Skip to main content

Comparison between non-vitamin K oral antagonist versus warfarin in atrial fibrillation with and without valvular heart disease: a systematic review and meta-analysis

Abstract

Background

Atrial fibrillation (AF) poses a significant stroke risk in heart disease patients. This systematic review aims to evaluate the efficacy and safety of non-vitamin K oral antagonists (NOACs) versus vitamin K antagonists (VKAs) in AF patients with and without any valvular heart disease (VHD/N-VHD).

Methods

A systematic search was conducted on PubMed, Scopus, and Google Scholar up to March 3, 2022. Efficacy and safety parameters were analyzed.

Results

A total of 85,423 subjects from 10 studies were included in this meta-analysis. NOACs and VKAs showed similar effects on ischemic stroke in AF patients with VHD/N-VHD (RR 0.97; 95% CI 0.72–1.30; p = 0.83) and also on systemic embolic events (RR 1.02; 95% CI 0.83–1.25; p = 0.86). Similar effects were seen in VHD and N-VHD subgroups. Both treatments had similar effects on myocardial infarction in AF patients with VHD/N-VHD (RR 0.79; 95% CI 0.49–1.26; p = 0.32), VHD (RR 0.78; 95% CI 0.59–1.02; p = 0.07), and N-VHD subgroups (RR 0.82; 95% CI 0.30–2.21; p = 0.69). NOACs reduced the risk of intracranial bleeding in AF VHD/N-VHD (RR 0.64; 95% CI 0.54–0.77; p < 0.0001), VHD (RR 0.59; 95% CI 0.42–0.82; p = 0.002), and N-VHD subgroups (RR 0.70; 95% CI 0.57–0.85; p = 0.0003). Additionally, NOACs reduced the risk of gastrointestinal bleeding in AF VHD/N-VHD (RR 0.80; 95% CI 0.66–0.96; p = 0.02), specifically in the VHD subgroup (RR 0.69; 95% CI 0.54–0.89; p = 0.004). Moreover, NOACs were associated with a decreased risk for minor and non-fatal bleeding in AF patients with VHD/N-VHD (RR 0.86; 95% CI 0.75–0.99; p = 0.04).

Conclusion

NOACs are effective and safe for ischemic stroke, systemic embolic events, myocardial infarction, intracranial bleeding, and gastrointestinal bleeding in AF patients with VHD/N-VHD.

Background

Atrial fibrillation (AF) is a common kind of heart arrhythmia. AF develops when tachyarrhythmia, an irregular electrical action in the heart’s atrium, starts fibrillation. A variety of symptoms, including but not limited to chest pain, palpitations, fast heartbeat, difficulty breathing, nausea, vertigo, profuse perspiration, and weakness, can accompany atrial fibrillation (AF) [1,2,3]. In establishing the diagnosis of AF, the electrocardiogram (ECG) examinations are crucial in confirming the diagnosis of atrial fibrillation (AF). The electrocardiogram (ECG) reveals a complex narrow pattern that is “irregularly irregular” and does not contain any discernible p waves. With ventricular rates ranging from 80 to 180 beats per minute, fibrillar waves might or might not be visible [4]. Age, hypertension, preexisting cardiac conditions, congenital heart defects, and alcohol consumption are all risk factors for atrial fibrillation [1,2,3].

Management of cardioversion in AF can be achieved through pharmacological or electrical means. Intravenous amiodarone is the only available antiarrhythmic medication in Egypt for acute AF cardioversion, and it can take between 6 and 16 h to be effective [5]. Identifying clinical factors that predict early successful cardioversion can help in deciding whether to use pharmacological or electrical cardioversion, potentially reducing hospital stay and associated costs. For instance, the use of antiarrhythmic agents such as encainide, digoxin, and amiodarone has been associated with an increased propensity for rhythm conversion in patients undergoing cardioversion [6, 7].

Atrial fibrillation (AF) in valvular heart disease (VHD) remains a concern. According to the 2023 ACC/AHA/ACCP/HRS guidelines, VHD encompasses any dysfunction or abnormality in one or more of the heart’s four valves: the aortic, mitral, pulmonary, and tricuspid valves. These issues can manifest as stenosis (a narrowing that restricts blood flow) or regurgitation (backward leakage due to improper valve closure). If not properly managed, VHD can result in significant morbidity and mortality [8]. Based on the 2021 ESC/EACTS guidelines, the management of valvular heart disease (VHD) varies significantly depending on the type and severity of the valve dysfunction. For mild cases, conservative management with regular monitoring is typically recommended; lifestyle modifications and medical management are needed; contributing conditions like hypertension or heart failure may be recommended. In moderate cases, medical therapy is often employed to manage symptoms and prevent complications, with interventions considered if symptoms worsen. For severe cases, more aggressive interventions such as valve repair or replacement are necessary; options include valve repair or replacement, which can be done via traditional open-heart surgery or minimally invasive techniques like transcatheter aortic valve replacement (TAVR) or mitral valve repair. The choice between surgical and transcatheter techniques depends on various factors, including patient-specific characteristics, procedural risks, and the expertise of the heart team. The guidelines emphasize individualized treatment plans and the importance of a multidisciplinary approach to optimize patient outcomes [9].

In cases of cardiac disease, AF is the primary determinant of stroke risk [10]. Because of the irregular heartbeat, the patient’s blood flow becomes turbulent, increasing the risk of thrombus formation and, in the worst-case scenario, a stroke [1,2,3]. The occurrence of AF has been on the rise worldwide. The prevalence of AF seems to increase with age. By the year 2050, the number of individuals diagnosed with atrial fibrillation will probably have increased by a factor of two or three. Atrial fibrillation affects more than 9 percent of individuals 75 and older, despite a global prevalence of less than 1%. Atrial fibrillation (AF) is 22% more common in people aged 80 and up [11, 12]. The incidence of atrial fibrillation (AF) is higher in developed nations compared to less developed ones, and it affects males more often than women [13].

Anticoagulants, rate-controlling medications, rhythm, cardioversion, ablation, and other cardiac operations can lower the risk of stroke in persons with atrial fibrillation (AF) [1,2,3]. Vitamin K antagonists (VKAs) and non-vitamin K oral anticoagulants (NOACs) are necessary for the anticoagulation of atrial fibrillation (AF), which is intended to prevent stroke. The use of VKAs persisted in falling after the four NOACs—dabigatran, rivaroxaban, apixaban, and edoxaban—were agreed upon [14]. When comparing warfarin with NOAC, the only group that warrants an exception are patients who have mechanical heart valves and moderate to severe mitral stenosis (MS) [15, 16]. When compared to VKA, NOAC is just as effective and safer in reducing the risk of stroke or bleeding, if not safer [17, 18].

A decreased incidence of blood loss events equivalent to VKA was associated with the combined therapy strategy involving NOAC. Triple antithrombotic therapy (TAT) considerably decreased risk when added to dual antithrombotic therapy (DAT). Compared to TAT with VKA and dual antiplatelet therapy (DAPT), DAT with NOAC and single antiplatelet therapy (SAPT) reduced the relative risk (RR 0.63; 95% CI 0.50–0.80) by 37%. Neither VKA nor treatment approaches involving a combination of NOAC were associated with a lower risk of stroke or death in prior research [19]. Thus, in order to prevent AF patients from developing valvular heart disease (VHD), a comprehensive evaluation of therapeutic approaches is required, including the use of NOAC or VKA for stroke prevention.

Methods

This systematic review was conducted on the PRISMA protocol [20]. The registration number for this review procedure is CRD42022357998, and it is part of the International Prospective Register of Systematic Reviews (PROSPERO).

Eligibility criteria

For the sake of this meta-analysis, we strictly adhered to inclusion and exclusion criteria. Valvular AF is defined as AF occurring in patients with mitral stenosis (MS), mitral regurgitation (MR), aortic stenosis (AS), aortic regurgitation (AR), mechanical heart valves, or those who have undergone valve replacement. These conditions are considered inclusion criteria for valvular AF. In contrast, non-valvular AF is defined as AF not caused by a heart valve issue. Studies included in this meta-analysis specifically addressed either valvular or non-valvular AF. We excluded studies that did not focus on individuals with AF related to valvular or non-valvular heart disease (N-VHD). The following criteria were also considered: (1) efficacy in the form of an ischemic stroke; (2) safety in the form of myocardial infarction, intracranial bleeding, or gastrointestinal bleeding; (3) the use of a cross-sectional, cohort, or case–control study design; and (4) the research had to have been written in English. On the flip side, we did not include trials that did not compare NOACs to warfarin or that did not have appropriate outcome measures. Additionally, in order to guarantee that the results were applicable to the intended group of AF patients, data from non-human studies were also omitted from the study.

Search strategy and selection of studies

A number of databases were searched in order to find pertinent subjects up until March 2022 including PubMed, ScienceDirect, and Google Scholar. Formula search terms included “anticoagulant,” “NOAC,” “novel oral anticoagulant,” “VKA,” “vitamin K antagonist,” “atrial fibrillation,” “stroke,” “intracranial hemorrhage,” “vascular heart disease,” and “non-vascular heart disease.” Boolean “AND” and “OR” were also utilized. By perusing the publications’ citations, we were able to uncover additional studies that were comparable or pertinent.

Data extraction

Designated investigators (A.S.A. and B.G.L.) painstakingly extracted relevant data using a predetermined data extraction form after relevant studies were selected. Study features including author, publication year, and study design were among the many aspects covered by the extracted data. Additionally, participant demographics, interventions administered (including type and dosage of NOACs or warfarin), and reported outcome measures were systematically recorded. To maintain the integrity and precision of the data extraction process, a thorough cross-checking procedure was implemented. Another investigator independently reviewed the extracted data to verify its accuracy and completeness, thereby mitigating the risk of errors or omissions. This stringent validation process ensured the reliability and robustness of the extracted data for subsequent analysis.

Quality assessment

The Newcastle–Ottawa Scale (NOS), developed for use in non-randomized research designs, was used to conduct additional evaluations of each of the publications that were included in the list. There are three points for exposure determination, four for patient selection, and two for group comparability in the NOS. Total scores could range from 0 (very bad) to 9 (excellent). The Cochrane risk-of-bias instrument for randomized trials, second edition (RoB 2), was utilized to assess RCT publications for potential bias in a more comprehensive manner. The RoB2 tools automatically do all computations and evaluations in accordance with five critical areas [21, 22]. The quality evaluation was carried out by two researchers (A.S.A.). If any conflicts arise during this evaluation period, the researchers will work together to find a solution.

Outcome measure

The analysis considered several outcome measures, encompassing efficacy and safety. Efficacy was evaluated in terms of ischemic stroke incidence, while safety endpoints included myocardial infarction, intracranial bleeding, and gastrointestinal bleeding.

Data synthesis and statistical analysis

Every outcome measure in this study had its own pooled risk ratio (RR) and 95% CI, which were determined by a meta-analysis. To determine how heterogeneous the included studies were, we used the I2 statistic. Patients undergoing atrial fibrillation can be categorized into subgroups according to the presence or absence of ventricular HF and non-ventricular HF. The results will also be subjected to sensitivity analysis to ensure their robustness. Statistical significance was defined as a p value less than 0.05. With the help of Review Manager 5.4, we ran the statistical analysis [23].

Results

Study selection process and quality assessment

After searching online sources such as PubMed, ScienceDirect, and Google Scholar, the authors discovered four studies out of 4730. After being rejected for reasons that are yet unclear, 49 studies were kept. Six studies were deemed eligible for assessment following paper content and paper-accessible screening. The total report of 6 new included studies, and the previous is 10. Here is a flowchart that summarizes the complete literature search process according to the PRISMA Guideline: (Fig. 1).

Fig. 1
figure 1

PRISMA flow diagram of the study selection process

Study characteristics

Table 1 shows the outcomes of interest associated with AF with or without VHD, as determined by a total of ten investigations [24,25,26,27,28,29,30,31,32,33], involving 85,423 individuals. With four studies each, Asia, the United States, and Europe accounted for the bulk of the participants. Three months to three and a half years was the range of the follow-up duration.

Table 1 Data characteristics

Risk of bias

Table 2 shows the categorization of the ten included studies according to the methodologies used to determine their risk of bias. Using the Newcastle–Ottawa Scale (NOS), six studies were evaluated; five of them were deemed to have good quality, while one was deemed to have fair/moderate quality. All of the studies that were considered by RoB to have a low risk of bias, with the exception of four others, indicate that the studies that were included are of good quality.

Table 2 Study outcome

Efficacy of non-vitamin k oral antagonists versus warfarin

Table 3 offers a concise evaluation of the safety and effectiveness of NOACs compared to warfarin in treating atrial fibrillation (AF) in individuals with and without valvular heart disease (VHD).

Table 3 Summary of results

Ischemic stroke

In Fig. 2, a total of 10 studies represents the total number of participants in each treatment group across all the included studies. There were 53,750 participants in the studies treated with NOACs and 45,185 participants treated with VKAs. We found no statistical difference between NOACs and VKAs in relation to ischemic stroke in VHD “(RR 0.88; 95% CI 0.75–1.04; p = 0.12; I2 = 90%)” and N-VHD “(RR 1.13; 95% CI 0.64–1.99; p = 0.68; I2 = 100%)” when we pooled our data. When it came to ischemic stroke, NOACs and VKAs had comparable effects in AF patients with VHD/N-VHD “(RR 0.97; 95% CI 0.72–1.30; p = 0.83; I2 = 99%).”

Fig. 2
figure 2

Forest plot of NOACs vs VKAs in ischemic stroke

Systemic embolic events (SEE)

In Fig. 3, the comparison of systemic embolic events between non-vitamin K oral anticoagulants (NOACs) and warfarin in atrial fibrillation (AF) patients with and without valvular heart disease (VHD) shows varied results. For patients with VHD, the relative risk (RR) of systemic embolic events was 1.09 (95% CI 0.78–1.53), indicating no significant difference between NOACs and warfarin. For patients without VHD (N-VHD), the RR was 0.94 (95% CI 0.83–1.07), also suggesting no significant difference between the two treatments. Overall, the pooled analysis found no statistical difference between NOACs and warfarin regarding systemic embolic events in AF patients regardless of the presence of VHD, with an RR of 0.86.

Fig. 3
figure 3

Forest plot of NOACs vs VKAs in systemic embolic events

Safety of non-vitamin k oral antagonists versus warfarin

Myocardial infraction

Figure 4 shows the results of the ten studies that looked at the risk ratio of AF patients with and without VHD in MI and how NOACs compared to VKAs. In the VHD “(RR 0.78; 95% CI 0.59–1.02; p = 0.07; I2 = 94%)” and N-VHD “(RR 0.82; 95% CI 0.30–2.21; p = 0.69; I2 = 99%)” groups, our combined analysis did not find a statistically significant difference in the risk of MI between NOACs and VKAs. Comparing NOAC and VKA effects on MI in AF patients with VHD/N-VHD, we find that they are similar “(RR 0.79; 95% CI 0.49–1.26; p = 0.32; I2 = 99%).”

Fig. 4
figure 4

Forest plot of NOACs vs VKAs in myocardial infraction

Intracranial bleeding

Figure 5 shows the results of 10 trials that demonstrated a decreased incidence of intracranial bleeding in patients with atrial fibrillation, regardless of whether they had valvular heart disease (VHD) or not, and in those who used NOACs instead of VKAs. We found that compared to VKAs, NOACs significantly reduce the incidence of intracranial hemorrhage in valvular heart disease patients by 41% in VHD “(RR 0.59; 95% CI 0.42–0.82; p = 0.002; I2 = 88%)” and by 30% in non-valvular heart disease patients “(RR 0.70; 95% CI 0.57–0.85; p = 0.0003; I2 = 66%).” The risk of cerebral hemorrhage was generally decreased by NOACs “(RR 0.64; 95% CI 0.54–0.77; p < 0.0001; I2 = 82%).”

Fig. 5
figure 5

Forest plot of NOACs vs VKAs in intracranial bleeding

Gastrointestinal bleeding

In terms of the risk ratio of gastrointestinal bleeding in patients with AF and those without VHD, Fig. 6 displays the results of ten trials that compared NOACs and VKAs. When we combined the data from the studies, we found that NOACs were far more likely to cause gastrointestinal bleeding than VKAs. A 31% reduction in the incidence of cerebral hemorrhage was observed in patients with VHD when NOACs were used instead of VKAs “(RR 0.69; 95% CI 0.54–0.89; p = 0.004; I2 = 95%).” However, in patients without VHD, the opposite was true “(RR 0.96; 95% CI 0.69–1.34; p = 0.83; I2 = 96%).” Patients with atrial fibrillation who did not have ventricular fibrillation and who used NOACs had a reduced risk of gastrointestinal bleeding “(RR 0.80; 95% CI 0.66–0.96; p = 0.02; I2 = 96%).”

Fig. 6
figure 6

Forest plot of NOACs vs VKAs in gastrointestinal bleeding

Minor and non-fatal bleeding

Figure 7 shows the results of the three studies that looked at the risk ratio of AF patients with and without VHD in minor and non-fatal bleeding and how NOACs compared to VKAs. In the VHD “(RR 0.93; 95% CI 0.81–1.07; p = 0.32; I2 = 31%)” and N-VHD “(RR 0.82; 95% CI 0.66–1.02; p = 0.07; I2 = 93%)” groups, our analyses did not find a statistically significant difference in the risk of minor and non-fatal bleeding between NOACs and VKAs. However, comparing NOAC and VKA effects in AF patients with VHD/N-VHD, we found that NOACs were associated with a decreased risk for minor and non-fatal bleeding “(RR 0.86; 95% CI 0.75–0.99; p = 0.04; I2 = 85%).”

Fig. 7
figure 7

Forest plot of NOACs vs VKAs in minor and non-fatal bleeding

The end points that were evaluated showed generally symmetrical funnel plots with estimable odds ratios (Fig. 8), suggesting that there was no major publication bias.

Fig. 8
figure 8

Funnel plot of publication bias for A Ischemic Stroke, B Systemic Embolic Events, C Myocardial Infraction, D Intracranial Bleeding, E Gastrointestinal Bleeding and F Minor and Non-Fatal Bleeding

Discussions

This meta-analysis assessed the safety and effectiveness of oral anticoagulants with and without vitamin K antagonists, drawing on data from 10 trials with 85,423 participants. The trials included warfarin, a drug that blocks the effects of vitamin K. Our research shows that NOACs greatly decrease the likelihood of bleeding in the brain and gastrointestinal system. This study shows that there is no statistical difference between NOACs and VKAs in relation to ischemic stroke, systemic embolic events (SEE), risk of MI, and minor and non-fatal bleeding. However, comparing NOAC and VKA effects in AF patients with VHD/N-VHD, we found that NOACs were associated with a decreased risk for minor and non-fatal bleeding. This result is in line with the previous study which reported that there is no significant difference between the NOACs compared to warfarin in terms of all-cause mortality and MI, but the risk of major bleeding events was found lower in patients receiving NOACs [34]. The results of the comparison between VKAs and NOACs may have different meanings and may differ significantly when analyzed separately for AF VHD and N-VHD. Combining these results may obscure potential differences as this study reported no significant difference was observed between DOACs and VKAs in AF regardless of AF etiology.

In this study, VHD classifications included aortic stenosis (AS), aortic regurgitation (AR), tricuspid regurgitation (TR), MS, and mitral regurgitation (MR). Among the included studies, the population receiving NOACs and warfarin for ischemic stroke and SEE was more frequent in AF patients with MR (22,250 patients). This study found no statistical difference between NOACs and VKAs in preventing ischemic stroke and SEE. According to Fanaroff et al., in patients with AF and MR, NOACs have been shown to be as effective as VKAs in reducing the risk of stroke and systemic embolism. Pivotal trials demonstrated that the effect of NOACs compared to VKAs on the primary efficacy endpoint of stroke and systemic embolism was similar for patients with and without valvular heart disease, including those with MR. Specifically, the rate of stroke and systemic embolism per 100 patient-years in patients with valvular heart disease treated with VKAs was 1.56, while it was 0.83 for those treated with NOACs in the ROCKET-AF trial [35, 36]. Connolly et al. (2022) enrolled 4531 patients with rheumatic heart disease-associated atrial fibrillation to compare the efficacy of non-vitamin K antagonist oral anticoagulants (NOACs), specifically rivaroxaban, with vitamin K antagonists (VKAs). The study found that VKA therapy led to a lower rate of a composite of cardiovascular events or death compared to rivaroxaban, without a higher rate of major bleeding. Additionally, VKA therapy resulted in a lower rate of ischemic stroke and lower mortality due to vascular causes. These findings support current guidelines recommending VKA therapy for stroke prevention in patients with rheumatic heart disease and atrial fibrillation [37]. Consequently, NOACs are recommended over VKAs in patients with AF and MR. In terms of efficacy and safety, Melgaard et al. found that NOACs were associated with a significantly higher risk of thromboembolism compared to warfarin in AF patients with AS, but NOACs were associated with a significantly lower risk of major bleeding compared to warfarin in the same patient group. Furthermore, according to the guidelines from the American College of Cardiology (ACC) and the American Heart Association (AHA), NOACs are not recommended for AF patients with moderate to severe MS due to the absence of supporting evidence in the literature [38].

A number of studies have looked at the effectiveness and safety of various anticoagulants in atrial fibrillation patients in relation to bleeding [34]. In order to lessen the likelihood of bleeding and systemic embolism, this research illuminate the best ways to manage this group of patients. Dabigatran reduced the risk of bleeding in patients with non-valvular AF (NVAF) in the RE-LY trial when compared to other NOACs, including rivaroxaban and apixaban. Patients with NVAF who are more likely to experience bleeding should be evaluated for the possibility of using dabigatran as a therapeutic option. However, each patient’s specific circumstances and considerations should be considered before selecting an anticoagulant [39,40,41].

Continuing from the previous point, it is crucial to think about the effects of ischemic stroke and MI when managing AF [42]. Ischemic stroke is a major concern for patients with atrial fibrillation due to the higher frequency of thromboembolic events [43]. There is evidence that NOACs are superior to warfarin in preventing ischemic stroke and systemic embolism in patients with atrial fibrillation and native VHD. Apixaban, Dabigatran, and Edoxaban are NOACs that have been shown to decrease hemorrhagic events; however, Rivaroxaban has been associated with an alarming increase in significant bleeding episodes. It is crucial to consider the patient’s risk profile and bleeding tendency before choosing an anticoagulant [44].

Preventing myocardial infarction (MI) is another important therapeutic concern in AF treatment [45]. Treatment decisions should take into consideration the risk of myocardial infarction (MI) and its effects, even if the primary objective of anticoagulant medication is to prevent thromboembolic events. It is important to closely monitor and alter dosages of warfarin as previous research has shown that its effects might vary and that it can interact with other drugs and foods. In contrast, NOACs have the potential to improve patient adherence and treatment results through their streamlined dose regimens and reduced requirement for anticoagulation monitoring [46].

Treatment choices are further complicated when patients with AF undergoing percutaneous coronary intervention (PCI) must also decide on antithrombotic medication [47]. When compared to warfarin-based regimens, the bleeding risk and effectiveness of NOACs combined with single or double antiplatelet therapy are equal, suggesting that they are not inferior. When choosing an antithrombotic medication for atrial fibrillation patients having a percutaneous coronary intervention (PCI) at the same time, it is important to weigh the risks of stent thrombosis, stroke, and bleeding complications [48, 49].

Compliance with NOAC doses and the efficacy of AF medication may interact with one another. Dose once daily has been linked to better patient adherence without sacrificing effectiveness or safety, whereas twice-day dose regimens may provide a more stable risk–benefit profile for stroke prevention. In addition, the patient’s preferences, lifestyle characteristics, renal function, and bleeding risk should be considered when deciding between NOACs and VKAs [50, 51].

Overall, AF therapy necessitates a holistic strategy that harmonizes thromboembolic event avoidance with bleeding and other adverse outcome risk assessment [52]. Anticoagulant therapy, particularly NOACs, has revolutionized the treatment of AF by offering improved safety, efficacy, and convenience compared to traditional VKA therapy. However, the selection of the most appropriate anticoagulant should be guided by careful consideration of individual patient characteristics and preferences, as well as the presence of concomitant conditions such as ischemic stroke and myocardial infarction. By tailoring treatment strategies to the specific needs of each patient, healthcare providers can optimize outcomes and enhance the quality of care for individuals with AF [51].

Conclusions

The findings of this study indicate that both NOACs and VKAs exhibit comparable efficacy in preventing ischemic stroke in AF patients, regardless of the presence of VHD or N-VHD. Both treatment modalities also demonstrate similar effectiveness in reducing the incidence of myocardial infarction in these patient populations. Importantly, NOACs significantly reduce the risk of intracranial bleeding in both VHD and N-VHD patients. In addition, NOACs are associated with a decreased risk of gastrointestinal bleeding specifically in VHD patients, although this effect does not reach statistical significance in the N-VHD subgroup. Moreover, NOACs are linked to a lower risk of minor and non-fatal bleeding in AF patients with either VHD or N-VHD. These results suggest that NOACs offer a favorable safety profile compared to VKAs, particularly in terms of bleeding complications. However, it is crucial to interpret these findings in the context of current clinical guidelines and to consider potential variations in treatment recommendations for different patient populations.

Limitation

The included studies presented heterogeneity in the duration of the treatment given to patients. Moreover, in some studies, with a rather short follow-up period, long-term outcomes may not be captured. Furthermore, mitral stenosis (MS) was absent in 50% of this research, and only about 1.6% of the total patient number in the review had MS. This significant underrepresentation of patients with MS is not clearly mentioned or discussed as a limitation. In short, although NOACs appear to be a safer choice over VKAs, especially for bleeding risks, further research with a more homogeneous study population and longer follow-up is needed to confirm these findings and guide clinical guidelines.

Availability of data and materials

Data available within the article. The authors confirm that the data supporting the findings of this study are available within the article.

Abbreviations

AF:

Atrial fibrillation

CI:

Confidence interval

DAPT:

Dual antiplatelet therapy

DAT:

Dual antithrombotic therapy

ECG:

Electrocardiogram

MI:

Myocardial infarction

NOS:

Newcastle–Ottawa Scale

NOAC:

Non-vitamin K oral anticoagulant

NVAF:

Non-valvular atrial fibrillation

PCI:

Percutaneous coronary intervention

PRISMA:

Preferred Reporting Items for Systematic Review and Meta-Analysis

PROSPERO:

International Prospective Register of Systematic Reviews

RCT:

Randomized controlled trial

RR:

Risk ratio

SAPT:

Single antiplatelet therapy

SEE:

Systemic embolic event

TAT:

Triple antithrombotic therapy

VHD:

Valvular heart disease

VKA:

Vitamin K antagonist

References

  1. McManus DD, Rienstra M, Benjamin EJ (2012) An update on the prognosis of patients with atrial fibrillation. Circulation 126:e143–e146

    Article  PubMed  PubMed Central  Google Scholar 

  2. Amin A, Houmsse A, Ishola A, Tyler J, Houmsse M (2016) The current approach of atrial fibrillation management. Avicenna J Med 6:8–16

    Article  PubMed  PubMed Central  Google Scholar 

  3. Markides V, Schilling RJ (2003) Atrial fibrillation: classification, pathophysiology, mechanisms and drug treatment. Heart 89:939–943

    Article  PubMed  PubMed Central  Google Scholar 

  4. Nesheiwat Z, Goyal A, Jagtap M (2022) Atrial fibrillation. StatPearls Publishing, Treasure Island

    Google Scholar 

  5. Roshdy HS, Naguib TA, Elthalawi MA (2016) A new score for detection of early cardioversion using intravenous amiodarone in recent onset atrial fibrillation. Egpt Heart J 68:245–252

    Article  Google Scholar 

  6. Kapadia M, Jagadish PS, Hutchinson M, Lee H (2023) Atrial fibrillation, electroconvulsive therapy, stroke risk, and anticoagulation. Springer Science and Business Media, Deutschland GmbH

    Book  Google Scholar 

  7. Arrigo M, Jeger N, Seifert B, Spahn DR, Bettex D, Rudiger A (2015) Disappointing success of electrical cardioversion for new-onset atrial fibrillation in cardiosurgical ICU patients. Intensive Care Medicine Experimental, SpringerOpen

  8. Joglar JA, Chung MK, Armbruster AL, Benjamin EJ, Chyou JY, Cronin EM, et al (2023) ACC/AHA/ACCP/HRS guideline for the diagnosis and management of atrial fibrillation: a report of the American college of cardiology/American Heart association joint committee on clinical practice guidelines. Circulation. Lippincott Williams and Wilkins 2024. pp E1–156

  9. Vahanian A, Beyersdorf F, Praz F, Milojevic M, Baldus S, Bauersachs J et al (2022) 2021 ESC/EACTS guidelines for the management of valvular heart disease. Eur Heart J 43:561–632

    Article  PubMed  Google Scholar 

  10. Lippi G, Sanchis-Gomar F, Cervellin G (2021) Global epidemiology of atrial fibrillation: an increasing epidemic and public health challenge. Int J Stroke 16:217–221

    Article  PubMed  Google Scholar 

  11. Peters SAE, Woodward M (2019) Established and novel risk factors for atrial fibrillation in women compared with men. Heart 105:226–234

    Article  CAS  PubMed  Google Scholar 

  12. Choi Y-J, Choi E-K, Han K-D, Jung J-H, Park J, Lee E et al (2018) Temporal trends of the prevalence and incidence of atrial fibrillation and stroke among Asian patients with hypertrophic cardiomyopathy: a nationwide population-based study. Int J Cardiol 273:130–135

    Article  PubMed  Google Scholar 

  13. Chugh SS, Havmoeller R, Narayanan K, Singh D, Rienstra M, Benjamin EJ et al (2014) Worldwide epidemiology of atrial fibrillation: a global burden of disease 2010 study. Circulation 129:837–847

    Article  PubMed  Google Scholar 

  14. Schäfer A, Flierl U, Berliner D, Bauersachs J (2020) Anticoagulants for stroke prevention in atrial fibrillation in elderly patients. Cardiovasc Drugs Ther 34:555–568

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bai CJ, Madan N, Alshahrani S, Aggarwal NT, Volgman AS (2018) Sex differences in atrial fibrillation-update on risk assessment, treatment, and long-term risk. Curr Treat Options Cardiovasc Med 20:79

    Article  PubMed  Google Scholar 

  16. Laäs DJ, Naidoo M (2018) Oral anticoagulants and atrial fibrillation: a South African perspective. S Afr Med J 108:640–646

    Article  PubMed  Google Scholar 

  17. Tellor KB, Patel S, Armbruster AL, Daly MW (2015) Evaluation of the appropriateness of dosing, indication and safety of rivaroxaban in a community hospital. J Clin Pharm Ther 40:447–451

    Article  CAS  PubMed  Google Scholar 

  18. Armbruster AL, Buehler KS, Min SH, Riley M, Daly MW (2014) Evaluation of dabigatran for appropriateness of use and bleeding events in a community hospital setting. Am Health Drug Benefits 7:376–384

    PubMed  PubMed Central  Google Scholar 

  19. Eyileten C, Postula M, Jakubik D, Toma A, Mirowska-Guzel D, Patti G, Renda G, Siller-Matula JM (2020) Non-vitamin K oral anticoagulants (Noac) versus vitamin K antagonists (vka) for atrial fibrillation with elective or urgent percutaneous coronary intervention: a meta-analysis with a particular focus on combination type. J Clin Med 9(4):1120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, The PRISMA et al (2020) statement: an updated guideline for reporting systematic reviews. Int J Surg 2021:88

    Google Scholar 

  21. Wells G, Shea B, O’Connell D, Peterson J, Welch V, Losos M (2014) Newcastle–Ottawa quality assessment scale cohort studies. University of Ottawa, Ottawa

    Google Scholar 

  22. Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al (2019) RoB 2: a revised tool for assessing risk of bias in randomised trials. The BMJ 366

  23. The cochrane collaboration. Review manager (RevMan) version 5.4 [Internet]. 2020 [cited 2024 Apr 13]. Available from: https://training.cochrane.org/System/Files/Uploads/Protected_file/RevMan5.4_user_guide.Pdf

  24. Avezum A, Lopes RD, Schulte PJ, Lanas F, Gersh BJ, Hanna M et al (2015) Apixaban in comparison with warfarin in patients with atrial fibrillation and valvular heart disease: Findings From the Apixaban for reduction in stroke and other thromboembolic events in atrial fibrillation (Aristotle) trial. Circulation 132:624–632

    Article  CAS  PubMed  Google Scholar 

  25. Chaudhry UA, Ezekowitz MD, Gracely EJ, George WT, Wolfe CM, Harper G et al (2021) Comparison of low-dose direct acting anticoagulant and Warfarin in patients aged ≥80 years with atrial fibrillation. Am J Cardiol 152:69–77

    Article  CAS  PubMed  Google Scholar 

  26. Reinecke H, Nabauer M, Gerth A, Limbourg T, Treszl A, Engelbertz C et al (2015) Morbidity and treatment in patients with atrial fibrillation and chronic kidney disease. Kidney Int 87:200–209

    Article  CAS  PubMed  Google Scholar 

  27. Patti G, Cavallari I, Hanon O, De Caterina R (2018) The safety and efficacy of non-vitamin K antagonist oral anticoagulants in atrial fibrillation in the elderly. Int J Cardiol 265:118–124

    Article  PubMed  Google Scholar 

  28. Kanai Y, Oguro H, Tahara N, Matsuda H, Takayoshi H, Mitaki S et al (2018) Analysis of recurrent stroke volume and prognosis between warfarin and four non-vitamin K antagonist oral anticoagulants’ administration for secondary prevention of stroke. J Stroke Cerebrovasc Dis 27:338–345

    Article  PubMed  Google Scholar 

  29. Kim M, You SC, Sung J-H, Jang E, Yu HT, Kim T-H et al (2021) Safety and long-term outcomes of catheter ablation according to sex in patients with atrial fibrillation: a nationwide cohort study. Int J Cardiol 338:95–101

    Article  PubMed  Google Scholar 

  30. Okumura K, Lip GYH, Akao M, Tanizawa K, Fukuzawa M, Abe K et al (2017) Edoxaban for the management of elderly Japanese patients with atrial fibrillation ineligible for standard oral anticoagulant therapies: rationale and design of the ELDERCARE-AF study. Am Heart J 194:99–106

    Article  CAS  PubMed  Google Scholar 

  31. Melgaard L, Overvad TF, Jensen M, Christensen TD, Lip GY, Larsen TB, Nielsen PB (2021) Effectiveness and safety of noac versus warfarin in patients with atrial fibrillation and aortic stenosis. J Am Heart Assoc 10(23):e022628

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Strange JE, Sindet-Pedersen C, Staerk L, Grove EL, Gerds TA, Torp-Pedersen C, Gislason GH, Olesen JB (2021) All-cause mortality, stroke, and bleeding in patients with atrial fibrillation and valvular heart disease. Eur Heart J-Cardiovasc Pharmacother 7(FI1):f93-100. https://doi.org/10.1093/ehjcvp/pvaa011/5739442

    Article  PubMed  Google Scholar 

  33. Li W, Wang S, Li S, Chen X, Xiong Y (2019) Clinical outcomes of valvular versus nonvalvular atrial fibrillation in acute anterior circulation occlusive stroke undergoing endovascular treatment. J Stroke Cerebrovasc Dis 28:31–37

    Article  PubMed  Google Scholar 

  34. Clemens A, Noack H, Brueckmann M, Lip GYH (2014) Twice- or once-daily dosing of novel oral anticoagulants for stroke prevention: a fixed-effects meta-analysis with predefined heterogeneity quality criteria. PLoS ONE 9:1–8

    Article  Google Scholar 

  35. Fanaroff AC, Vora AN, Lopes RD (2022) Non-vitamin K antagonist oral anticoagulants in patients with valvular heart disease. Eur Heart J Suppl 24:A19-31

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hohnloser SH, Lopes RD (2014) Atrial fibrillation, valvular heart disease, and use of target-specific oral anticoagulants for stroke prevention. Eur Heart J 35(47):3323–3325

    Article  CAS  PubMed  Google Scholar 

  37. Connolly SJ, Karthikeyan G, Ntsekhe M, Haileamlak A, El Sayed A, El Ghamrawy A et al (2022) Rivaroxaban in rheumatic heart disease-associated atrial fibrillation. N Engl J Med 387:978–988

    Article  CAS  PubMed  Google Scholar 

  38. Al Rawahi MN, Al-Maqbali JS, Al Noumani J, Al Alawi AM, Essebag V (2023) Novel oral anticoagulants in patients with atrial fibrillation and moderate to severe mitral stenosis: a systematic review. Cureus

  39. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A et al (2009) Dabigatran versus Warfarin in patients with atrial fibrillation. N Engl J Med 361:1139–1151

    Article  CAS  PubMed  Google Scholar 

  40. Schaefer JK, McBane RD, Wysokinski WE (2016) How to choose appropriate direct oral anticoagulant for patient with nonvalvular atrial fibrillation. Ann Hematol 95:437–449

    Article  CAS  PubMed  Google Scholar 

  41. Adeboyeje G, Sylwestrzak M, Barron JJ, White J, Rosenberg A, Abarca J, et al (2017) Major bleeding risk during anticoagulation with warfarin, dabigatran, apixaban, or rivaroxaban in patients with nonvalvular atrial fibrillation. J Manag Care Spec Pharm. 2017. Available from: www.jmcp.org

  42. Emren SV, Zoghi M, Berilgen R, Özdemir İH, Çelik O, Çetin N et al (2018) Safety of once-or twice-daily dosing of non-vitamin K antagonist oral anticoagulants (NOACs) in patients with nonvalvular atrial fibrillation: a NOAC-TR study. Bosn J Basic Med Sci 18:185–190

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bang OY, Park KM, Jeong DS (2023) Occurrence of ischemic stroke in patients with atrial fibrillation receiving non-vitamin K oral anticoagulants: causes and prevention strategies. J Stroke 25(2):199–213

    Article  PubMed  PubMed Central  Google Scholar 

  44. Pan KL, Singer DE, Ovbiagele B, Wu YL, Ahmed MA, Lee M (2017) Effects of non–vitamin K antagonist oral anticoagulants versus warfarin in patients with atrial fibrillation and valvular heart disease: a systematic review and meta-analysis. J Am Heart Assoc 6(7):e005835

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lip GYH, Lane DA, Lenarczyk R, Boriani G, Doehner W, Benjamin LA et al (2022) Integrated care for optimizing the management of stroke and associated heart disease: a position paper of the European society of cardiology council on stroke. Eur Heart J 43:2442–2460

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Caldeira D, David C, Costa J, Ferreira JJ, Pinto FJ (2018) Non-Vitamin K antagonist oral anticoagulants in patients with atrial fibrillation and valvular heart disease: systematic review and meta-analysis. Eur Heart J Cardiovasc Pharmacother 4:111–118

    Article  PubMed  Google Scholar 

  47. Pan KL, Singer DE, Ovbiagele B, Wu YL, Ahmed MA, Lee M (2017) Effects of non-vitamin K antagonist oral anticoagulants versus warfarin in patients with atrial fibrillation and valvular heart disease: a systematic review and meta-analysis. J Am Heart Assoc 6:1–12

    Article  Google Scholar 

  48. Botto GL, Ameri P, De Caterina R (2021) Many good reasons to switch from vitamin k antagonists to non-vitamin k antagonists in patients with non-valvular atrial fibrillation. J Clin Med 10(13):2866

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bellin A, Berto P, Themistoclakis S, Chandak A, Giusti P, Cavalli G, Bakshi S, Tessarin M, Deambrosis P, Chinellato A (2019) New oral anti-coagulants versus vitamin K antagonists in high thromboembolic risk patients. PLoS ONE 14(10):e0222762

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yamamoto K, Koretsune Y, Akasaka T, Kisanuki A, Ohte N, Takenaka T, Takeuchi M, Yoshida K, Iwade K, Okuyama Y, Hirano Y (2017) Effects of vitamin K antagonist on aortic valve degeneration in non-valvular atrial fibrillation patients: prospective 4-year observational study. Thrombosis Res 1(160):69–75

    Article  Google Scholar 

  51. A. Foster and Dasgupta. Oral Anticoagulants Guideline for prescribing, monitoring and management. 2018.

  52. Botto GL, Ameri P, De Caterina R (2021) Many good reasons to switch from vitamin K antagonists to non-vitamin k antagonists in patients with non-valvular atrial fibrillation. J Clin Med 10:1–14

    Article  Google Scholar 

Download references

Acknowledgements

None.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design were done by ASA. Analysis and interpretation of the data were done by ASA and BGL. Drafting of the article was done by ASA and BGL. Critical revision of the article for important intellectual content was done by AS. Final approval of the article was done by ASA and BGL. Provision of study materials or patients was done by ASA.

Corresponding author

Correspondence to Arga Setyo Adji.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All of the authors have reviewed the final version of the manuscript and agreed to publish this manuscript.

Competing interests

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Adji, A.S., de Liyis, B.G. Comparison between non-vitamin K oral antagonist versus warfarin in atrial fibrillation with and without valvular heart disease: a systematic review and meta-analysis. Egypt Heart J 76, 102 (2024). https://doi.org/10.1186/s43044-024-00535-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43044-024-00535-w

Keywords