• In this nationwide cohort study, apixaban, dabigatran, and rivaroxaban had similar rates of any stroke or systemic embolism.

  • Rivaroxaban had higher rates of major bleeding than apixaban and dabigatran but lower rates of myocardial infarction than dabigatran.

In the pivotal randomized controlled trials (RCTs) for patients with atrial fibrillation, direct oral anticoagulants (DOACs) had similar or even superior efficacy and safety compared with warfarin. However, RCTs comparing different DOACs are nonexistent and previous observational studies have yielded conflicting results. In this nationwide cohort study, rates of any stroke or systemic embolism (stroke/SE) and major bleeding were compared among new users of apixaban, dabigatran, and rivaroxaban with atrial fibrillation from 2014 to 2019. Inverse probability weighting was used to yield balanced study groups, and outcomes were compared using Cox regression. Stroke/SE rates were similar in patients receiving apixaban, dabigatran, and rivaroxaban. Dabigatran was associated with twofold higher rates of myocardial infarction (MI) than rivaroxaban (1.4 events/100 person-years (py) vs 0.7 events/100-py, hazard ratio [HR] 2.21, 95% confidence interval [CI], 1.00-4.90) and apixaban (1.4 events/100-py vs 0.7 events/100-py, HR 2.26, 95% CI, 0.90-5.67), although the second comparison included the possibility of a null effect. Rivaroxaban was associated with higher major bleeding rates compared with apixaban (2.9 events/100-py vs 1.8 events/100-py, HR 1.64, 95% CI, 1.13-2.37) and dabigatran (2.9 events/100-py vs 1.4 events/100-py, HR 2.18, 95% CI, 1.21-3.93). Specifically, rivaroxaban had higher rates of major gastrointestinal bleeding and other major bleeding than apixaban. In conclusion, although stroke/SE rates were similar for DOACs, rivaroxaban was associated with higher rates of major bleeding than other DOACs and lower rates of MI than dabigatran. These results may help guide oral anticoagulant selection, especially in patients at high risk of bleeding or MI.

Pivotal randomized controlled clinical trials (RCTs) demonstrated that direct oral anticoagulants (DOACs) had similar or even superior efficacy compared with warfarin.1-4 In addition, these trials reported higher rates of major bleeding with warfarin than with apixaban, edoxaban, and low-dose dabigatran, whereas major bleeding rates were similar between warfarin and rivaroxaban and high-dose dabigatran.1,4 Whether thromboembolic and major bleeding events differ between individual DOACs remains unclear because no RCT has compared DOACs head-to-head. Assessments of thromboembolic event rates have yielded conflicting results in previous observational studies5-10 whereas rivaroxaban has generally been associated with higher major bleeding rates than other DOACs.5-10 

In the initial RCTs, major bleeding was defined according to the International Society of Thrombosis and Hemostasis (ISTH) as a bleeding event leading to hemoglobin drop of ≥20 g/L, transfusion of ≥2 units of red blood cells, bleeding into a critical area (such as the cranium or retroperitoneum), or death.1-4 To the best of our knowledge, no large observational study has used this definition for major bleeding, although a modified version of the ISTH definition was used in a nationwide Norwegian registry study, omitting hemoglobin drop as 1 of the criteria.11 Otherwise, most studies have defined major bleeding as a bleeding event requiring hospitalization, using relevant hospital discharge codes only and without manual verification of events. How closely this resembles the ISTH criteria for major bleeding is unknown. A previous study from our group demonstrated that only 77% of hospitalized patients with gastrointestinal bleeding (GIB) fulfilled the ISTH criteria of major bleeding.12 The proportion is likely lower for other bleeding events such as hematuria, epistaxis, postmenopausal bleeding, or unspecified anemia, which have been included as major bleeding in previous studies.7,8,10,13-15 Thus, population-based real-world studies with well-characterized patients with adequate follow-up are important to elucidate potential differences in the effectiveness and safety of DOACs.

The aim of this study was to compare the rates of any stroke or systemic embolism (stroke/SE) and major bleeding associated with apixaban, dabigatran, and rivaroxaban for patients with atrial fibrillation in a nationwide propensity score–weighted cohort.

Data source

The creation of the Icelandic oral anticoagulation database has been previously described with emphasis on GIB.16 The database includes data on all patients in Iceland receiving oral anticoagulants (OACs) from 1 March 2014 to 28 February 2019. Using the unique national identification number of each patient, assigned to all Icelanders at birth or upon immigration, data were collected from the Icelandic Medicine Registry, the 5 major hospitals in Iceland, and the Icelandic death registry. This study was approved by the National Bioethics Committee of Iceland (VSN-16-057-V4) and was conducted according to the Declaration of Helsinki.

Patient selection and follow-up

Patients included were those with atrial fibrillation who started treatment with apixaban, dabigatran, and rivaroxaban from 1 March 2014 to 28 February 2019. To establish an OAC naïve cohort, patients were excluded if they had filled a prescription for any OAC in the preceding 12 months before their eligibility into the study. In addition, patients were excluded if their permanent residence was outside Iceland, if they had end-stage renal disease, mitral stenosis, or a mechanical heart valve.

Patients were followed from the date of first drug dispensation to 28 February 2019, or earlier if either the treatment was stopped, primary outcome was achieved, death occurred, they moved out of the country, or were switched to a different OAC.

Exposure and outcomes

The exposure of interest was treatment with apixaban, dabigatran, or rivaroxaban. The primary effectiveness outcome was any stroke/SE. The primary safety outcome was major bleeding. Secondary outcomes were myocardial infarction (MI), venous thromboembolism, arterial thromboembolism, transient ischemic attack (TIA), intracranial hemorrhage, major GIB, other major bleeding, all-cause mortality, vascular mortality, ischemic stroke, and hemorrhagic stroke. Stroke was defined as a focal neurological deficit in an area consistent with the findings of diagnostic imaging or autopsy. TIA was defined as a focal neurological deficit in an area corresponding to a major cerebral artery that lasted for <24 hours and, if applicable, no evidence of infarction or hemorrhage on diagnostic imaging. MI was defined as acute chest pain with 1 of 1) new-onset ST-segment elevation on EKG, 2) >threefold increase in Troponin T from baseline, or 3) MI as cause of death on autopsy. Major bleeding was defined according to the ISTH criteria as bleeding leading to a hemoglobin drop of ≥20 g/L, transfusion of ≥2 units of packed red blood cells, symptomatic bleeding into a closed compartment (such as the cranium or retroperitoneum), or death.17 

Identification of events

Data on study outcomes were gathered by 4 separate pathways. First, by use of a thorough International Classification of Diseases, 10th revision (ICD-10) code search (from hospital discharges and emergency department visits) (supplemental Table 1). Second, by querying the Icelandic death registry. Third, by examining endoscopic procedures for GIB. Fourth, by examining computed tomographies of the head and pulmonary arteries to identify missed diagnoses of stroke, intracranial hemorrhage, or pulmonary embolism. Each diagnosis was reviewed and confirmed by manual chart review. Events were excluded if they occurred before the start of the patient's follow-up, the patient had not been receiving OACs in the preceding 2 days, or a bleeding event did not fulfil the ISTH criteria.

For comparison, we identified events by using only previously verified ICD-10 codes and without manual chart review, as reported by other investigators.14,18,19 The sensitivity, specificity, positive predictive value (PPV), and negative predictive value for that method were calculated in comparison to our more robust searching algorithm.

Data on baseline characteristics (ie, treatment indication, comorbidities, concomitant drug use, and area of residence) were collected from the start of the patient's follow-up or earlier (Figure 1; supplemental Tables 1 and 2). The data acquisition for these variables has been previously described in detail.16 

Figure 1.

Cohort creation diagram. ∗The exclusion criteria were residence outside of Iceland at the index date, prior use of oral anticoagulants within 12 months of the index date, prescription of 2.5 mg of rivaroxaban at the index date, and diagnosis of end-stage renal disease, a mechanical heart valve, or mitral stenosis from the index date or earlier.

Figure 1.

Cohort creation diagram. ∗The exclusion criteria were residence outside of Iceland at the index date, prior use of oral anticoagulants within 12 months of the index date, prescription of 2.5 mg of rivaroxaban at the index date, and diagnosis of end-stage renal disease, a mechanical heart valve, or mitral stenosis from the index date or earlier.

Close modal

Statistical analysis

To account for potential indication bias, inverse probability weighting (IPW) was used to yield balanced study groups. IPW assigns weights to patients based on propensity scores calculated from potential confounders, thereby creating a balanced pseudopopulation that includes the whole study population. The model accounted for age, sex, DOAC dosing (standard or low dose), all variables in the Charlson comorbidity index (except for AIDS, which was too sporadic), bleeding or coagulation disorders, hypertension, history of venous thromboembolism or GIB, concomitant drug use (antihistamines, antiplatelets, corticosteroids, nonsteroidal anti-inflammatory drugs, proton pump inhibitors, selective serotonin reuptake inhibitors, and statins), and area of residence. Study outcomes were compared using propensity score-weighted Cox regression, and the data were visualized using propensity score-weighted Kaplan–Meier survival curves. Finally, the E-value was calculated to estimate the effect of any residual confounding factors.20 All statistical tests were 2-tailed and all CIs were 95%. Statistical analysis was performed in R, version 4.1.2 (R Foundation for Statistical Computing), using RStudio, version 2021.09.2.

Study population

In total, 8892 patients filled an OAC prescription during the study period. Of those, 2869 patients were excluded because they had received an OAC in the preceding 12 months, 1256 patients were excluded because they had treatment indication other than atrial fibrillation, and 97 patients were excluded for other reasons, leaving 4670 patients in the final study cohort (Figure 2). Of those, 1787 patients received apixaban, 420 received dabigatran, and 2463 patients received rivaroxaban. IPW yielded balanced study groups with an average weighted follow-up period of 1.3 years for patients receiving apixaban, 1.9 years for patients receiving dabigatran, and 1.8 years for patients receiving rivaroxaban (Table 1).

Figure 2.

Flowchart for study selection. AF, atrial fibrillation; DOACs, direct oral anticoagulants; OAC, oral anticoagulant.

Figure 2.

Flowchart for study selection. AF, atrial fibrillation; DOACs, direct oral anticoagulants; OAC, oral anticoagulant.

Close modal
Table 1.

Baseline characteristics of the study population 

Apixaban (n = 1787)Dabigatran (n = 420)Rivaroxaban (n = 2463)SMD 
BeforeAfter
Age 73.8 (12.0) 71.3 (12.0) 70.7 (11.4) 0.175 0.074 
Sex (% male) 976 (54.6) 245 (58.3) 1526 (62.0) 0.099 0.089 
CHA2DS2-VASc score 2.8 (1.6) 2.6 (1.5) 2.4 (1.5) 0.202 0.082 
Charlson comorbidity index 0.9 (1.4) 0.7 (1.1) 0.6 (1.1) 0.156 0.082 
Ischemic heart disease 142 (7.9) 34 (8.1) 187 (7.6) 0.012 0.013 
Congestive heart failure 158 (8.8) 34 (8.1) 156 (6.3) 0.063 0.025 
Peripheral vascular disease 81 (4.5) 16 (3.8) 91 (3.7) 0.028 0.024 
Cerebrovascular disease 207 (11.6) 25 (6.0) 125 (5.1) 0.159 0.038 
Hemiplegia 15 (0.8) 2 (0.5) 8 (0.3) 0.046 0.020 
Diabetes mellitus 84 (4.7) 22 (5.2) 84 (3.4) 0.060 0.033 
Diabetes mellitus with end-organ damage 50 (2.8) 12 (2.9) 57 (2.3) 0.023 0.043 
Chronic lung disease 105 (5.9) 20 (4.8) 90 (3.7) 0.070 0.030 
Moderate/severe renal disease 61 (3.4) 9 (2.1) 59 (2.4) 0.052 0.066 
Liver disease 17 (1.0) 0 (0.0) 13 (0.5) 0.097 0.084 
Peptic ulcer disease 42 (2.4) 9 (2.1) 27 (1.1) 0.064 0.013 
Connective tissue disease 35 (2.0) 8 (1.9) 45 (1.8) 0.006 0.028 
Dementia 45 (2.5) 6 (1.4) 25 (1.0) 0.077 0.060 
Any tumor 207 (11.6) 38 (9.0) 191 (7.8) 0.087 0.055 
Metastatic solid tumor 8 (0.4) 1 (0.2) 12 (0.5) 0.028 0.032 
Hypertension 1245 (69.7) 298 (71.0) 1613 (65.5) 0.078 0.023 
Bleeding disease 13 (0.7) 1 (0.2) 10 (0.4) 0.048 0.044 
Prior GIB 80 (4.5) 19 (4.5) 68 (2.8) 0.063 0.039 
Prior VTE 27 (1.5) 4 (1.0) 59 (2.4) 0.076 0.028 
Dosing (low dose) 394 (22.0) 190 (45.2) 515 (20.9) 0.356 0.040 
Concomitant drug use      
Antihistamines 9 (0.5) 5 (1.2) 16 (0.6) 0.050 0.019 
Antiplatelets 488 (27.3) 114 (27.1) 532 (21.6) 0.089 0.021 
Corticosteroids 356 (19.9) 82 (19.5) 455 (18.5) 0.025 0.031 
NSAIDs 384 (21.5) 87 (20.7) 586 (23.8) 0.049 0.045 
PPIs 735 (41.1) 158 (37.6) 882 (35.8) 0.073 0.024 
SSRIs 313 (17.5) 46 (11.0) 322 (13.1) 0.126 0.131 
Statins 829 (46.4) 198 (47.1) 1088 (44.2) 0.040 0.021 
Area of residence    0.319 0.129 
Capital area 1269 (71.0) 258 (61.4) 1529 (62.1)   
Eastern 58 (3.2) 15 (3.6) 58 (2.4)   
Northern 120 (6.7) 46 (11.0) 361 (14.7)   
Southern 220 (12.3) 90 (21.4) 335 (13.6)   
Western 87 (4.9) 10 (2.4) 135 (5.5)   
Westfjords 33 (1.8) 1 (0.2) 45 (1.8)   
Apixaban (n = 1787)Dabigatran (n = 420)Rivaroxaban (n = 2463)SMD 
BeforeAfter
Age 73.8 (12.0) 71.3 (12.0) 70.7 (11.4) 0.175 0.074 
Sex (% male) 976 (54.6) 245 (58.3) 1526 (62.0) 0.099 0.089 
CHA2DS2-VASc score 2.8 (1.6) 2.6 (1.5) 2.4 (1.5) 0.202 0.082 
Charlson comorbidity index 0.9 (1.4) 0.7 (1.1) 0.6 (1.1) 0.156 0.082 
Ischemic heart disease 142 (7.9) 34 (8.1) 187 (7.6) 0.012 0.013 
Congestive heart failure 158 (8.8) 34 (8.1) 156 (6.3) 0.063 0.025 
Peripheral vascular disease 81 (4.5) 16 (3.8) 91 (3.7) 0.028 0.024 
Cerebrovascular disease 207 (11.6) 25 (6.0) 125 (5.1) 0.159 0.038 
Hemiplegia 15 (0.8) 2 (0.5) 8 (0.3) 0.046 0.020 
Diabetes mellitus 84 (4.7) 22 (5.2) 84 (3.4) 0.060 0.033 
Diabetes mellitus with end-organ damage 50 (2.8) 12 (2.9) 57 (2.3) 0.023 0.043 
Chronic lung disease 105 (5.9) 20 (4.8) 90 (3.7) 0.070 0.030 
Moderate/severe renal disease 61 (3.4) 9 (2.1) 59 (2.4) 0.052 0.066 
Liver disease 17 (1.0) 0 (0.0) 13 (0.5) 0.097 0.084 
Peptic ulcer disease 42 (2.4) 9 (2.1) 27 (1.1) 0.064 0.013 
Connective tissue disease 35 (2.0) 8 (1.9) 45 (1.8) 0.006 0.028 
Dementia 45 (2.5) 6 (1.4) 25 (1.0) 0.077 0.060 
Any tumor 207 (11.6) 38 (9.0) 191 (7.8) 0.087 0.055 
Metastatic solid tumor 8 (0.4) 1 (0.2) 12 (0.5) 0.028 0.032 
Hypertension 1245 (69.7) 298 (71.0) 1613 (65.5) 0.078 0.023 
Bleeding disease 13 (0.7) 1 (0.2) 10 (0.4) 0.048 0.044 
Prior GIB 80 (4.5) 19 (4.5) 68 (2.8) 0.063 0.039 
Prior VTE 27 (1.5) 4 (1.0) 59 (2.4) 0.076 0.028 
Dosing (low dose) 394 (22.0) 190 (45.2) 515 (20.9) 0.356 0.040 
Concomitant drug use      
Antihistamines 9 (0.5) 5 (1.2) 16 (0.6) 0.050 0.019 
Antiplatelets 488 (27.3) 114 (27.1) 532 (21.6) 0.089 0.021 
Corticosteroids 356 (19.9) 82 (19.5) 455 (18.5) 0.025 0.031 
NSAIDs 384 (21.5) 87 (20.7) 586 (23.8) 0.049 0.045 
PPIs 735 (41.1) 158 (37.6) 882 (35.8) 0.073 0.024 
SSRIs 313 (17.5) 46 (11.0) 322 (13.1) 0.126 0.131 
Statins 829 (46.4) 198 (47.1) 1088 (44.2) 0.040 0.021 
Area of residence    0.319 0.129 
Capital area 1269 (71.0) 258 (61.4) 1529 (62.1)   
Eastern 58 (3.2) 15 (3.6) 58 (2.4)   
Northern 120 (6.7) 46 (11.0) 361 (14.7)   
Southern 220 (12.3) 90 (21.4) 335 (13.6)   
Western 87 (4.9) 10 (2.4) 135 (5.5)   
Westfjords 33 (1.8) 1 (0.2) 45 (1.8)   

GIB, gastrointestinal bleeding; NSAID, nonsteroidal anti-inflammatory drug; PPI, proton pump inhibitor; SMD, standardized mean difference; SSRI, selective serotonin receptor inhibitor; VTE, venous thromboembolism.

Data are represented as either mean (standard deviation) or n (%).

SMD <0.1 was considered ideal balance, whereas SMD <0.2 was considered acceptable balance.

A total of 44 stroke/SE events were identified during the follow-up period. In total, 38 (86%) events were identified using ICD-10 codes and 4 (9%) from the death registry. In addition, 2 (5%) events were identified during chart review of another diagnosis.

Overall, 191 major bleeding events were identified during the follow-up period. Of those, 137 (72%) originated from the gastrointestinal tract, 28 (15%) had an intracranial location, and 26 (14%) originated from other locations. Of the 191 major bleeding episodes, 159 (83%) were identified using ICD-10 codes, 25 (13%) from endoscopic procedure codes, and 4 (2%) from the death registry. In addition, 3 (2%) events were identified during chart review of another diagnosis.

When only previously validated primary discharge ICD-10 codes were used to identify events and without confirmation by manual chart review, 46 stroke/SE and 73 major bleeding events were identified. Compared with our robust searching algorithm, this method had a sensitivity of 52.3% for identifying any stroke/SE, and sensitivity of 30.4% for any major bleeding (Table 2). The specificity was 99.5% and 99.7% for stroke/SE and major bleeding events, respectively. The PPV for stroke/SE was 50.0%. The PPV was 79.5% for any major bleeding. Specifically, the PPV was 100% for intracranial hemorrhage, 90.2% for GIB, and 39.1% for bleeding from other locations. For other outcomes, the PPV ranged from 14.3% for venous thromboembolism to 91.7% for MI. The negative predictive value was 99.5% for stroke/SE and 97.1% for major bleeding (Table 2).

Table 2.

Sensitivity, specificity, and predictive values of conventional ICD-10 code searches compared with our robust searching algorithm

OutcomeSensitivity (%)Specificity (%)Positive predictive value (%)Negative predictive value (%)
Only primary discharge diagnoses 
Major bleeding 30.4 99.7 79.5 97.1 
Gastrointestinal bleeding 17.5 99.9 90.2 96.2 
Intracranial hemorrhage 48.3 100 100 99.7 
Other bleeding 34.6 99.7 39.1 99.6 
Any stroke or systemic embolism 52.3 99.5 50.0 99.5 
Ischemic stroke 52.9 99.5 43.9 99.7 
Hemorrhagic stroke 40.0 100 100 99.9 
Arterial thromboembolism 100 ∼100 50.0 100 
Myocardial infarction 54.1 99.9 91.7 99.4 
Venous thromboembolism 10.0 99.9 14.3 99.8 
All hospital discharge codes and codes from emergency department visits 
Major bleeding 64.9 96.4 43.4 98.5 
Gastrointestinal bleeding 51.8 99.1 62.3 98.6 
Intracranial hemorrhage 89.7 99.9 86.7 99.9 
Other bleeding 88.5 96.9 15.2 99.9 
Any stroke or systemic embolism 81.8 98.8 39.1 99.8 
Ischemic stroke 82.4 98.8 34.1 99.9 
Hemorrhagic stroke 70.0 99.9 63.6 99.9 
Arterial thromboembolism 100 ∼100 50.0 100 
Myocardial infarction 78.7 99.6 71.6 99.7 
Venous thromboembolism 70.0 99.6 29.2 99.9 
OutcomeSensitivity (%)Specificity (%)Positive predictive value (%)Negative predictive value (%)
Only primary discharge diagnoses 
Major bleeding 30.4 99.7 79.5 97.1 
Gastrointestinal bleeding 17.5 99.9 90.2 96.2 
Intracranial hemorrhage 48.3 100 100 99.7 
Other bleeding 34.6 99.7 39.1 99.6 
Any stroke or systemic embolism 52.3 99.5 50.0 99.5 
Ischemic stroke 52.9 99.5 43.9 99.7 
Hemorrhagic stroke 40.0 100 100 99.9 
Arterial thromboembolism 100 ∼100 50.0 100 
Myocardial infarction 54.1 99.9 91.7 99.4 
Venous thromboembolism 10.0 99.9 14.3 99.8 
All hospital discharge codes and codes from emergency department visits 
Major bleeding 64.9 96.4 43.4 98.5 
Gastrointestinal bleeding 51.8 99.1 62.3 98.6 
Intracranial hemorrhage 89.7 99.9 86.7 99.9 
Other bleeding 88.5 96.9 15.2 99.9 
Any stroke or systemic embolism 81.8 98.8 39.1 99.8 
Ischemic stroke 82.4 98.8 34.1 99.9 
Hemorrhagic stroke 70.0 99.9 63.6 99.9 
Arterial thromboembolism 100 ∼100 50.0 100 
Myocardial infarction 78.7 99.6 71.6 99.7 
Venous thromboembolism 70.0 99.6 29.2 99.9 

Comparison of thromboembolic and mortality rates

Rates of any stroke/SE were similar among the 3 drugs (Figures 3A and 4). Specifically, the rates of ischemic stroke or hemorrhagic stroke were not markedly different between groups, although the study was likely underpowered for this analysis. Interestingly, dabigatran was associated with >twofold higher rates of MI compared with both apixaban (1.4 events/100 person-years (py) vs 0.7 events/100-py, hazard ratio [HR] 2.26, 95% CI, 0.90-5.67) and rivaroxaban (1.4 events/100-py vs 0.7 events/100-py, HR 2.21, 95% CI 1.00-4.90), although the former comparison included the possibility of a null effect (Figure 4). All-cause mortality was similar among the drugs (Figure 4). Dabigatran was associated with twofold higher vascular mortality compared with apixaban and rivaroxaban, although the CIs had a wide range for these comparisons and included the possibility of a null effect in both instances (Figure 4). The number of venous thromboembolic and TIA events was too low to make any meaningful comparisons among the groups (supplemental Table 3).

Figure 3.

Kaplan-Meier cumulative event plots comparing the primary study outcomes. (A) compares rates of any stroke or systemic embolism (SE) for patients receiving apixaban, dabigatran, and rivaroxaban. (B) compares rates of any major bleeding between patients receiving apixaban, dabigatran, and rivaroxaban.

Figure 3.

Kaplan-Meier cumulative event plots comparing the primary study outcomes. (A) compares rates of any stroke or systemic embolism (SE) for patients receiving apixaban, dabigatran, and rivaroxaban. (B) compares rates of any major bleeding between patients receiving apixaban, dabigatran, and rivaroxaban.

Close modal
Figure 4.

Propensity score-weighted incidence rates and hazard ratios of thromboembolic and mortality rates. Comparison between patients receiving apixaban, dabigatran, and rivaroxaban.

Figure 4.

Propensity score-weighted incidence rates and hazard ratios of thromboembolic and mortality rates. Comparison between patients receiving apixaban, dabigatran, and rivaroxaban.

Close modal

Comparison of major bleeding rates

Rivaroxaban was associated with higher rates of major bleeding compared with apixaban (2.9 events/100-py vs 1.8 events/100-py, HR 1.64, 95% CI, 1.13-2.37) and dabigatran (2.9 events/100-py vs 1.4 events/100-py, HR 2.18, 95% CI, 1.21-3.93) (Figures 3B and 5). Rivaroxaban users had higher rates of both major GIB and other major bleeding events compared with apixaban. No differences were noted in intracranial hemorrhage rates among the 3 drugs (Figure 5).

Figure 5.

Propensity score-weighted incidence rates and hazard ratios of major bleeding rates. Comparison between patients receiving apixaban, dabigatran, and rivaroxaban.

Figure 5.

Propensity score-weighted incidence rates and hazard ratios of major bleeding rates. Comparison between patients receiving apixaban, dabigatran, and rivaroxaban.

Close modal

Estimation of the effect of potential confounders

To estimate the effect of potential confounders, the E-value was calculated. The E-value for the comparison of major bleeding rates between rivaroxaban and apixaban users was 2.66 for the point estimate and 1.51 for the lower limit of the 95% CI. This suggests that an unmeasured confounder, unrelated to the covariates included in the IPW model, would have to be 166% more common in the rivaroxaban group and increase the risk for major bleeding by 166% to explain away the observed difference, or be 51% more common in the rivaroxaban group and increase the risk for major bleeding by 51%, for the CI to include the possibility of a null effect. Similarly, for the comparison between dabigatran and rivaroxaban users, the E-value for the point estimate was 3.79 and 1.72 for the lower limit of the CI.

For the comparison of MI rates between rivaroxaban and dabigatran, the E-value was 3.85 for the point estimate and 1.02 for the lower limit of the CI.

In this nationwide propensity score-weighted cohort study, rivaroxaban was associated with higher rates of major bleeding compared with apixaban and dabigatran. Additionally, dabigatran users had twofold higher rates of MI compared with users of apixaban and rivaroxaban, although the former comparison included the possibility of a null effect. Importantly, the rates of stroke/SE were similar among the 3 drugs.

The similar rates of stroke/SE observed in this study are consistent with previous population-based studies from Denmark, the United Kingdom, and Chinese Taipei.7,8,21 However, it contrasts with a study from the USA which found apixaban to be associated with lower rates of ischemic stroke or SE compared with those of rivaroxaban.6 That study was based on the Optum database, which mostly includes privately insured patients, and may therefore be susceptible to selection bias.6 Given the comparable effectiveness of the 3 drugs, oral anticoagulant selection may be guided by the different bleeding and MI risks of the 3 drugs.

The twofold higher rates of MI compared with apixaban and rivaroxaban in the current study contrasts with previous registry studies from the USA, France, and Chinese Taipei that failed to demonstrate significantly different MI rates between dabigatran and rivaroxaban.21-23 The discrepancy between previous studies and this one might, at least partly, be explained by differences in study design. The study from the US was based on patients aged ≥65 years receiving health insurance through the Medicare insurance coverage and only assessed comorbid conditions using relevant ICD-9 codes within 6 months of cohort entry.22 This is likely to lead to missed diagnoses of important baseline covariates. The study from France was limited to the first year after marketing, which, owing to the increased usage of DOACs in recent years, is unlikely to be representative of today's DOAC population.23 Finally, the study from Chinese Taipei was based on a population that was likely at reduced risk of MI compared with that of this study. In that study, only 3% of rivaroxaban and dabigatran users had prior history of MI and 41% to 45% were receiving concomitant antiplatelet treatment. For comparison, 7.6% to 8.1% of rivaroxaban and dabigatran users had history of previous MI in this study and 22% to 27% of patients were receiving concomitant antiplatelet treatment. Therefore, MI rates were, perhaps unsurprisingly, twice as common in this study compared with the one from Chinese Taipei. However, these differences in incidence rates are unlikely to be only because of differences in the study populations. Given that our searching algorithm identified 69% more MI events compared with using ICD-10 codes alone (61 events vs 36 events), previous observational studies have likely suffered from low sensitivity.

A similar discrepancy has been observed between RCTs and observational studies examining MI rates between dabigatran and warfarin. Although a meta-analysis of 14 RCTs demonstrated that dabigatran was associated with higher MI rates compared with warfarin,24 a second meta-analysis limited to observational studies failed to observe any difference in MI rates between the 2 drugs.25 The increased MI risk of dabigatran has been hypothesized to be because of platelet activation. It has been shown that dabigatran increases thromboxane excretion, a marker of platelet activation whereas warfarin usage does not affect thromboxane excretion.26 Similarly, 3 studies have demonstrated increased platelet aggregation in patients after dabigatran initiation but not in rivaroxaban or warfarin users.27-29 These results were replicated using human atherosclerotic plaque homogenates and a mouse arterial injury model using intravital microscopy.29 

The higher rates of major bleeding observed for rivaroxaban compared with other DOACs in this study is generally consistent with previous observational studies comparing rates of bleeding events requiring hospitalization.5-10 Although a registry study from Denmark found no difference in bleeding rates among the 3 drugs,8 rivaroxaban has been associated with higher bleeding rates compared with apixaban and/or dabigatran in previous studies from the USA and United Kingdom.5-7,10 The higher bleeding rates of rivaroxaban has been suggested to be because of the different pharmacokinetics of the drugs,6,16 but rivaroxaban is administered once daily whereas apixaban and dabigatran are both administered twice daily. This may cause a higher peak plasma concentration making these patients more susceptible to bleeding. Indeed, previous studies have demonstrated that rivaroxaban has higher peak plasma concentration, higher maximal anti-Xa activity, and higher 24-hour area under the curve for anti-Xa activity compared with apixaban.30,31 

This study has several strengths. A centralized national drug prescription database was used that allowed for identification of all DOAC users in the country. Furthermore, study outcomes were gathered from all the major hospitals in Iceland, using a robust searching algorithm that, in addition to the traditional method of using ICD-10 codes, included reviewing diagnostic imaging studies and endoscopic procedures undertaken during the study period, and searching the Icelandic death registry. This study also used the ISTH definition for major bleeding similar to the pivotal RCTs but opposed to most observational studies that have defined major bleeding as a bleeding that leads to hospitalization. In addition, all outcomes were manually verified, greatly increasing the accuracy of the data. In comparison, previous studies have identified outcome events using only a few specific ICD-10 primary discharge codes and without manual verification of events.14,18,19 Although this method is highly specific, the sensitivity was only 52.3% for identification of stroke/SE and 30.4% for identification of major bleeding compared with our searching algorithm. In addition, the PPV was suboptimal for this method, that is, 50.0% for stroke/SE and 79.5% for major bleeding. The low PPV for major bleeding was mostly explained by the fact that a considerable proportion of bleeding events that required hospitalization did not fulfil the ISTH criteria for major bleeding. Meanwhile, the low PPV for stroke/SE was mostly explained by the fact that patients were often issued ICD-10 codes for stroke/SE events that had occurred before the start of the patient's follow-up, ie, a stroke event that led to the patient starting DOAC treatment. This underlines the importance of manual verification of study outcomes and suggests that the results of previous observational studies may be unreliable.

The study also has several limitations. First, although a robust IPW model was used to account for indication bias, it cannot be excluded that some unmeasured confounding exists. For example, this study did not account for socioeconomic or lifestyle variables, such as smoking, alcohol consumption, or obesity. However, because all DOACs in Iceland have equivalent prices and are reimbursed through a universal health insurance, the risk of bias owing to socioeconomic status is unlikely. Second, the study did not have information on baseline laboratory values such as hemoglobin or creatinine values. It did, however, account for prior history of bleeding and diagnosis of chronic kidney disease. Third, data on over-the-counter medication usage were not available. This is important because over-the-counter usage of both nonsteroidal anti-inflammatory drugs and proton pump inhibitors is relatively common in Iceland. However, there was no difference between the groups in concomitant drug prescriptions of nonsteroidal anti-inflammatory drugs or of proton pump inhibitors before or after IPW, making potential differences in over-the-counter consumption less likely. Lastly, the dabigatran group was relatively small compared with previous registry studies.

In conclusion, rates of stroke/SE were similar among DOACs. Rivaroxaban was associated with higher rates of major bleeding compared with other DOACs, but lower rates of MI compared with dabigatran. These results may help guide oral anticoagulant selection, especially in patients at high risk of bleeding or MI.

This study was funded by the Landspitali University Hospital Research Fund (A-2018-012 to E.S.B.) and by the Icelandic Centre for Research (207113-051 to A.B.I.). The funding source had no role in the design, conduct, or reporting of the study.

Contributions: A.B.I., E.S.B., J.P.H., and P.T.O. designed the study; A.B.I. and E.S.B. were involved in obtaining funding for the study; A.B.I., A.S.A., E.R., D.A.P., I.E.R., and B.R.G. performed the data acquisition; A.B.I. performed the statistical analysis and visualized the data with help from J.P.H. and S.H.L.; A.B.I. wrote the first draft of the manuscript; All other coauthors critically reviewed the manuscript, had access to the primary data, and approved the final manuscript before submission.

Conflicts of interest: P.T.O. and B.R.G. together with the Landspitali University Hospital and the University of Iceland hold a patent for the FIIX-prothrombin measurement. All other authors declare no competing interests.

Correspondence: Einar S. Bjornsson, MD, PhD, Professor of Gastroenterology and Hepatology, University of Iceland, Faculty of Medicine, Chief Physician, Landspitali University Hospital, Department of Gastroenterology and Hepatology; e-mail: einarsb@landspitali.is.

1.
Connolly
SJ
,
Ezekowitz
MD
,
Yusuf
S
, et al
.
Dabigatran versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2009
;
361
(
12
):
1139
-
1151
.
2.
Granger
CB
,
Alexander
JH
,
McMurray
JJ
, et al
.
Apixaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2011
;
365
(
11
):
981
-
992
.
3.
Giugliano
RP
,
Ruff
CT
,
Braunwald
E
, et al
.
Edoxaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
.
2013
;
369
(
22
):
2093
-
2104
.
4.
Patel
MR
,
Mahaffey
KW
,
Garg
J
, et al
.
Rivaroxaban versus warfarin in nonvalvular atrial fibrillation
.
N Engl J Med
.
2011
;
365
(
10
):
883
-
891
.
5.
Dawwas
GK
,
Leonard
CE
,
Lewis
JD
,
Cuker
A
.
Risk for recurrent venous thromboembolism and bleeding with apixaban compared with rivaroxaban: an analysis of real-world data
.
Ann Intern Med
.
2022
;
175
(
1
):
20
-
28
.
6.
Fralick
M
,
Colacci
M
,
Schneeweiss
S
,
Huybrechts
KF
,
Lin
KJ
,
Gagne
JJ
.
Effectiveness and safety of apixaban compared with rivaroxaban for patients with atrial fibrillation in routine practice: a cohort study
.
Ann Intern Med
.
2020
;
172
(
7
):
463
-
473
.
7.
Vinogradova
Y
,
Coupland
C
,
Hill
T
,
Hippisley-Cox
J
.
Risks and benefits of direct oral anticoagulants versus warfarin in a real world setting: cohort study in primary care
.
BMJ
.
2018
;
362
(
8160
):
k2505
.
8.
Andersson
NW
,
Svanstrom
H
,
Lund
M
,
Pasternak
B
,
Melbye
M
.
Comparative effectiveness and safety of apixaban, dabigatran, and rivaroxaban in patients with non-valvular atrial fibrillation
.
Int J Cardiol
.
2018
;
268
:
113
-
119
.
9.
Bai
Y
,
Deng
H
,
Shantsila
A
,
Lip
GY
.
Rivaroxaban versus dabigatran or warfarin in real-world studies of stroke prevention in atrial fibrillation: systematic review and meta-analysis
.
Stroke
.
2017
;
48
(
4
):
970
-
976
.
10.
Mueller
T
,
Alvarez-Madrazo
S
,
Robertson
C
,
Wu
O
,
Bennie
M
.
Comparative safety and effectiveness of direct oral anticoagulants in patients with atrial fibrillation in clinical practice in Scotland
.
Br J Clin Pharmacol
.
2019
;
85
(
2
):
422
-
431
.
11.
Halvorsen
S
,
Ghanima
W
,
Fride Tvete
I
, et al
.
A nationwide registry study to compare bleeding rates in patients with atrial fibrillation being prescribed oral anticoagulants
.
Eur Heart J Cardiovasc Pharmacother
.
2017
;
3
(
1
):
28
-
36
.
12.
Ingason
AB
,
Hreinsson
JP
,
Ágústsson
AS
, et al
.
Warfarin is associated with higher rates of upper but not lower gastrointestinal bleeding compared with direct oral anticoagulants: a population-based propensity-weighted cohort study
.
Clin Gastroenterol Hepatol
.
2023
;
21
(
2
):
347
-
357
.
13.
Larsen
TB
,
Skjoth
F
,
Kjaeldgaard
JN
,
Lip
GYH
,
Nielsen
PB
,
Sogaard
M
.
Effectiveness and safety of rivaroxaban and warfarin in patients with unprovoked venous thromboembolism: a propensity-matched nationwide cohort study
.
Lancet Haematol
.
2017
;
4
(
5
):
e237
-
e244
.
14.
Nielsen
PB
,
Skjoth
F
,
Sogaard
M
,
Kjaeldgaard
JN
,
Lip
GY
,
Larsen
TB
.
Effectiveness and safety of reduced dose non-vitamin K antagonist oral anticoagulants and warfarin in patients with atrial fibrillation: propensity weighted nationwide cohort study
.
BMJ
.
2017
;
356
(
8094
):
j510
.
15.
Souverein
PC
,
van den Ham
HA
,
Huerta
C
, et al
.
Comparing risk of major bleeding between users of different oral anticoagulants in patients with nonvalvular atrial fibrillation
.
Br J Clin Pharmacol
.
2020
;
87
(
3
):
988
-
1000
.
16.
Ingason
AB
,
Hreinsson
JP
,
Ágústsson
AS
, et al
.
Rivaroxaban is associated with higher rates of gastrointestinal bleeding than other direct oral anticoagulants: a nationwide propensity score-weighted study
.
Ann Intern Med
.
2021
;
174
(
11
):
1493
-
1502
.
17.
Schulman
S
,
Kearon
C
.
Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients
.
J Thromb Haemost
.
2005
;
3
(
4
):
692
-
694
.
18.
Sjalander
S
,
Sjogren
V
,
Renlund
H
,
Norrving
B
,
Sjalander
A
.
Dabigatran, rivaroxaban and apixaban vs. high TTR warfarin in atrial fibrillation
.
Thromb Res
.
2018
;
167
:
113
-
118
.
19.
Maura
G
,
Blotiere
PO
,
Bouillon
K
, et al
.
Comparison of the short-term risk of bleeding and arterial thromboembolic events in nonvalvular atrial fibrillation patients newly treated with dabigatran or rivaroxaban versus vitamin K antagonists: a French nationwide propensity-matched cohort study
.
Circulation
.
2015
;
132
(
13
):
1252
-
1260
.
20.
VanderWeele
TJ
,
Ding
P
.
Sensitivity analysis in observational research: introducing the E-value
.
Ann Intern Med
.
2017
;
167
(
4
):
268
-
274
.
21.
Chan
YH
,
Kuo
CT
,
Yeh
YH
, et al
.
Thromboembolic, bleeding, and mortality risks of rivaroxaban and dabigatran in Asians with nonvalvular atrial fibrillation
.
J Am Coll Cardiol
.
2016
;
68
(
13
):
1389
-
1401
.
22.
Graham
DJ
,
Reichman
ME
,
Wernecke
M
, et al
.
Stroke, bleeding, and mortality risks in elderly Medicare beneficiaries treated with dabigatran or rivaroxaban for nonvalvular atrial fibrillation
.
JAMA Intern Med
.
2016
;
176
(
11
):
1662
-
1671
.
23.
Blin
P
,
Dureau-Pournin
C
,
Cottin
Y
, et al
.
Comparative effectiveness and safety of standard or reduced dose dabigatran vs. rivaroxaban in nonvalvular atrial fibrillation
.
Clin Pharmacol Ther
.
2019
;
105
(
6
):
1439
-
1455
.
24.
Douxfils
J
,
Buckinx
F
,
Mullier
F
, et al
.
Dabigatran etexilate and risk of myocardial infarction, other cardiovascular events, major bleeding, and all-cause mortality: a systematic review and meta-analysis of randomized controlled trials
.
J Am Heart Assoc
.
2014
;
3
(
3
):
e000515
.
25.
Carmo
J
,
Moscoso Costa
F
,
Ferreira
J
,
Mendes
M
.
Dabigatran in real-world atrial fibrillation. Meta-analysis of observational comparison studies with vitamin K antagonists
.
Thromb Haemost
.
2016
;
116
(
4
):
754
-
763
.
26.
Ezekowitz
MD
,
Reilly
PA
,
Nehmiz
G
, et al
.
Dabigatran with or without concomitant aspirin compared with warfarin alone in patients with nonvalvular atrial fibrillation (PETRO Study)
.
Am J Cardiol
.
2007
;
100
(
9
):
1419
-
1426
.
27.
Achilles
A
,
Mohring
A
,
Dannenberg
L
, et al
.
Dabigatran enhances platelet reactivity and platelet thrombin receptor expression in patients with atrial fibrillation
.
J Thromb Haemost
.
2017
;
15
(
3
):
473
-
476
.
28.
Olivier
CB
,
Weik
P
,
Meyer
M
, et al
.
TRAP-induced platelet aggregation is enhanced in cardiovascular patients receiving dabigatran
.
Thromb Res
.
2016
;
138
:
63
-
68
.
29.
Petzold
T
,
Thienel
M
,
Konrad
I
, et al
.
Oral thrombin inhibitor aggravates platelet adhesion and aggregation during arterial thrombosis
.
Sci Transl Med
.
2016
;
8
(
367
). 367ra168.
30.
Frost
C
,
Song
Y
,
Barrett
YC
, et al
.
A randomized direct comparison of the pharmacokinetics and pharmacodynamics of apixaban and rivaroxaban
.
Clin Pharmacol
.
2014
;
6
:
179
-
187
.
31.
Kreutz
R
,
Persson
PB
,
Kubitza
D
, et al
.
Dissociation between the pharmacokinetics and pharmacodynamics of once-daily rivaroxaban and twice-daily apixaban: a randomized crossover study
.
J Thromb Haemost
.
2017
;
15
(
10
):
2017
-
2028
.

Author notes

Study protocol and statistical code are available from E.S.B. through email requests (einarsb@landspitali.is). Data set is available by license through the National Bioethics Committee of Iceland (vsn@vsn.is).

The full-text version of this article contains a data supplement.

Supplemental data