Abstract
Background
The COOL-AF (Cohort of Antithrombotic Use and Optimal International Normalized Ratio Levels in Patients with Atrial Fibrillation) risk scores for death, bleeding, and thromboembolic events (TEs) were derived from the COOL-AF cohort from Thailand and require external validation.
Objectives
The authors sought to externally validate the COOL-AF scores in the APHRS (Asia-Pacific Heart Rhythm Society) registry and to compare their performance in the ESC-EHRA (European Society of Cardiology-European Heart Rhythm Association) EORP-AF (EURObservational Research Programme in Atrial Fibrillation) General Long-Term Registry.
Methods
We studied 3,628 APHRS and 8,825 EORP-AF patients. Receiver operating characteristic (ROC) curves and Cox regression analyses were used to test the predictive value of COOL-AF scores and to compared them with the CHA2DS2-VASc and HAS-BLED scores.
Results
Patients in the EORP-AF were older, had a higher prevalence of male sex, and were at higher thromboembolic and hemorrhagic risk than APHRS patients. After 1 year of follow-up in APHRS and EORP-AF, the following events were recorded: 87 (2.4%) and 435 (4.9%) death for any causes, 37 (1.0%) and 111 (1.3%) major bleeding, and 25 (0.7%) and 109 (1.2%) TEs, respectively. In APHRS, the COOL-AF scores showed moderate-to-good predictive value for all-cause mortality (area under the curve [AUC]: 0.77; 95% CI: 0.71-0.83), major bleeding (AUC: 0.68; 95% CI: 0.60-0.76), and TEs (AUC: 0.61; 95% CI: 0.51-0.71), and were similar to the CHA2DS2-VASc and HAS-BLED scores. In EORP-AF, the predictive value of COOL-AF for all-cause mortality (AUC: 0.68; 95% CI: 0.65-0.70) and major bleeding (AUC: 0.61; 95% CI: 0.60-0.62) was modest and lower than in APHRS. In EORP-AF, the COOL-AF score for TE was inferior to the CHA2DS2-VASc score.
Conclusions
The COOL-AF risk scores may be an easy tool to identify Asian patients with AF at risk for death and major bleeding and performs better in Asian than in European patients with AF. (Clinical Survey on the Stroke Prevention in Atrial Fibrillation in Asia [AF-Registry]; NCT04807049)
Key Words: Asians, atrial fibrillation, bleeding, mortality, thromboembolism
Central Illustration
Atrial fibrillation (AF) is the most common arrhythmia worldwide and is associated with an increased risk of systemic embolism and death.1 Oral anticoagulation (OAC) with vitamin K antagonists (VKAs) or non-vitamin K anticoagulants (NOACs), has been shown to reduce thromboembolic events (TEs) and to improve survival in patients with AF; however, the use of OAC has to balance reduction of stroke against the potential risk of bleeding events.2
Several risk-stratification scores have been proposed to stratify the thrombotic and hemorrhagic risk in patients with AF; the most commonly used clinical practices are the CHA2DS2-VASC and HAS-BLED scores.3,4 Despite these scores having good predictive value and their recommendation by international guidelines,5, 6, 7 they have been derived from studies mainly performed in Western countries, and their application in Asian populations has been debated. Indeed, compared with Western patients, Asian patients with AF have a higher predisposition to bleeding and a different TE risk profile, characterized by a higher prevalence of heart failure (HF) and diabetes and a lower prevalence of vascular disease.8, 9, 10
Recently, 3 new predictive risk models to identify patients with AF at high risk of death for any causes, major bleeding, and TEs have been proposed from the COOL-AF (Cohort of Antithrombotic Use and Optimal International Normalized Ratio [INR] Level in Patients With Nonvalvular Atrial Fibrillation in Thailand) study.11 In these patients, the COOL-AF scores showed more improved performances than the CHA2DS2-VASc and HAS-BLED scores and have been proposed as possible Asian-specific risk scores for all-cause mortality, major bleeding, and TEs.11 These risk scores require external validation in other Asian and non-Asian cohorts.
The aim of this study was to validate the COOL-AF score externally in the APHRS (Asia-Pacific Heart Rhythm Society) registry and to compare its performance in the ESC-EHRA (European Society of Cardiology-European Heart Rhythm Association) EORP-AF (EURObservational Research Programme in Atrial Fibrillation) general long-term registry.
Methods
The study protocol for patient selection and data collection for the APHRS and EORP-AF were similar, as reported previously.12,13 In brief, the APHRS registry was started in 2015, and the enrollment finished in 2017. The population was composed of consecutive inpatients and outpatients with AF who had undergone cardiology examinations in tertiary and general hospitals in 5 Asian countries (Hong Kong, South Korea, Japan, Singapore, and Taiwan). All eligible patients had electrocardiogram (ECG)-documented AF within 12 months before their enrollment visits and had signed written informed consent forms according to the local regulations. After the baseline clinical assessment, the 1-year follow-up was performed by the local investigators. The study protocol was approved by local ethics committees, and the trial was registered on ClinicalTrials.gov (NCT04807049).
Study outcomes
Adverse outcomes were registered after 1 year of follow-up observation. The primary endpoints of the study were all-cause mortality, major bleedings (including intracranial hemorrhage and extracranial major bleedings) and TEs (including stroke, transient ischemic attack [TIA], and any peripheral embolism).
Risk scores
The CHA2DS2-VASc score was calculated as follows: congestive HF (1 point); hypertension (1 point); age 65 to 74 (1 point) and >75 years (2 points); diabetes (1 point); stroke (2 points); vascular disease (1 point); and female sex category (1 point). Patients with CHA2DS2-VASc ≥2 were considered at high-risk for TEs.5 The HAS-BLED score was calculated as follows: uncontrolled hypertension (1 point), abnormal renal, or liver function (1 point); history of stroke (1 point); history of bleeding (1 point); labile INR (1 point); age >65 years (1 point); and drugs (eg, aspirin or nonsteroidal anti-inflammatory drugs or alcohol) (1 point). Patients with HAS-BLED ≥3 were considered at high risk for bleeding.5 In APHRS, the HAS-BLED score was calculated by the investigators (including the labile INR criterion, when applicable) and reported in the case records.
The COOL-AF scores were calculated as was previously reported14:
-
•
COOL-AF score for all-cause mortality at 1-year was calculated as follows: 1 − 0.94712516exp (Prognostic Index) when prognostic index = 0.020319 · age − 0.087589 · body mass index (BMI) − 0.456114 · paroxysmal AF + 0.518448 · bleeding + 0.805105 · anemia.
-
•
COOL-AF score for major bleeding at 1 year was calculated as follows: 1 − 0.99950939 exp (Prognostic Index) when prognostic index = 0.042377 · age − 0.512419 · female + 1.679421 · renal replacement therapy + 0.601297 · anemia + 0.849478 · OAC.
-
•
COOL-AF score for TE 1 year was calculated as follows: 1 − 0.99501052 exp (Prognostic Index) when prognostic index = 0.841293 · hypertension + 1.326225 · chronic kidney disease (CKD) − 0.716626 · OAC.
The COOL-AF score components were defined according to the COOL-AF original study as follows: Anemia was defined according to the World Heart Organization criteria as a hemoglobin level <13 g/dL for male subjects and <12 g/dL for female subjects15; CKD was defined as an estimated glomerular function <60 mL/min/1.73 m2, according to the Kidney Disease Improving Global Outcomes (KIDGO) guidelines.16 Previous bleeding was defined as the occurrence of major bleeding or clinically relevant non-major bleeding according to the International Society on Thrombosis and Haemostasis (ISTH) guidelines.17,18 Paroxysmal AF and hypertension diagnosis were determined according to the ESC recommendations.5,19
Statistical analyses
Continuous variables are reported as median(IQR), whereas categorial variables are reported as percentages. Comparison among groups has been done with a nonparametric test (Mann-Whitney test) and chi-square test. Receiver operating characteristic (ROC) curves were used to assess the ability of the COOL-AF scores to predict the primary endpoints. Comparisons of the predictive ability of COOL-AF scores with CHA2DS2-VASc for all-cause of mortality and TEs and HAS-BLED for major bleeding were performed in each population by ROC pairwise comparison. Area under the curve (AUC) values were calculated using the method described by Delong et al.14 In addition, we estimated the clinical usefulness and net benefit of COOL-AF scores and CHA2DS2-VASc or HAS-BLED scores using the decision curve analysis (DCA) with the method proposed by Vickers et al.20
In each population, we used the ROC curve with Youden’s J statistic (J index) to find the specific optimal cutoff to dichotomize the COOL-AF scores.
J Index was calculated as follows: sensitivity/specificity-1 = (true positives/true positives + false negatives) + (true negatives/true negatives + false positives)-1
Plots of Kaplan-Meier curves for time to all-cause mortality, major bleeding, and any TE according to the dichotomized COOL-AF scores were performed. Survival distributions were compared using the log-rank test. Cox proportional hazards regression time to the first event analysis was used to calculate adjusted relative HRs and 95% CI of outcomes. All the multivariable Cox regression analyses, performed to investigate the association between COOL-AF scores and the primary outcomes, were adjusted for the following covariates: age, sex, and CHA2DS2-VASc ≥2 or HAS-BLED ≥3. Only a 2-sided P value <0.05 was considered statistically significant. Patients without available data to calculate the COOL-AF scores or follow-up were excluded from the analysis. All statistics were performed by SPSS statistical software, version 25.0 (IBM SPSS Statistics), and MedCalc (MedCalc Software Ltd).
Results
Of the 4,666 and 11,096 patients enrolled in the APHRS and EORP-AF, the total number of patients with all the data needed to calculate COOL-AF scores and available 1-year follow-up was 3,628 (77.7%) and 8,825 (79.5%), respectively. APHRS patients not included in this analysis were younger, with a higher prevalence of male sex, and a lower risk for TEs compared with included patients. EORP-AF patients not included were older, but no significative other differences were found for thrombotic and hemorrhagic baseline risks.
As reported in Table 1, EORP-AF patients were older; had a higher prevalence of female patients; and a higher median value of systolic and diastolic blood pressure, heart rate, and BMI. Regarding the clinical history, EORP-AF patients showed a higher prevalence of HF, coronary artery disease (CAD), stroke and TIA, dyslipidemia, chronic obstructive pulmonary disease, and CKD, whereas APHRS patients had a higher prevalence of paroxysmal AF, previous bleeding, diabetes, and anemia. EORP-AF patients were at higher risk of both thrombotic and hemorrhagic events, as showed by the higher prevalence of patients with CHA2DS2-VASc ≥2 and HAS-BLED ≥3. EORP-AF patients were more often prescribed with antithrombotic treatment and showed a higher use of both antiplatelet and OAC. The most used OACs were VKAs in EORP-AF and non-VKA oral anticoagulants (NOACs) in APHRS.
Table 1.
Baseline Characteristics of Study Populations
APHRS (n = 3,628) | EORP-AF (n = 8,825) | P Value | |
---|---|---|---|
Age (y) | 68.5 (61.0-76.0) | 71.0 (63.0-77.0) | <0.001 |
Female | 33.4 (1,210) | 40.1 (8,358) | <0.001 |
BMI (kg/m2) | 24.7 (22.3-27.3) | 27.5 (24.8-31.1) | <0.001 |
SBP (mm Hg) | 128 (117-140) | 130 (120-142) | <0.001 |
DBP (mm Hg) | 74 (66-82) | 80 (70-87) | <0.001 |
HR (beats/min) | 75 (66-85) | 78 (66-92) | <0.001 |
Paroxysmal AF | 42.0 (1,520) | 26.2 (2,308) | <0.001 |
HF | 20.8 (749) | 38.3 (3,355) | <0.001 |
CAD | 19.5 (695) | 27.8 (2,339) | <0.001 |
Stroke/TIA | 9.8 (355) | 8.6 (754) | 0.026 |
Bleeding | 7.5 (271) | 5.4 (476) | <0.001 |
Diabetes | 24.8 (891) | 22.8 (2,007) | 0.021 |
Hypertension | 61.4 (2,218) | 61.8 (5,457) | 0.668 |
Smoking | 8.8 (320) | 9.4 (778) | 0.326 |
Dyslipidemia | 39.1 (1405) | 41.9 (3,563) | 0.005 |
Dementia | 1.5 (54) | 1.1 (95) | 0.055 |
COPD | 2.6 (95) | 8.7 (759) | <0.001 |
CKD | 7.5 (271) | 12.3 (1,084) | <0.001 |
Dialysis | 1.2 (44) | 0.4 (37) | <0.001 |
Anemia | 6.6 (241) | 5.0 (437) | <0.001 |
CHA2DS2-VASc ≥2 | 72.6 (2,634) | 80.5 (7,102) | <0.001 |
HAS-BLED ≥3 | 14.2 (516) | 17.4 (1,532) | <0.001 |
Antiplatelet | 15.0 (543) | 18.3 (1,617) | <0.001 |
OAC | 3,016 (83.1) | 86.7 (7,654) | <0.001 |
VKA | 20.0 (725) | 50.5 (4,455) | <0.001 |
NOAC | 63.1 (2,291) | 36.3 (3,205) | <0.001 |
Beta-blocker | 51.6 (1,861) | 68.8 (2,751) | <0.001 |
CCB | 23.4 (848) | 16.9 (1,490) | <0.001 |
Statin | 37.8 (1,363) | 42.3 (3,730) | <0.001 |
ACEI/ARB | 39.5 (1,428) | 60.5 (5,326) | <0.001 |
Values are median (IQR) or % (n).
ACEI = ACE inhibitor; AF = atrial fibrillation; APHRS = Asian Pacific Heart Rhythm Society; ARB = angiotensin receptor blocker; BMI = body mass index; bpm = beats per minute; CAD = coronary artery disease; CCB = calcium channel blocker; CKD = chronic kidney disease; COPD = chronic obstructive pulmonary disease; DBP = diastolic blood pressure; EORP-AF = EURObservational Research Programme in Atrial Fibrillation; HF = heart failure; HR = heart rate; NOAC = non-vitamin K anticoagulant; OAC = oral anticoagulation; SBP = systolic blood pressure; TIA = transient ischemic attack; VKA = vitamin K antagonist.
Prediction of mortality, bleeding, and thromboembolism
In APHRS, the COOL-AF scores for all-cause mortality and major bleeding showed moderate-to-good predictive value (AUC: 0.77; 95% CI: 0.71-0.83; P < 0.001 and AUC: 0.68; 95% CI: 0.60-0.76; P < 0.001, respectively) (Figures 1A and 1C), and AUCs were nonstatistically significant different from those of CHA2DS2-VASc (AUC: 0.78; 95% CI: 0.74-0.82; P = 0.642) (Figure 1B) and HAS-BLED (AUC: 0.69; 95% CI: 0.61-0.76; P = 0.909) (Figure 1D) scores. The COOL-AF score for TEs showed a low predictive value (AUC: 0.61; 95% CI: 0.51-0.71; P = 0.031) (Figure 1E), with no statistically significant differences with CHA2DS2-VASc score (AUC: 0.68; 95% CI: 0.57-0.79; P = 0.228) (Figure 1F).
Figure 1.
Receiver Operating Curves for COOL-AF Scores
COOL-AF score for all-cause mortality (A) and its comparison with CHA2DS2-VASC (B); COOL-AF score for major bleeding (C) and its comparison with HAS-BLED (D); COOL-AF score for thromboembolism (E) and its comparison with CHA2DS2-VASc (F) in APHRS. COOL-AF score for all-cause mortality (G) and its comparison with CHA2DS2-VASC (H), COOL-AF score for major bleeding (I) and its comparison with HAS-BLED (J); COOL-AF score for thromboembolism (K) and its comparison with CHA2DS2-VASc (L) in EORP-AF. APHRS = Asian Pacific Heart Rhythm Society; COOL-AF = Cohort of Antithrombotic Use and Optimal International Normalized Ratio Levels in Patients With Atrial Fibrillation; EORP-AF = EURObservational Research Programme in Atrial Fibrillation.
In EORP-AF, the COOL-AF scores for all-cause mortality and major bleeding showed a moderate predictive value (AUC: 0.68; 95 % CI: 0.65-0.70; P < 0.001 and AUC: 0.61, 95% CI: 0.60-0.62; P = 0.001, respectively) (Figures 1G and 1I) with no statistically significant differences compared with CHA2DS2-VASc (AUC: 0.69; 95% CI: 0.66-0.71, 0.491; P = 491) (Figure 1H) and HAS-BLED scores (AUC: 0.61; 95% CI: 0.56-0.67; P = 0.802) (Figure 1J). COOL-AF for TEs was nonsignificant (AUC: 0.52; 95% CI: 0.47-0.58; P = 0.450) (Figure 1K) and inferior to CHA2DS2-VASc (AUC: 0.59; 95% CI: 0.54-0.65; P = 0.025) (Figure 1L).
For the specific optimal cutoff to dichotomize the COOL-AF scores, the J indexes identified in APHRS and EORP-AF for each COOL-AF score were, respectively, –0.024122609 and –0.0553404034 for COOL-AF all-cause mortality; 0.001869861 and 0.001995343 for COOL-AF major bleeding; and –0.003590973 and 0.000623386 for COOL-AF TEs. The prevalence of patients with COOL-AF score > J index in APHRS and EORP-AF was 25.4% (n = 922) and 45.9% (n = 4,049) for COOL-AF all-cause mortality; 30.7% (n = 1,112) and 17.2% (n = 1,514) for COOL-AF major bleeding; and 70.5% (n = 2,558) and 65.3% (n = 5,762) for COOL-AF TEs.
Survival analysis
The median follow up was 365 (IQR: 343-373) days in APHRS and 367 (IQR: 357-383) days in EORP-AF. During the 1-year follow-up, in APHRS and EORP-AF the following events were recorded: 87 (2.4%) and 435 (4.9%) death for any causes, 37 (1.0%) and 111 (1.3%) major bleeding, and 25 (0.7%) and 109 (1.2%) TEs, respectively.
In APHRS patients with COOL-AF scores > J index, the 1-year incidence of all-cause mortality, major bleeding, and TEs was higher than patients ≤ J index (6.4% [n =59] vs 1.0% (n = 28), log-rank test <0.001; 2.1% (n = 23) vs 0.6% (n = 14), log-rank test <0.001; and 0.9% (n = 22) vs 0.6% (n = 3), log-rank test = 0.038, respectively) (Figures 2A, 2C, and 2E). In EORP-AF, in patients with COOL-AF all-cause mortality and major bleeding scores > J index, the 1-year incidence of all-cause mortality and major bleeding was higher than patients ≤ J index (7.3% [n = 297] vs 2.9% [n = 138], log-rank test <0.001; and 2.7% [n = 41] vs 1.0% [n = 70], log-rank test <0.001; respectively) (Figures 2B and 2D), whereas no significative difference was found for COOL-AF TEs (1.2% [n = 70] vs 1.3% [n = 39], log-rank test = 0.765) (Figure 2F).
Figure 2.
Multivariate Cox-Regression Analysis for COOL-AF Scores
COOL-AF all-cause mortality in APHRS (A) and EORP-AF (B), COOL-AF major bleeding in APHRS (C) and EORP-AF (D), COOL-AF thromboembolism in APHRS (E) and EORP-AF (F). Abbreviations as in Figure 1.
Cox regression models
The dichotomized COOL-AF scores were tested in different Cox regression models adjusted for age, sex, and CHA2DS2-VASc ≥2 or HAS-BLED≥3 (Table 2). COOL-AF score for all-cause mortality was significantly associated with all-cause mortality after adjustment for age, sex, and CHA2DS2-VASc ≥2 in both APHRS (HR: 4.35; 95% CI: 2.65-7.14) and EORP-AF (HR: 1.70; 95% CI: 1.36-2.14) (Table 2, Model A). COOL-AF major bleeding was significantly associated with major bleeding after adjustment for age, sex and HAS-BLED≥3 in both APHRS (HR: 3.41; 95% CI: 1.32-8.80) and EORP-AF (HR: 2.52; 95% CI: 1.56-3.46) (Table 2, Model B). COOL-AF TE was not associated with TEs after adjustment for age, sex, and CHA2DS2-VASc ≥2 in both populations (Table 2, Model C).
Table 2.
Predictive Models for COOL-AF Scores in APHRS and EORP-AF
APHRS |
EORP-AF |
|||||
---|---|---|---|---|---|---|
HR | 96% CI | P Value | HR | 96% CI | P Value | |
Model A | ||||||
Age (continuous) | 1.04 | 1.01-1.07 | 0.004 | 1.06 | 1.04-10.7 | <0.001 |
Female | 0.93 | 0.61-1.44 | 0.758 | 0.88 | 0.72-1.08 | 0.219 |
COOL-AF score all-cause mortality > J index | 4.35 | 2.65-7.14 | <0.001 | 1.70 | 1.36-2.14 | <0.001 |
CHA2DS2-VASc ≥2 | 3.50 | 1.18-12.06 | 0.047 | 1.95 | 1.16-3.64 | 0.011 |
Model B | ||||||
Age (continuous) | 1.02 | 0.97-1.06 | 0.461 | 1.00 | 0.98-1.03 | 0.704 |
Female | 1.49 | 0.69-3.20 | 0.310 | 1.23 | 0.80-1.90 | 0.350 |
COOL-AF score major bleeding > J index | 3.41 | 1.32-8.80 | 0.011 | 2.52 | 1.49-4.25 | 0.001 |
HAS-BLED ≥3 | 2.56 | 1.26-5.18 | 0.009 | 2.32 | 1.56-3.46 | <0.001 |
Model C | ||||||
Age (continuous) | 1.05 | 1.01-1.10 | 0.022 | 1.02 | 0.99-1.04 | 0.151 |
Female | 1.42 | 0.63-3.22 | 0.404 | 1.22 | 0.83-1.80 | 0.309 |
COOL-AF score thromboembolism > J index | 3.14 | 0.90-10.93 | 0.073 | 1.50 | 0.97-2.34 | 0.071 |
CHA2DS2-VASc ≥2 | 0.62 | 0.16-2.43 | 0.493 | 1.46 | 0.71-2.99 | 0.301 |
Abbreviations as in Table 1.
Discrimination analyses
We tested the clinical usefulness and net clinical benefit of the 3 COOL-AF scores using DCA (Supplemental Figures 1 and 2). The comparison between COOL-AF all-cause mortality and CHA2DS2-VASc showed marginal improvement in net clinical benefit in APHRS (but not EORP-AF), but the COOL-AF major bleeding and HAS-BLED and COOL-AF TE and CHA2DS2-VASc showed overlapping curves in both populations.
Discussion
In this study, we have performed the first external validation of the COOL-AF scores, testing their predictive value in a large prospective cohort of Asian patients with AF and comparing their performance with Europeans. We found that all the COOL-AF scores are significantly associated with the primary outcomes in Asian patients with AF at risk for death and major bleeding and seems to perform better in Asian patients with AF compared with Europeans (Central Illustration).
Central Illustration.
Cohort of Antithrombotic Use and Optimal International Normalized Ratio Levels in Patients With Atrial Fibrillation Scores Predictive Value in Asian Pacific Heart Rhythm Society and EURObservational Research Programme in Atrial Fibrillation
The COOL-AF scores for all-cause mortality and major bleeding had good predictive value in both the registries, whereas the COOL-AF score for thromboembolism had a modest predictive value only in APHRS. Overall, the COOL-AF scores generally performed better in Asian than in European patients with AF. APHRS = Asian Pacific Heart Rhythm Society; AUC = area under the curve; AF = atrial fibrillation; COOL-AF = Cohort of Antithrombotic Use and Optimal International Normalized Ratio Levels in Patients With Atrial Fibrillation.
The predictive value of COOL-AF score for all-cause mortality in APHRS (AUC: 0.77) was similar to that reported in the COOL-AF study (AUC: 0.73),11 had a moderately good performance also in EORP-AF (AUC: 0.66), and was independent and similar to the CHA2DS2-VASc score in both populations. Of note, the COOL-AF score for all-cause mortality is calculated with several factors not included in the CHA2DS2-VASc score, and these results take into account additional variables such as BMI, history of bleeding, anemia, and the type of AF, in addition to the traditional risk factors, leading to better death risk stratification in Asian patients with AF. This may be of importance in Asian patients in whom the association among BMI, body mass composition, and health risk is different from Europeans.21 Indeed, a recent study in Korean patients with AF has shown that each BMI increase of 5 kg/m2 was associated with lower risks of ischemic stroke (HR: 0.89; 95% CI: 0.80-0.99), major bleeding (HR: 0.79; 95% CI: 0.69-0.92), and all-cause mortality (HR: 0.66, 95% CI: 0.60-0.72).22 Furthermore, a very low BMI may be associated with alterations in OAC metabolism that could be responsible for bleeding episodes and anemia.23,24 Of note, the COOL-AF score for all-cause mortality include the AF pattern and consider paroxysmal AF as a factor associated with lower TE and mortality risk when compared with persistent and permanent AF.25
In our study, the incidence of major bleeding was higher in EORP-AF patients than in the APHRS cohort. These results, besides the higher baseline HAS-BLED score in EORP-AF, should be also related to the different type of OAC used in these 2 populations. The most used OAC treatment was VKAs in EORP-AF and NOACs in APHRS patients. Several studies showed that NOAC treatment compared with VKA treatment is associated with an absolute lower risk of bleeding and that the magnitude of this reduction was more pronounced in Asian compared with non-Asian patients.26 Even in patients only treated with NOACs, the risk of intracranial bleeding is still numerically higher in Asians than Western patients,27, 28, 29, 30 showing that a specific Asian risk score for bleeding is still needed. The predictive value of COOL-AF score for major bleeding in APHRS (AUC: 0.68) was similar to that reported in the COOL-AF study (AUC: 0.71),11 but had a lower predictive role in EORP-AF (AUC: 0.61) and was independent and similar to the HAS-BLED risk score in both the populations.
These results suggest the importance of detecting anemia in patients with AF to stratify the risk of all-cause mortality and major bleeding. This point is in accordance with 2 previous studies in 4,824 Chinese and 1,562 Thai patients with AF in which anemia was an independent risk factor for major bleeding, HF, and death31,32 but is in contrast to a recent study on 15,606 Chinese patients in which anemia was independently associated with all-cause death, cardiovascular death, but not with major bleeding.33 The conflicting results from studies investigating the associations between anemia and major bleeding in AF are probably caused by several factors: In Asia, the proportion of patients on OAC differs considerately in the different areas influencing the hemorrhagic risk; no information about the type of anemia was reported making impossible to understand its clinical weight; and no study was designed to clarify if the lower hemoglobin is a result rather than a cause of bleeding.
The COOL-AF score for major bleeding considers female sex as a possible protective factor for hemorrhagic events and could represent another possible explanation to its independency from HAS-BLED. The protective value of female sex for major bleeding in the COOL-AF study is supported by growing evidence that female sex is a Janus-faced factor associated with a higher risk of systemic embolism and lower risk of bleeding than male patients.3,34,35
In our study, the predictive value of COOL-AF score for TEs was significantly lower (AUC: 0.61) than that reported in the COOL-AF study (AUC: 0.70). In APHRS, COOL-AF for TEs showed a low predictive value and was similar to CHA2DS2-VASc, whereas in EORP-AF, it showed no association with TE, and its performance was inferior to CHA2DS2-VASc. When interpretating this result, some possible confounding factors should be considered. In the COOL-AF score for TE, 1 of the strongest risk factors is represented by CKD. In the APHRS registry, we found a 7-fold lower prevalence of CKD (7.5% vs 51.6%) and a significantly lower 1-year incidence of TEs (1.4 of 100 patients per year vs 0.7 of 100 patients per year) than the COOL-AF study, which could be associated with a loss of statistical power in the current study. The lack of any predictive value of COOL-AF score for TE in EORP-AF seems to confirm its low performance, underlying that the determinants of the thrombotic risk in Europeans may be different from that seen in Asians.
To date, although growing evidence showed the presence of ethnic-specific factors in determining the risk of death, bleeding, and thromboembolic events in patients with AF, the international guidelines have not still adopted or proposed any approach that considers these aspects.5, 6, 7 The APHRS guidelines for management of AF in Asians underline the importance of preferring NOACs to VKAs for prevention of stroke because of the higher predisposition to bleeding in those patients, but do not provide any clear indication regarding the possibility to consider ethnic-specific factors to characterize the risk of adverse events.6 Although the CHA2DS2-VASc and HAS-BLED risk scores represent the best predictive tools to identify high-risk patients, the management of patients classified as low-risk is still debated.36 In these patients, a regular risk (re)assessment has been proposed to detect the onset of incident cardiovascular risk factors early that can drive therapeutic decisions.37 In this context, adding to the information given by the classical risk scores, such as those derived from the COOL-AF scores, could potentially help the clinician to better stratify the risk of adverse events in Asian patients with AF. Furthermore, in the era of a more holistic or integrated care approach to management of AF, the spread of this concept to the ethnic-associated factors could lead to a further improvement of the short- and long-term outcomes.
Study limitations
The observational nature limits the strength of the evidence derived from this study. The presence of baseline differences between Asian and European patients could have influenced the predictive performance of the COOL-AF scores. Furthermore, given the high degree of heterogeneity within the different Asian (and European) geographic areas considered in this study, we cannot exclude the effect of ethnic and geographic factors in the performance of the COOL-AF scores. However, the main clinical and demographic characteristics of our APHRS population are similar to those reported in other Asian-based AF cohorts from China38 and Japan,39 allowing us to consider it as a representative Asian cohort of patients with AF.
Given the relatively low sample size of the study, when stratifying results by countries that participated in the APHRS registry, we were unable to analyze whether the performance of the COOL-AF scores differ among recruiting countries as well as to implement newer techniques (such as machine-learning algorithms), which may improve our ability to predict prognosis and major outcomes; further studies with larger sample size are therefore needed to answer these open questions. The low mortality rate observed in the APHRS registry is unlikely to affect the risk of competitive events, whereas the 1-year risk of outcomes was assessed by Cox proportional hazards regression time to the first event analysis, and no competing risk models for multiple events were used.
Finally, some of the variable needed for COOL-AF scores, such as anemia and CKD, are dynamic; as the data were collected at baseline, we cannot exclude that some patients with mild anemia or CKD ameliorate their status or that some patients with barely normal hemoglobin or creatinine values got worse during the observation period. Although we considered approximately the 80% of the initial cohort in both registries, the clinical differences between patients included and excluded from this analysis could have introduced some selection bias.
Conclusions
The COOL-AF risk scores may be an easy tool to identify Asian patients with AF at risk for all-cause mortality and major bleeding and seems to perform better in Asian patients than in European patients with AF.
Perspectives.
COMPETENCY IN PATIENTS CARE AND PROCEDURAL SKILLS: The COOL-AF scores have been proposed as Asian predictive models for patients with AF. The different predictive value in Asians and Europeans suggest that the risk of death, major bleeding, and thromboembolic events is influenced by ethnic-specific risk factors.
TRANSLATIONAL OUTLOOK: Further studies are needed to better characterize the specific weight of each cardiovascular risk factor in determining the death, bleeding, and thrombotic-risk profile in different ethnic groups.
Funding Support and Author Disclosures
Dr Romiti has done consultancy for Boehringer Ingelheim and has received an educational grant from Anthos, outside the submitted work; no fees are directly received personally. Dr Shimizu has received grants from Daiichi Sankyo Co, Ltd and Nippon Boehringer Ingelheim Co, Ltd; and remuneration for lectures, presentations, Speakers Bureau, manuscript writing, or educational events from Daiichi Sankyo Co, Ltd, Nippon Boehringer Ingelheim Co, Ltd, Bristol-Meyers Squibb, KK; Bayer Yakuhin, Ltd, Pfizer Japan, Inc, Ono Pharmaceutical Co Ltd, and Medtronic Japan Co, Ltd. Dr Boriani has received small speaker fees from Boston Scientific, Bayer, Boehringer, Bristol Myers Squibb, Janssen, and Sanofi, outside the submitted work. Dr Lip is a consultant and speaker for BMS/Pfizer, Boehringer Ingelheim, Anthos, and Daiichi Sankyo Co, Ltd; no fees were received personally. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.
Footnotes
Minglong Chen, MD, served as Guest Associate Editor for this paper. Nathan Wong, PhD, served as Guest Editor-in-Chief for this paper.
The authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’ institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information, visit the Author Center.
Appendix
For supplemental figures, please see the online version of this paper.
Contributor Information
Tommaso Bucci, Email: tommaso.bucci@liverpool.ac.uk.
Gregory Y.H. Lip, Email: gregory.lip@liverpool.ac.uk.
Appendix
References
- 1.Chugh S.S., Havmoeller R., Narayanan K., et al. Worldwide epidemiology of atrial fibrillation: a Global Burden of Disease 2010 Study. Circulation. 2014;129:837–847. doi: 10.1161/CIRCULATIONAHA.113.005119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lane D.A., Lip G.Y.H. Stroke and bleeding risk stratification in atrial fibrillation: a critical appraisal. Eur Heart J. 2020;22(Suppl):O14–O27. doi: 10.1093/eurheartj/suaa178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lip G.Y., Nieuwlaat R., Pisters R., Lane D.A., Crijns H.J. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach: the Euro Heart Survey on Atrial Fibrillation. Chest. 2010;137:263–272. doi: 10.1378/chest.09-1584. [DOI] [PubMed] [Google Scholar]
- 4.Pisters R., Lane D.A., Nieuwlaat R., de Vos C.B., Crijns H.J., Lip G.Y. A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest. 2010;138:1093–1100. doi: 10.1378/chest.10-0134. [DOI] [PubMed] [Google Scholar]
- 5.Hindricks G., Potpara T., Dagres N., 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. doi: 10.1093/eurheartj/ehaa612. [DOI] [PubMed] [Google Scholar]
- 6.Chao T.F., Joung B., Takahashi Y., et al. 2021 Focused Update Consensus Guidelines of the Asia Pacific Heart Rhythm Society on Stroke Prevention in Atrial Fibrillation: Executive Summary. Thromb Haemost. 2022;122:20–47. doi: 10.1055/s-0041-1739411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.January C.T., Wann L.S., Calkins H., 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. J Am Coll Cardiol. 2019;74(1):104–132. doi: 10.1016/j.jacc.2019.01.011. [DOI] [PubMed] [Google Scholar]
- 8.Krittayaphong R., Winijkul A., Methavigul K., et al. Risk profiles and pattern of antithrombotic use in patients with non-valvular atrial fibrillation in Thailand: a multicenter study. BMC Cardiovasc Disord. 2018;18:174. doi: 10.1186/s12872-018-0911-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kim S.M., Jeon E.T., Jung J.M., Lee J.S. Real-world oral anticoagulants for Asian patients with non-valvular atrial fibrillation: a PRISMA-compliant article. Medicine (Baltimore) 2021;100 doi: 10.1097/MD.0000000000026883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gorog D.A., Gue Y.X., Chao T.F., et al. Assessment and mitigation of bleeding risk in atrial fibrillation and venous thromboembolism: a position paper from the ESC Working Group on Thrombosis, in collaboration with the European Heart Rhythm Association, the Association for Acute CardioVascular Care and the Asia-Pacific Heart Rhythm Society. Europace. 2022;24(11):1844–1871. doi: 10.1093/europace/euac020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Krittayaphong R., Kanjanarutjawiwat W., Wisaratapong T., Lip G.Y.H. Improving risk prediction for death, stroke and bleeding in Asian patients with atrial fibrillation. Eur J Clin Invest. 2022 doi: 10.1111/eci.13886. [DOI] [PubMed] [Google Scholar]
- 12.Tse H.F., Teo W.S., Siu C.W., et al. Prognosis and treatment of atrial fibrillation in Asian cities: 1-year review of the Asia-Pacific Heart Rhythm Society Atrial Fibrillation Registry. Europace. 2022;24(12):1889–1898. doi: 10.1093/europace/euab327. [DOI] [PubMed] [Google Scholar]
- 13.Bucci T., Proietti M., Shantsila A., et al. Integrated care for atrial fibrillation using the ABC pathway in the prospective APHRS-AF registry. JACC: Asia. 2023;3(4):580–591. doi: 10.1016/j.jacasi.2023.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.DeLong E.R., DeLong D.M., Clarke-Pearson D.L. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837–845. [PubMed] [Google Scholar]
- 15.Garcia-Casal M.N., Pasricha S.R., Sharma A.J., Pena-Rosas J.P. Use and interpretation of hemoglobin concentrations for assessing anemia status in individuals and populations: results from a WHO technical meeting. Ann N Y Acad Sci. 2019;1450:5–14. doi: 10.1111/nyas.14090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Stevens P.E., Levin A., Kidney Disease Improving Global Outcomes Chronic Kidney Disease Guideline Development Work Group Members. Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med. 2013;158:825–830. doi: 10.7326/0003-4819-158-11-201306040-00007. [DOI] [PubMed] [Google Scholar]
- 17.Schulman S., Kearon C. Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost. 2005;3:692–694. doi: 10.1111/j.1538-7836.2005.01204.x. [DOI] [PubMed] [Google Scholar]
- 18.Kaatz S., Ahmad D., Spyropoulos A.C., Schulman S., Subcommittee on Control of Anticoagulation Definition of clinically relevant non-major bleeding in studies of anticoagulants in atrial fibrillation and venous thromboembolic disease in non-surgical patients: communication from the SSC of the ISTH. J Thromb Haemost. 2015;13:2119–2126. doi: 10.1111/jth.13140. [DOI] [PubMed] [Google Scholar]
- 19.Williams B., Mancia G., Spiering W., et al. 2018 practice guidelines for the management of arterial hypertension of the European Society of Hypertension and the European Society of Cardiology: ESH/ESC task force for the management of arterial hypertension. J Hypertens. 2018;36:2284–2309. doi: 10.1097/HJH.0000000000001961. [DOI] [PubMed] [Google Scholar]
- 20.Vickers A.J., Cronin A.M., Elkin E.B., Gonen M. Extensions to decision curve analysis, a novel method for evaluating diagnostic tests, prediction models and molecular markers. BMC Med Inform Decis Mak. 2008;8:53. doi: 10.1186/1472-6947-8-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.WHO Expert Consultation Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet. 2004;363:157–163. doi: 10.1016/S0140-6736(03)15268-3. [DOI] [PubMed] [Google Scholar]
- 22.Lee S.R., Choi E.K., Jung J.H., et al. Body mass index and clinical outcomes in Asian patients with atrial fibrillation receiving oral anticoagulation. Stroke. 2021;52:521–530. doi: 10.1161/STROKEAHA.120.030356. [DOI] [PubMed] [Google Scholar]
- 23.Krittayaphong R., Chichareon P., Komoltri C., Kornbongkotmas S., Yindeengam A., Lip G.Y.H. Low body weight increases the risk of ischemic stroke and major bleeding in atrial fibrillation: the COOL-AF registry. J Clin Med. 2020;9:2713. doi: 10.3390/jcm9092713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Upreti V.V., Wang J., Barrett Y.C., et al. Effect of extremes of body weight on the pharmacokinetics, pharmacodynamics, safety and tolerability of apixaban in healthy subjects. Br J Clin Pharmacol. 2013;76:908–916. doi: 10.1111/bcp.12114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ganesan A.N., Chew D.P., Hartshorne T., et al. The impact of atrial fibrillation type on the risk of thromboembolism, mortality, and bleeding: a systematic review and meta-analysis. Eur Heart J. 2016;37:1591–1602. doi: 10.1093/eurheartj/ehw007. [DOI] [PubMed] [Google Scholar]
- 26.Wang K.L., Lip G.Y., Lin S.J., Chiang C.E. Non-vitamin K antagonist oral anticoagulants for stroke prevention in Asian patients with nonvalvular atrial fibrillation: meta-analysis. Stroke. 2015;46:2555–2561. doi: 10.1161/STROKEAHA.115.009947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hori M., Connolly S.J., Zhu J., et al. Dabigatran versus warfarin: effects on ischemic and hemorrhagic strokes and bleeding in Asians and non-Asians with atrial fibrillation. Stroke. 2013;44:1891–1896. doi: 10.1161/STROKEAHA.113.000990. [DOI] [PubMed] [Google Scholar]
- 28.Wong K.S., Hu D.Y., Oomman A., et al. Rivaroxaban for stroke prevention in East Asian patients from the ROCKET AF trial. Stroke. 2014;45:1739–1747. doi: 10.1161/STROKEAHA.113.002968. [DOI] [PubMed] [Google Scholar]
- 29.Goto S., Zhu J., Liu L., et al. Efficacy and safety of apixaban compared with warfarin for stroke prevention in patients with atrial fibrillation from East Asia: a subanalysis of the Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation (ARISTOTLE) trial. Am Heart J. 2014;168:303–309. doi: 10.1016/j.ahj.2014.06.005. [DOI] [PubMed] [Google Scholar]
- 30.Yamashita T., Koretsune Y., Yang Y., et al. Edoxaban vs. warfarin in East Asian patients with atrial fibrillation: an ENGAGE AF-TIMI 48 subanalysis. Circ J. 2016;80:860–869. doi: 10.1253/circj.CJ-15-1082. [DOI] [PubMed] [Google Scholar]
- 31.Guo Y.T., Zhang Y., Shi X.M., et al. Assessing bleeding risk in 4824 Asian patients with atrial fibrillation: the Beijing PLA Hospital Atrial Fibrillation Project. Sci Rep. 2016;6 doi: 10.1038/srep31755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Krittayaphong R., Pumprueg S., Thongsri T., et al. Impact of anemia on clinical outcomes of patients with atrial fibrillation: the COOL-AF registry. Clin Cardiol. 2021;44:415–423. doi: 10.1002/clc.23559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang Z., Jiang C., He L., et al. Associations of anemia with death and major bleeding in patients with atrial fibrillation: a report from the Chinese Atrial Fibrillation Registry Study. Clin Cardiol. 2022;45:91–100. doi: 10.1002/clc.23764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yong C.M., Tremmel J.A., Lansberg M.G., Fan J., Askari M., Turakhia M.P. Sex differences in oral anticoagulation and outcomes of stroke and intracranial bleeding in newly diagnosed atrial fibrillation. J Am Heart Assoc. 2020;9 doi: 10.1161/JAHA.120.015689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Law S.W.Y., Lau W.C.Y., Wong I.C.K., et al. Sex-based differences in outcomes of oral anticoagulation in patients with atrial fibrillation. J Am Coll Cardiol. 2018;72:271–282. doi: 10.1016/j.jacc.2018.04.066. [DOI] [PubMed] [Google Scholar]
- 36.Sulzgruber P., Wassmann S., Semb A.G., et al. Oral anticoagulation in patients with non-valvular atrial fibrillation and a CHA2DS2-VASc score of 1: a current opinion of the European Society of Cardiology Working Group on Cardiovascular Pharmacotherapy and European Society of Cardiology Council on Stroke. Eur Heart J Cardiovasc Pharmacother. 2019;5:171–180. doi: 10.1093/ehjcvp/pvz016. [DOI] [PubMed] [Google Scholar]
- 37.Chang T.Y., Lip G.Y.H., Chen S.A., Chao T.F. Importance of risk reassessment in patients with atrial fibrillation in guidelines: assessing risk as a dynamic process. Can J Cardiol. 2019;35:611–618. doi: 10.1016/j.cjca.2019.01.018. [DOI] [PubMed] [Google Scholar]
- 38.Wang Y.F., Jiang C., He L., et al. Integrated care of atrial fibrillation using the ABC (Atrial fibrillation Better Care) pathway improves clinical outcomes in Chinese population: an analysis from the Chinese Atrial Fibrillation Registry. Front Cardiovasc Med. 2021;8 doi: 10.3389/fcvm.2021.762245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sakuma I., Uchiyama S., Atarashi H., et al. Clinical risk factors of stroke and major bleeding in patients with non-valvular atrial fibrillation under rivaroxaban: the EXPAND study sub-analysis. Heart Vessels. 2019;34:1839–1851. doi: 10.1007/s00380-019-01425-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.