Abstract
The prevalence and economic burden of atrial fibrillation (AF) are predicted to more than double over the next few decades. In addition to anticoagulation and treatment of concomitant cardiovascular conditions, early and standardized rhythm control therapy reduces cardiovascular outcomes as compared with a rate control approach, favouring the restoration, and maintenance of sinus rhythm safely. Current therapies for rhythm control of AF include antiarrhythmic drugs (AADs) and catheter ablation (CA). However, response in an individual patient is highly variable with some remaining free of AF for long periods on antiarrhythmic therapy, while others require repeat AF ablation within weeks. The limited success of rhythm control therapy for AF is in part related to incomplete understanding of the pathophysiological mechanisms and our inability to predict responses in individual patients. Thus, a major knowledge gap is predicting which patients with AF are likely to respond to rhythm control approach. Over the last decade, tremendous progress has been made in defining the genetic architecture of AF with the identification of rare mutations in cardiac ion channels, signalling molecules, and myocardial structural proteins associated with familial (early-onset) AF. Conversely, genome-wide association studies have identified common variants at over 100 genetic loci and the development of polygenic risk scores has identified high-risk individuals. Although retrospective studies suggest that response to AADs and CA is modulated in part by common genetic variation, the development of a comprehensive clinical and genetic risk score may enable the translation of genetic data to the bedside care of AF patients. Given the economic impact of the AF epidemic, even small changes in therapeutic efficacy may lead to substantial improvements for patients and health care systems.
Keywords: Atrial fibrillation, Antiarrhythmic drugs, Catheter ablation, Response, Arrhythmias, Genetic variation, Genotype
Graphical Abstract
1. Introduction
More than 30 million people around the world were affected by atrial fibrillation (AF) in 2010.1 The lifetime risk of developing AF in people of European descent older than 40 years one in three and AF is associated with significant morbidity and increased mortality.2 Even on optimal current therapy, approximately half of this mortality is cardiovascular, including sudden death and heart failure.3 The aging of the population, accumulation of comorbidities associated with AF such as the hypertension, chronic kidney disease, obesity, and heart failure, and improved detection methods will fuel the growing AF epidemic.4 The prevalence of AF in the USA (5.2 million cases in 2010) is projected to increase to over 12 million cases by 2030.5 Likewise, in whites of European descent the prevalence will rise from 7.6 million cases in 2016 to over 14 million in 2060.6
Besides treating the underlying conditions to prevent recurrent AF and reducing AF-related complications, oral anticoagulation can prevent most strokes in patients and rate control therapy reduces AF-associated symptoms. In fact, most patients with AF are managed with these treatment modalities.7 However, even on anticoagulation and rate control therapy, and with good management of comorbidities, AF-related complications remain high with 5% annual risk of cardiovascular death, stroke, or hospitalization for worsening of heart failure or acute coronary syndrome.3
The current ‘one-size fits all’ approach to selecting rhythm control therapy especially antiarrhythmic drugs (AADs) is based largely on minimizing the risk of adverse effects rather than on therapeutic efficacy.8 Possible reasons for the high variability in response to AAD therapy include heterogeneity of the underlying electrical and structural substrate, limited understanding of the underlying pathophysiological mechanisms, and our inability to predict individual responses.9 The genomic variability underlying AF also contributes to the variable response, as suggested by recent translational hypothesis-generating studies.10 Thus, a major knowledge gap is predicting which patients with AF are most likely to respond to rhythm control therapy. The heritability of AF has been reported as ∼22% in the UK Biobank and as high as ∼62% in a study of Danish twins.11,12
Following the first description of familial AF over seven decades ago,13 and almost 15 years after the first publication of a genetic locus associated with AF,14 great progress has been made in understanding the genetic architecture of the arrhythmia. Advances in next-generation sequencing have enabled comprehensive genotyping at reasonable cost. Although genetic approaches to AF have revealed that susceptibility to AF development and response to therapy is modulated in part by the underlying genetic substrate, the impact of these discoveries on the bedside care of patients has been limited. In this monograph, we review whether the increased knowledge on common genetic variation associated with AF can improve our management of the arrhythmia and how and when this may be best accomplished.
2. Genetic basis of AF
In 1936, Wolff13 described a family of three brothers with AF in one of the first reports of a hereditary form of AF. The first discovered gene mutation associated with familial AF was KCNQ1 coding for the Kv7.1 potassium channel that mediates the slow rectifying potassium current (IKs).15 Other ion channel genes implicated in AF include SCN5A, the gene coding the cardiac sodium channel. For instance, in a Japanese family with autosomal dominant familial AF, a novel gain-of-function mutation in SCN5A (M1875T) was identified.16 Mutations in SCN5A have been identified in ∼5% of unselected AF population, with a higher prevalence in younger cohorts.17 Single-nucleotide polymorphisms (SNPs) encoding sodium channels (SCN family) have also been associated with AF.18 Interestingly, a loss-of-function (LOF) mutation in the potassium voltage-gated channel, shaker-related subfamily member 5 (KCNA5) modulated tyrosine kinase regulation which suggests more subtle disruptions to cardiac electrophysiology than just dysfunctional ion channels.19
The first and only non-ion channel gene (NPPA) linked with familial AF was identified in a large family of 11 affected members.20 This frameshift mutation produced a mutant peptide that shortened the action potential duration in an isolated rat heart model and predisposed to re-entrant AF. Importantly, this was the first report that established a link between a non-ion channel mutation and altered atrial electrophysiology. Recently, next-generation sequencing identified two novel variants in myosin heavy chain 6 (MYH6) and myosin light chain 4 (MYL4) that are associated with familial AF emphasizing the important role of myocardial structural proteins in the pathogenesis of AF.21,22 Mutations of MYL4 associated with AF and structural cardiac myopathy have been later replicated in humans and animal models including zebrafish and rats.23,24 Of the genes implicated in AF, the strongest level of evidence exists for MYH6,21 MYL4,22,23,25 titin (TTN),26–28 and NPPA.20
Although research into rare variants in affected families has provided important insights into the underlying genetic mechanisms in isolated families, the attributable risk at the population is modest at best.29 Traditional linkage analysis and candidate gene approaches are complemented by genome-wide association studies (GWAS). Common variants associated with AF susceptibility have a weaker effect in the individual, but their population-attributable risk is considerable and may enable a genotype-based approach in large clinical trials. The reporting of novel variants in PLEC and TTN associated with early-onset AF have emphasized the importance of genes encoding for structural proteins in AF pathogenesis. In a GWAS of an Icelandic cohort, a PLEC missense variant, which is an important part of the cytoskeleton, was associated with an increased AF risk and cardioembolic stroke.30 The effect was, however, weaker compared with variants of MYH6.30 In a Danish study, titin-truncating variants (TTNtv) were associated with familial and early-onset AF with compelling evidence.26,31,32 Remarkably, 16% of probands with familial AF and 4.7% of patients with early-onset AF carried TTNtv. In zebrafish, these variants were associated with a sarcomere defect and atrial fibrosis.31 Titin is the largest protein in humans and central to sarcomere integrity by connecting the Z- and M-line. In a case–control study, TTN LOF variants were associated with early-onset AF.26 Around 2.1% of AF patients carried TTN LOF variants compared with 1.1% in control subjects. The prevalence of TTN LOF increased with younger age with 6.5% in early-onset AF patients under 30 with the findings replicated in the UK Biobank and the MyCode Geisinger cohort.
Testing for TTNtv is increasing in patients with heart failure because of robust data linking these variants with dilated cardiomyopathy (DCM).33,34 Patients with DCM and TTNtv have also been reported to be at higher risk for arrhythmias.35 Increased surveillance in patients with DCM and TTNtv has therefore been proposed to identify patients at risk for worsening heart failure.36 However, the benefit of TTNtv testing in AF patients without a history of heart failure is remains unclear. In a small case series of 25 patients with early-onset AF (<45 years, without significant comorbidities) referred to a tertiary care centre, 21 had at least one rare variant in a cardiomyopathy-associated gene (4 with TTN LOF and 1 with RMB20 which is implicated in titin splicing).27 Importantly, 8 out of 11 patients with structural left ventricular dysfunction by cardiac magnetic resonance imaging were missed by transthoracic echocardiography.27 Consequently, there may be a role for sequencing TTN in patients with early-onset AF who appear to have normal hearts but may be at risk for developing heart failure. Such an approach may include surveillance for peripartum cardiomyopathy or chemotherapy-induced cardiomyopathy, both of which have been associated with TTN variants.37–39 Yet, the process of structural or sarcomeric dysfunction in AF is not fully understood. An interesting zebrafish model of PITX2c LOF showed early sarcomeric changes in zebrafish embryos before extensive remodelling or electrophysiological defects were detectable.40 Metabolic changes with an increase of reactive oxygen species also contributed to sarcomeric changes and arrhythmia development.40 Further research will provide us with more insight into the mechanisms of genetic variation leading to AF and actionable findings. One example of that is the association of KCNN2 and KCNN3 variants with AF in several GWAS.41,42 Both genes encode proteins of a small conductance calcium-activated potassium channels (KCa2)43 that are targeted by a new class of AADs that are currently evaluated in an ongoing Phase 2 trial for conversion of AF to sinus rhythm (NCT04571385). Thus, variants in ion channels, signalling molecules, and myocardial structural proteins have been implicated in AF. Importantly, current antiarrhythmic therapy blocks cardiac ion channels only and a more personalized, i.e., patient-specific, approach needs to target the underlying substrate for AF including signalling molecules and myocardial structural proteins.
Thus far, the strongest data exist for variants on chromosome (chr) 4q25 adjacent to PITX2 which is known for its important role in pulmonary vein development and cardiogenesis.44,45 In 2007, Gudbjartsson et al.14 reported that a common variant on chr 4q25 in 35% of a European cohort and in 75% of a Chinese cohort increased the risk of AF by up to 1.72 per copy. Data from human and mouse models indicate high expression of PITX2 in adults and susceptibility to AF by action potential shortening.46,47 Consistently the chr4q25 locus has been replicated in European cohorts and Asian cohorts48 and new AF loci have been identified on chr16q22 (ZFHX3) and chr1q21 (KCNN3).41,49 Some of the known loci such as chr4q25 were replicated while others such as chr1q21 did not reach statistical significance.50,51 These findings suggest that AF-associated SNPs increase risk of AF and that at least some are likely ethnicity-specific.
Recently, large GWAS studies have reported new loci for common variants with increased AF risk. For instance, a GWAS for AF with ∼1 million subjects reported 163 independent risk variants at 111 genetic loci and 165 candidate AF genes.52 These loci include genes that code for structural functionality, embryogenetic cardiac development, intracellular calcium homeostasis, hormone signalling, and ion channels.52 However, the strongest association remains with SNPs located on chr4q25 across European, Japanese, and African-American populations.53 These findings further emphasize the multifactorial aetiology of AF and highlight the diagnostic difficulty to identify vulnerable populations.
2.1. Genetic risk scores
Although genome-wide polygenic risk scores (GPS) are calculated based on a multi-array of variants with the goal of identifying a population at high risk for developing AF, a number of preconditions have to be met. First, the GWAS must be large enough to identify all common variants associated with AF. Second, there must be sufficient power to replicate the GPS in the validation dataset. An early report used 12 variants on chr4q25 to identify patients with a five-fold increased AF risk; an effect observed in European whites and Japanese patients. Lubitz et al.54 showed that a comprehensive AF GPS was associated with incident AF exceeding associations of clinical risk factors emphasizing the complementary information provided by clinical and genetic factors. A follow-up study using a GPS with up to 719 SNPs reported that persons in the highest quartile were at 67% increased risk of developing AF.55 While the authors reported minimal benefit in addition to clinical risk factors, a GPS consisting of 127 variants identified patients with a two-fold increase of odds of cardioembolic stroke. This study illustrated the potential role of GPS for identifying AF populations with suspected cardioembolic stroke. The Stroke Genetics Network showed that a GPS with 934 variants was associated with cardioembolic stroke and stroke of undetermined source but not with large artery atherosclerosis or small artery occlusion.56 A GPS with 32 variants in over 50 000 patients from cardiovascular trials showed a four-fold increase in incident stroke in patients with high genetic risk but ‘low-risk’ CHA2DS2-VASc of 2.57 When examining the lifetime risk for AF in patients over 55 years, a GPS with 1000 variants was able to identify patients at high AF risk but low clinical risk.58
Recently, an AF GPS with 6.6 million variants in over 500 000 patients identified 6.1% of the general population at three-fold higher risk for AF.59 Identifying individuals at three-fold increased risk for developing AF is potentially ‘actionable’ and may lead to enhanced screening and earlier intervention with therapies and preventing transition to persistent or permanent forms of the arrhythmia. Another important report examined how monogenic and polygenic factors influence AF risk and penetrance in the UK Biobank with 0.44% of individuals carrying TTN LOF and an AF prevalence of 14% while the top 0.44% of the polygenic risk score subjects had an AF prevalence of 9.3%.32 The authors hypothesized that monogenic factors confer high penetrance but the common variants in the GPSGRS explained the broader risk of AF.
This work captures the essence of genetic testing in a clinical setting. While testing for certain variants has a role in specialized settings (e.g. TTNtv) because of high penetrance, the utilization in a broad clinical cohort will be dependent on the use of GPS to assess AF risk. Advances in the field will make GPS more feasible in identifying patients at risk in an inpatient and outpatient settings.
2.2. Catheter ablation therapy
The identification of ectopic triggers in the pulmonary veins initiating in vulnerable atria (substrate) established catheter ablation (CA) as the first-line strategy for treatment of symptomatic AF.60,61 Notably, CA is more effective than antiarrhythmic therapy for rhythm control therapy, yet maintenance of sinus rhythm often requires repeat ablation procedures.62 Despite the general safety of repeat ablation, procedure-related complications occur not infrequently.63
The Catheter Ablation vs. Anti-arrhythmic Drug Therapy for Atrial Fibrillation Trial (CABANA) compared CA vs. AADs and reported no significant difference in the primary outcome (composite of death, stroke, bleeding, and cardiac arrest).64 However, patients randomized to ablation reported significant improvement in symptomatic burden and quality of life. This finding was corroborated by the randomized Catheter Ablation Compared With Pharmacological Therapy for Atrial Fibrillation (CAPTAF Trial).65 While efficacy is high with CA (∼70–75% for paroxysmal AF), response is not uniform and some ∼10–15% of patients have recurrence of AF.66 Comorbidities such as obstructive sleep apnoea and obesity are important predictors of AF recurrence but for most patients the reason for recurrence remains unclear.67
Electrical reconnection of initially isolated pulmonary veins represents one of the most important drivers for AF recurrence.68 However, it remains unclear why some patients have pulmonary vein reconnection and others do not. The recent FIRE and ICE trial showed that cryoballoon is non-inferior to radiofrequency ablation but associated with fewer reconnected pulmonary veins.62,68 Here, genetics may provide important insights into pulmonary vein reconnection especially as rare variants in genes encoding structural cell integrity have been associated with AF.26,30 Importantly, determining which energy source for ablation is likely to have the best outcome for each patient would facilitate a personalized approach to CA of AF. The evaluation of the impact of AF ablation beyond the pulmonary veins, i.e., isolation of the left atrial appendage or the superior vena cava, ablation of complex fractionated electrograms, bidirectional block of linear lesions, and posterior wall ablation continues to evolve.69 To date, the clinical benefit of a genotype-based CA approach remains controversial, and additional data are required before definitive recommendations can be made for their use in clinical practice. Due to its importance in cardiogenesis and pulmonary vein formation, common genetic variation at the chr4q25/PITX2 locus is the most promising to pursue. A recent study showed that low concentrations of left atrial PITX2 and its surrogate, elevated BMP10 levels, were predictive for AF recurrence after thoracoscopic AF ablation.10
The recently reported primary outcome of the EAST-AFNET 4 trial showed that early initiation of rhythm control therapy, using both AAD and AF ablation, can improve outcomes compared to usual care in patients with recently diagnosed AF.70 This will increase the clinical use of rhythm control therapy, which thus far has mainly been used in selected patients.66 The role of GPS in predicting the population that will benefit the most from rhythm control therapy with better outcomes including less recurrence of AF, reduced strokes and cardiovascular death remains unclear and randomized controlled trials are required. In view of the expected increased use of rhythm control therapy, better ways to deliver effective, accessible therapy to a large population need to be found.
2.3. Antiarrhythmic therapy
The most recent ESC guidelines on the management of AF recommend antiarrhythmic therapy for rhythm control in favour of or at least with equal standing with CA in most indications.61 However, recommendations on which AAD to use are in part based on adverse events instead of therapeutic efficacy. Flecainide and propafenone have voltage-dependent effects, e.g. maximal effect at high heart rates, and a rapid onset of action after oral intake, which renders them suitable for the ‘pill-in-the-pocket’ approach for symptomatic AF episodes.71
Variants of the gene encoding the β1-adrenergic receptor (ADRB1) has been an early focus of a pharmacogenomic approach. In an analysis of 543 patients from the Vanderbilt AF Registry, carriers of the Gly389 genotype responded better to rate control in AF and needed lower doses.72 However, in a Japanese cohort of 159 patients with supraventricular arrhythmias, the Gly389 polymorphism decreased antiarrhythmic efficacy of flecainide when co-administered with β-blockers.73 Earlier, a substudy of the Beta-Blocker Evaluation of Survival Trial (BEST) investigated the effect of bucindolol on new-onset AF. While 389 Gly carriers showed no difference, in those with 389 arginine homozygotes, bucindolol reduced new-onset AF by 74% (hazard ratio 0.26, 95% confidence interval 0.12–0.57).74 This led to the very first trial [GENETIC-AF trial (Phase 2 Genotype-Directed Comparative Effectiveness Trial of Bucindolol and Toprol-XL for the Prevention of Symptomatic Atrial Fibrillation/Atrial Flutter in Patients with Heart Failure)] to determine genetic risk prediction in pharmacotherapy of AF. When 267 heart failure patients carrying the ADRB1 Arg389Arg genotype were randomized to bucindolol or metoprolol75, there was no difference in new-onset AF or atrial flutter. However, the study was small and a larger Phase 2 trial is needed.
Other AADs used for the treatment of AF include amiodarone, dronedarone, and Class III drugs. In Europe, sotalol is often used to maintain sinus rhythm while dofetilide is prescribed frequently in the USA. Importantly, dronedarone and amiodarone affect multiple other channels, including inhibition of the cardiac sodium and potassium channels but also other currents (IKr, IKs, IK1, INa).76 Amiodarone is more potent than other AADs,77 and the only agent that can be safely used in patients with AF and heart failure with reduced ejection fraction.78 However, amiodarone’s non-cardiac side effects limit its use as a first-line agent in patients in whom other AADs are available. Importantly, amiodarone is not approved by the FDA for AF.79 Alternatives include dronedarone or sotalol which have been shown to have less efficacy and in some cases higher rates of QT prolongation and torsades de pointes.79 In conclusion, AAD therapy is complex and requires deep understanding of mechanisms of action and the associated physiological consequences. This leads to underutilization and discordance of clinical practice to guideline recommendations.80,81
2.4. Clinical predictors of response to AADs for AF
Due to the proarrhythmic effects of AADs in some patients, researchers have long sought predicting response. In 1980, therapeutic efficacy of AADs for ventricular tachycardia was assessed by suppression of pacing-induced arrhythmia during an electrophysiology study.82 Drugs that suppressed pacing-induced ventricular tachycardia were more effective in maintaining freedom from arrhythmia.82 Shortly after, a randomized trial confirmed the findings.83 However, predicting response to AAD treatment in AF is more challenging as this approach cannot be used to assess therapeutic efficacy due to the chaotic nature of AF and unclear mechanisms. Generally, rhythm control therapies are less efficacious in persistent or longstanding persistent forms of AF and in patients with enlarged atria or poorly treated comorbidities such as hypertension.84,85 However, most studies are limited to investigating AF recurrence after CA.
A recent study examined the impact of obesity on response to antiarrhythmic therapy in 311 patients with AF and in diet-induced obese mice.86 Obese patients were less likely to respond to sodium channel blocker AADs compared with non-obese controls. Similarly, treating obese mice with sotalol was associated with a greater reduction in pacing-induced AF burden as compared with sodium channel blockers.86 The same group showed in a separate study that there was reduced expression of the cardiac sodium channel in obese mice while potassium channel expression and atrial fibrosis were increased, confirming their findings in patients.87
2.5. Genotype-based approach to the treatment of AF—where are we now? Antiarrhythmic therapy
Variability in response to pharmacological and non-pharmacological therapy is established in cardiovascular medicine. Recognizing that common genetic variants increase susceptibility to AF, raises the possibility that they may also modulate responses to rhythm control therapy. One of the first pharmacogenetic studies investigated whether response to AADs for symptomatic AF was modified by the angiotensin-converting enzyme (ACE) I/D polymorphism.88 This 287 base pair region within intron 16, associated with increased ACE activity and cardiac fibrosis, was a significant predictor of failure to respond to AADs in patients with early-onset AF.89 There was a graded response with patients carrying the ACE II genotype experiencing the greatest reduction of symptoms while those with the DD genotype responded poorly. A second study evaluated if SNPs on chr4q25, chr16q22, and chr1q21 modulated response to AADs for AF. Only the rs10033464 SNP on chr4q25 was an independent predictor of successful rhythm control with patients carrying the ancestral allele having ∼four-fold increased odds of maintaining sinus rhythm.90 There was also a differential response to Class I vs. Class III AADs with individuals carrying the rs10033464 variant allele responding better to Class I as compared to Class III. However, both analyses were performed in whites of European descent, and it remains unclear if the same effect is seen across race–ethnicity. While the findings have to be replicated in other, contemporary cohorts, in 2016, Syeda et al.91 reported that variable PITX2 expression not only modulated atrial resting membrane potential but also confirmed the clinical observation that flecainide was more effective at suppressing AF than sotalol in Pitx2c+/− mice. There is, however, no data on PITX2 guided antiarrhythmic therapy in humans.
Overall, the observed effects in genetic risk prediction in AAD therapy are modest and with the rising importance of CA, GPS studies on AAD response have not been pursued. As rare variants do not confer a large population-attributable risk, a GPS approach will need to be developed. With an expected rise of rhythm control therapy after EAST, CA centres will not be able to offer enough patients a procedure, consequently many patients will remain on AAD. There is tremendous potential in the application of genetic risk scoring to guide AAD therapy in a broad and diverse population. It is important to emphasize that almost all pharmacogenetics studies evaluating response to AADs for AF have not been replicated and the effects are modest, reinforcing the need for large scale replication and performing randomized clinical trials before such approaches are introduced into clinical practice.
2.6. Ablation therapy
The prediction of AF recurrence after CA may help identify patients who would benefit from frequent follow-up and electrocardiographic surveillance (Figure 1). Earlier studies using a candidate gene approach have assessed recurrence of AF after ablation. Similar to risk for new-onset AF, PITX2 was a major focus for AF recurrence after CA. Two chr4q25 SNPs (rs2200733 and rs10033464; surrogate markers for PITX2) were investigated in a German cohort after CA.92 While clinical and echocardiographic variables failed to predict recurrence, any variant alleles were associated with early and late recurrence of atrial arrhythmias after CA.
Figure 1.
Overview of current (A) and proposed (B) genotype-guided therapy for atrial fibrillation. AAD, antiarrhythmic drug; QoL, quality of life.
One study examined whether the ACE I/D polymorphism predicted recurrence of AF after CA. Confirming earlier findings with antiarrhythmic therapy, the DD genotype and left atrial enlargement were significantly associated with AF recurrence.93 Importantly, these early studies suggested that genes implicated in the pathogenesis of AF may not only predict AF risk but also response to therapy and should be assessed individually. Epoxyeicosatrienoic acids encoded by the gene EPHX2 have been implicated as modulators of cardiac ion channels.94 A German study with 218 allele carriers and 268 controls showed that the rs751141 variant in EPHX2 is associated with increased risk of AF recurrence after ablation.95 As nitric oxide has been implicated in modulating cardiac vagal activity and cardiac remodelling,96 a study of 1000 Korean patients showed that the rs1799983 SNP in the eNOS3 gene was associated with early recurrence of AF.97 Several studies in patients of Asian ancestry showed that inflammatory pathways [interleukin-6 (ILR6) gene in a Chinese population],98 heme oxygenase-1 gene (HO-1) in a Taiwanese cohort,99 and angiotensinogen M235T polymorphism in a Chinese population though not significant in this cohort, may have value as predictors of AF recurrence after ablation.100 In another study, an SCN10A SNP (rs6795970) in a Chinese population independently increased the risk of AF recurrence.101 However, the value of screening for random rare variants as predictors of AF recurrence after ablation remains questionable. For example, rare variants in cardiac sodium channel genes (SCN5A, SCN1B–4B) were not significantly associated with outcome of CA in a Caucasian population.102
As earlier studies suggested that rs2200733 and rs10033464 SNPs on chr4q25 were significant predictors of early and late recurrence of AF, Shoemaker et al.103 performed a GWAS to verify these findings in 991 patients. Only rs2200733 on chr4q25 was significantly associated with recurrence of atrial arrhythmias. In contrast, a Korean study with 1068 AF patients found that no AF genetic loci predicted recurrence after CA.104 However, these findings should be viewed in the context of different race–ethnicity studied (European whites vs. Asians). Nonetheless, a recent Korean study showed that a GPS consisting of five SNPs on chr4q25 and one from chr16q22 was moderately predictive of AF recurrence after ablation.105 A histidine-rich calcium-binding protein SNP (rs3745297) was associated with a recurrence after CA in a Japanese cohort of 382 patients.106
Recently, a large multi-centre study investigated AF recurrence after CA in a large cohort of European ancestry (n = 3259) using a comprehensive polygenic risk score consisting of 929 SNPs.107 Although there was no significant correlation between the polygenic risk score and AF recurrence, common genetic variation particularly at the chr4q25 locus near PITX2 was significantly associated with AF recurrence. PITX2 variants are of clinical and pathophysiological interest, since pulmonary veins are the only established target for CA of AF. While this study suggested that AF recurrence after CA may represent a genetically different phenotype, identifying common genetic variants associated with recurrence is important and is being evaluated in ongoing studies. Their clinical value in predicting response to CA and/or other therapies for AF need to be demonstrated in randomized clinical trials. Many earlier studies using candidate SNPs have not been replicated and robust data are lacking. The value of a candidate SNP-based approach is debatable and questions remain on the number of candidate SNPs were initially investigated in each study. However, the GPS remains the most promising approach for the clinical setting but is not yet ready for prime time. Table 1 summarizes the existing literature on genomic risk for AF recurrence after CA.
Table 1.
Overview of studies investigating genome-guided therapy for AF
| Gene or SNP | Study design | Primary end point | Results | Statistical value | Reference |
|---|---|---|---|---|---|
| ACE I/D polymorphism | Response to AAD therapy (Caucasian, Vanderbilt AF Registry) | Success of therapy defined as ≥75% reduction in symptomatic AF burden (questionnaire based) | ACE DD/ID genotype in early-onset AF a significant predictor of inadequate response to AAD | OR 2.251, 95% CI 1.056–4.798; P = 0.036 | 88 |
|
Chr4q25: rs2200733, rs10033464 Chr16q22: rs7193343 Chr1q21: rs13376333 |
Response to AAD therapy (Caucasian, Vanderbilt AF Registry) | Success of therapy defined as ≥75% reduction in symptomatic AF burden (questionnaire based) | SNP rs10033464 on 4q25 predicts AF recurrence | OR 3.27, 95% CI 1.7–6.0; P < 0.001 | 90 |
| Variants in 4q25, 16q22, 1q21, β1-adrenergic receptor (ARDB1) | Response to AAD therapy in relation to OSA (Caucasian, Vanderbilt AF Registry) | Success of therapy defined as ≥75% reduction in symptomatic AF burden (questionnaire based) | 4q25 (rs10033464) locus associated with increase of AAD response in patients with mild OSA | OR 10.0, 95% CI 1.03–97.5; P < 0.05 | 108 |
| Recurrence after CA | |||||
| Chr4q25 SNPs (rs2200733, rs10033464) | Single-centre study (Caucasian, Leipzig Heart Center AF ablation registry) | Recurrence of AF after CA | rs2200733 and rs10033464 associated with early and late AF recurrence |
Early recurrence (OR 1.994, 95% CI 1.036–3.837; P = 0.039) Late recurrence (OR 4.182, 95% CI 1.318–12.664; P = 0.011) |
92 |
|
HO-1 gene polymorphism (number of GT repeats) |
Single-centre study (Taiwanese-Asian, Veterans General Hospital Taipeh) | Recurrence of AF after CA | <29 GT repeats associated with increased risk of AF recurrence | HR 1.79, 95% CI 1.10–2.94; P = 0.02 | 99 |
| Angiotensin-converting enzyme (ACE) I/D polymorphism | Single-centre study (Caucasian, Leipzig Heart Center AF ablation registry) | Recurrence of AF after CA | ACE DD genotype associated with AF recurrence | OR 2.251, 95% CI 1.056–4.798; P = 0.036 | 93 |
| EPHX2 polymorphisms (rs41507953, rs751141) | Single-centre study (Caucasian, Charité Berlin) | Recurrence of AF after CA | rs751141 significantly increases risk of AF recurrence |
12 months (OR 3.2, 95% CI 1.237–8.276; P = 0.016) 24 months (OR 6.076, 95% CI 2.244–6.451; P < 0.0001) |
95 |
| IL6R gene polymorphism (rs4845625) | Retrospective study (Chinese-Han, GeneID population) | Recurrence of AF after CA | rs4845625 increases risk of early and late AF recurrence | OR 1.92, 95% CI 1.30–2.81; P = 0.001 | 98 |
| eNOS3 gene polymorphism (rs1799983) | Single-centre study (Korean-Asian, Yonsei AF Ablation Cohort) | Recurrence of AF after CA | rs1799983 increases risk of early recurrence of AF | OR 1.71, 95% CI 1.06–2.79; P = 0.028 | 97 |
|
Chr4q25 (rs2200733, rs10033464), Chr1q21 (rs13376333) Chr 16q22 (rs7193343) |
Multi-centre meta-analysis (Caucasian) | Recurrence of AF after CA | 4q25 SNP (rs2200733) predicted increase of risk of recurrence | HR 1.3, 95% CI 1.1–1.6; P = 0.011 | 103 |
| Chr4q25 (rs6817105, rs2200733), Chr16q22(rs2106261), Chr1q21(rs13376333) | Single-centre study (Korean-Asian, Yonsei AF Ablation Cohort) | Recurrence of AF after CA | No significant correlation between SNPs and outcome | 1068 AF patients and 1068 age- and sex-matched controls | 104 |
| M235T polymorphism | Single-centre study (Chinese-Han, Shanghai Jiao Tong University) | Recurrence of AF after CA | M235T variants were not significantly associated with AF recurrence | 150 symptomatic drug-refractory AF patients | 100 |
| SCN5A, SCN1B–4B, CAV3, GPD1L, SNTA1 and MOG1 variants | Single-centre study (Caucasian, Leipzig Heart Center AF ablation registry) | Recurrence of AF after CA | No association between rare variants and ablation outcome | 137 patients | 102 |
| SCN10A SNP (rs6800541) | Single-centre study (Chinese-Han, Shanghai Jiao Tong University) | Recurrence of AF after CA | rs6800541 is and independent risk factor for AF recurrence | OR 0.36, 95% CI 0.22–0.60; P = 7.04 × 10−5 | 101 |
| The HRC SNP, rs3745297 (T > G, Ser96Ala) | Single-centre (Japanese, Hiroshima University Hospital) | Recurrence of AF after CA | rs3745297 associated with increased risk of AF recurrence | HR 2.66, CI 1.32–5.0; P = 0.007 | 106 |
| Polygenic risk score of 5 SNPs [rs1448818, rs2200733, rs6843082, rs6838973 at Chr4q25 (PITX2) and 1 SNP rs2106261 at Chr 16q22 (ZFHX3)] | Two-centre study (Korean) | Recurrence of AF after CA | Association of recurrence risk and risk score at intermediate or high risk | Intermediate risk (HR 2.00, 95% CI 0.99–4.04) and high risk (HR 2.66, 95% CI 1.32–5.37) | 105 |
| Polygenic risk score of 929 SNPs | Multi-centre study (Caucasian cohort) | Recurrence of AF after CA | No association of polygenic risk score and AF recurrence | 3259 patients from 10 centres | 107 |
AAD, antiarrhythmic drugs; AF, atrial fibrillation; CA, catheter ablation; Chr, chromosome; CI, confidence interval; HR, hazard ratio; OR, odds ratio; OSA, obstructive sleep apnoea; SNP, single-nucleotide polymorphism.
3. Current value of genetics in clinical application
Over the last decade, tremendous progress has been made in defining the genetic architecture of AF with the identification of rare and common genetic variants. However, there is little evidence that existing data from genetic studies are used for clinical decision-making in AF patients in part because of poor understanding of the underlying genetic substrate for AF, the lack of prospective randomized trials, moderate effect sizes, and variability across race and ethnicity. Larger datasets and ongoing biobanking of samples from large cohorts and rhythm control studies may provide deeper genetic insights and guide selection of antiarrhythmic therapy. Although retrospective studies suggest that response to AADs and CA is modulated by common genetic variation, the development of a comprehensive clinical and genetic risk score may enable the translation of genetic discoveries to the bedside care of AF patients.
Polygenic risk scores may be useful tools to identify patients with good therapeutic response or upfront exclude patients from rhythm control therapies that may be associated with proarrhythmia or non-cardiac toxicities. However, the clinical use of a genetic risk score may be limited to the race–ethnicity of the initial GWAS. Many AF studies were performed with data from the UK Biobank, which is not representative of the general population as often indicated. Participants are healthier, more likely to be female and from less socioeconomically deprived locations.109 Studies suggest that not only does the genetic risk of AF vary by race–ethnicity but also response to rhythm control therapy (see Table 1). Since most genetic studies have been performed in whites of European descent, the risk of increasing health disparities is apparent and should be carefully considered. Greater diversification of GWAS may alleviate a bias towards white patients of European descent.110 Importantly, the translation of genetic data to the bedside is also hindered by technological limitations. GWAS and GPS remain a scientific approach that is available in only advanced cardiovascular research centres. Rapid genetic testing or point-of-care analysis to calculate the GPS are future tools that may enable the comprehensive use of AF genetic data in a clinical setting. The tools must be cost-effective to ensure they are available to all patients and not only benefit those who can afford it. The derivation of GPS for AF recurrence from GWAS of new-onset AF risk (e.g. two different clinical questions) may also be a hindrance to clinical application. The screening for undiagnosed AF achievable with far cheaper competing methods111 and clinical risk prediction using novel methods such as machine learning are being evaluated.112 Currently, novel loci for AF risk are valuable for understanding the complex pathophysiology of AF but are of limited use for a clinician to guide his therapy. The risk prediction for stroke of unknown source due to undetected AF would have immediate clinical impact, e.g. anticoagulation, if validated in a prospective and randomized clinical trial.
4. Future perspective on clinical applications of AF genetics
Currently, the major limitation to translation of genetic data to the care of AF patients is the lack of prospective, adequately powered randomized clinical trials demonstrating improved outcomes. Most studies on AF genetics focus on ‘lone AF’ (older studies) and ‘early-onset AF’ (recent studies) which are both loosely defined and rarely used at the bedside. The latter for instance is not mentioned in the most recent ESC AF guidelines.61 Future studies on genetic AF risk and outcomes after therapy must use the clinical terminology of paroxysmal AF, persistent AF, and longstanding persistent AF especially since most guideline recommendations are based on this classification. The often used binary classification of AF or no AF will be less important in the future with AF burden becoming the metric of choice to assess therapeutic efficacy.113 An ongoing cross-over study is investigating whether common genetic variants associated with AF modulate response to Class I vs. Class III AADs in patients with symptomatic AF (NCT02347111). Genetic sub-analysis of EAST-AFNET 4 and CABANA trials will also provide new perspectives for genetics-based targeted therapies for AF. The insights from these trials will not only provide new data to improve targeted genetic testing but also help develop a comprehensive clinical and genetic risk score enabling the translation of genetic data to the bedside care of patients with AF. All the major cardiovascular trials must include a genetic substudy to build genetic data on those that are treated (sicker, inpatient cohorts) rather than healthier biobank cohorts. This would also provide clinical and randomized data for the application of GPS in many trial populations whose results often shape guidelines.
Steps are needed to bring AF genetics from bench to bedside including:
Increase genetic data on AF patients and those with concomitant disease.
Accompany major cardiovascular trials with a genetic substudy.
Use polygenic risk scores to predict the probability of a disease-related event.
Derive polygenic risk scores for a specific prediction from GWAS data of the relevant cohort.
Integrate clinical and genetic risk data into a comprehensive risk prediction tool.
Use the clinically relevant classification of AF (paroxysmal, persistent, longstanding persistent).
Binary AF constellation will lose importance, use AF burden (via implanted loop recorder).
Be aware of racial and socio-economic disparities that GPS usage will increase.
Make genotyping cheaper and accessible to non-academic centres
Considering the economic burden and number of patients with AF by 2030 (∼12 million), even small improvements in personalized care by the identification of individuals who benefit the most from antiarrhythmic therapy or CA could lead to substantial improvements in patient outcomes and reduction in health care costs.
Funding
D.D. is supported by the National Institutes of Health (NIH) grants R01 HL138737, T32 HL139439, T32 HL139439, and 1R01 HL150586. R.B.S. has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program under the grant agreement No. 648131, from the European Union’s Horizon 2020 research and innovation program under the grant agreement No. 847770 (AFFECT-EU), and German Center for Cardiovascular Research (DZHK e.V.) (81Z1710103); German Ministry of Research and Education (BMBF 01ZX1408A) and ERACoSysMed3 (031L0239). P.K. was partially supported by European Union BigData@Heart (grant agreement EU IMI 116074), British Heart Foundation (FS/13/43/30324; PG/17/30/32961; and PG/20/22/35093), German Centre for Cardiovascular Research supported by the German Ministry of Education and Research (DZHK, via a grant to AFNET), and Leducq Foundation. P.K. is listed as inventor on two patents held by University of Birmingham (Atrial Fibrillation Therapy WO 2015140571, Markers for Atrial Fibrillation WO 2016012783). P.K. receives research support for basic, translational, and clinical research projects from European Union, British Heart Foundation, Leducq Foundation, Medical Research Council (UK), and German Centre for Cardiovascular Research.
Conflict of interest: R.B.S. has received lecture fees and advisory board fees from BMS/Pfizer outside this work. P.K. receives research support for basic, translational, and clinical research projects from European Union, British Heart Foundation, Leducq Foundation, Medical Research Council (UK), and German Centre for Cardiovascular Research, from several drug and device companies active in atrial fibrillation and has received honoraria from several such companies in the past, but not in the last 3 years. All other authors declared no conflict of interest.
This article is part of the Spotlight Issue on Atrial Fibrillation.
References
- 1. Chugh SS, Havmoeller R, Narayanan K, Singh D, Rienstra M, Benjamin EJ, Gillum RF, Kim YH, McAnulty JH Jr, Zheng ZJ, Forouzanfar MH, Naghavi M, Mensah GA, Ezzati M, Murray CJ. Worldwide epidemiology of atrial fibrillation: a Global Burden of Disease 2010 Study. Circulation 2014;129:837–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Magnussen C, Niiranen TJ, Ojeda FM, Gianfagna F, Blankenberg S, Njolstad I, Vartiainen E, Sans S, Pasterkamp G, Hughes M, Costanzo S, Donati MB, Jousilahti P, Linneberg A, Palosaari T, de Gaetano G, Bobak M, den Ruijter HM, Mathiesen E, Jorgensen T, Soderberg S, Kuulasmaa K, Zeller T, Iacoviello L, Salomaa V, Schnabel RB, BiomarCa REC. Sex differences and similarities in atrial fibrillation epidemiology, risk factors, and mortality in community cohorts: results from the BiomarCaRE consortium (Biomarker for Cardiovascular Risk Assessment in Europe). Circulation 2017;136:1588–1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Marijon E, Le Heuzey JY, Connolly S, Yang S, Pogue J, Brueckmann M, Eikelboom J, Themeles E, Ezekowitz M, Wallentin L, Yusuf S; RE-LY Investigators. Causes of death and influencing factors in patients with atrial fibrillation: a competing-risk analysis from the randomized evaluation of long-term anticoagulant therapy study. Circulation 2013;128:2192–2201. [DOI] [PubMed] [Google Scholar]
- 4. Schnabel RB, Yin X, Gona P, Larson MG, Beiser AS, McManus DD, Newton-Cheh C, Lubitz SA, Magnani JW, Ellinor PT, Seshadri S, Wolf PA, Vasan RS, Benjamin EJ, Levy D. 50 Year trends in atrial fibrillation prevalence, incidence, risk factors, and mortality in the Framingham Heart Study: a cohort study. Lancet 2015;386:154–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Miyasaka Y, Barnes ME, Gersh BJ, Cha SS, Bailey KR, Abhayaratna WP, Seward JB, Tsang TS. Secular trends in incidence of atrial fibrillation in Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future prevalence. Circulation 2006;114:119–125. [DOI] [PubMed] [Google Scholar]
- 6. Di Carlo A, Bellino L, Consoli D, Mori F, Zaninelli A, Baldereschi M, Cattarinussi A, D'Alfonso MG, Gradia C, Sgherzi B, Pracucci G, Piccardi B, Polizzi B, Inzitari D; National Research Program: Progetto FAI. La Fibrillazione Atriale in Italia. Prevalence of atrial fibrillation in the Italian elderly population and projections from 2020 to 2060 for Italy and the European Union: the FAI Project. Europace 2019;21:1468–1475. [DOI] [PubMed] [Google Scholar]
- 7. Kirchhof P, Ammentorp B, Darius H, De Caterina R, Le Heuzey JY, Schilling RJ, Schmitt J, Zamorano JL. Management of atrial fibrillation in seven European countries after the publication of the 2010 ESC Guidelines on atrial fibrillation: primary results of the PREvention oF thromboemolic events–European Registry in Atrial Fibrillation (PREFER in AF). Europace 2014;16:6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Fabritz L, Crijns HJGM, Guasch E, Goette A, Häusler KG, Kotecha D, Lewalter T, Meyer C, Potpara TS, Rienstra M, Schnabel RB, Willems S, Breithardt G, Camm AJ, Chan A, Chua W, de Melis M, Dimopoulou C, Dobrev D, Easter C, Eckardt L, Haase D, Hatem S, Healey JS, Heijman J, Hohnloser SH, Huebner T, Ilyas BS, Isaacs A, Kutschka I, Leclercq C, Lip GYH, Marinelli EA, Merino JL, Mont L, Nabauer M, Oldgren J, Pürerfellner H, Ravens U, Savelieva I, Sinner MF, Sitch A, Smolnik R, Steffel J, Stein K, Stoll M, Svennberg E, Thomas D, Van Gelder IC, Vardar B, Wakili R, Wieloch M, Zeemering S, Ziegler PD, Heidbuchel H, Hindricks G, Schotten U, Kirchhof P. Dynamic risk assessment to improve quality of care in patients with atrial fibrillation: the 7th AFNET/EHRA consensus conference. Europace 2021;23:329–344. [DOI] [PubMed] [Google Scholar]
- 9. Fabritz L, Guasch E, Antoniades C, Bardinet I, Benninger G, Betts TR, Brand E, Breithardt G, Bucklar-Suchankova G, Camm AJ, Cartlidge D, Casadei B, Chua WWL, Crijns HJGM, Deeks J, Hatem S, Hidden-Lucet F, Kääb S, Maniadakis N, Martin S, Mont L, Reinecke H, Sinner MF, Schotten U, Southwood T, Stoll M, Vardas P, Wakili R, West A, Ziegler A, Kirchhof P. Defining the major health modifiers causing atrial fibrillation: a roadmap to underpin personalized prevention and treatment. Nat Rev Cardiol 2016;13:230–237. [DOI] [PubMed] [Google Scholar]
- 10. Reyat JS, Chua W, Cardoso VR, Witten A, Kastner PM, Kabir SN, Sinner MF, Wesselink R, Holmes AP, Pavlovic D, Stoll M, Kaab S, Gkoutos GV, de Groot JR, Kirchhof P, Fabritz L. Reduced left atrial cardiomyocyte PITX2 and elevated circulating BMP10 predict atrial fibrillation after ablation. JCI Insight 2020;5:e139179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Christophersen IE, Ravn LS, Budtz-Joergensen E, Skytthe A, Haunsoe S, Svendsen JH, Christensen K. Familial aggregation of atrial fibrillation. Circ Arrhyth Electrophysiol 2009;2:378–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Weng L-C, Choi SH, Klarin D, Smith JG, Loh P-R, Chaffin M, Roselli C, Hulme OL, Lunetta KL, Dupuis J, Benjamin EJ, Newton-Cheh C, Kathiresan S, Ellinor PT, Lubitz SA. Heritability of atrial fibrillation. Circ Cardiovasc Genet 2017;10:e001838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wolff L. Familial auricular fibrillation. N Engl J Med 1943;229:396–398. [Google Scholar]
- 14. Gudbjartsson DF, Arnar DO, Helgadottir A, Gretarsdottir S, Holm H, Sigurdsson A, Jonasdottir A, Baker A, Thorleifsson G, Kristjansson K, Palsson A, Blondal T, Sulem P, Backman VM, Hardarson GA, Palsdottir E, Helgason A, Sigurjonsdottir R, Sverrisson JT, Kostulas K, Ng MC, Baum L, So WY, Wong KS, Chan JC, Furie KL, Greenberg SM, Sale M, Kelly P, MacRae CA, Smith EE, Rosand J, Hillert J, Ma RC, Ellinor PT, Thorgeirsson G, Gulcher JR, Kong A, Thorsteinsdottir U, Stefansson K. Variants conferring risk of atrial fibrillation on chromosome 4q25. Nature 2007;448:353–357. [DOI] [PubMed] [Google Scholar]
- 15. Chen Y-H, Xu S-J, Bendahhou S, Wang X-L, Wang Y, Xu W-Y, Jin H-W, Sun H, Su X-Y, Zhuang Q-N, Yang Y-Q, Li Y-B, Liu Y, Xu H-J, Li X-F, Ma N, Mou C-P, Chen Z, Barhanin J, Huang W. KCNQ1 gain-of-function mutation in familial atrial fibrillation. Science 2003;299:251–254. [DOI] [PubMed] [Google Scholar]
- 16. Makiyama T, Akao M, Shizuta S, Doi T, Nishiyama K, Oka Y, Ohno S, Nishio Y, Tsuji K, Itoh H, Kimura T, Kita T, Horie M. A novel SCN5A gain-of-function mutation M1875T associated with familial atrial fibrillation. J Am Coll Cardiol 2008;52:1326–1334. [DOI] [PubMed] [Google Scholar]
- 17. Darbar D, Kannankeril PJ, Donahue BS, Kucera G, Stubblefield T, Haines JL, George AL, Roden DM. Cardiac sodium channel (SCN5A) variants associated with atrial fibrillation. Circulation 2008;117:1927–1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Savio-Galimberti E, Weeke P, Muhammad R, Blair M, Ansari S, Short L, Atack TC, Kor K, Vanoye CG, Olesen MS, LuCamp Yang T, George AL Jr, Roden DM, Darbar D. SCN10A/Nav1.8 modulation of peak and late sodium currents in patients with early onset atrial fibrillation. Cardiovasc Res 2014;104:355–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Yang T, Yang P, Roden DM, Darbar D. Novel KCNA5 mutation implicates tyrosine kinase signaling in human atrial fibrillation. Heart Rhythm 2010;7:1246–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Hodgson-Zingman DM, Karst ML, Zingman LV, Heublein DM, Darbar D, Herron KJ, Ballew JD, de Andrade M, Burnett JC, Olson TM. Atrial natriuretic peptide frameshift mutation in familial atrial fibrillation. N Engl J Med 2008;359:158–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Holm H, Gudbjartsson DF, Sulem P, Masson G, Helgadottir HT, Zanon C, Magnusson OT, Helgason A, Saemundsdottir J, Gylfason A, Stefansdottir H, Gretarsdottir S, Matthiasson SE, Thorgeirsson G, Jonasdottir A, Sigurdsson A, Stefansson H, Werge T, Rafnar T, Kiemeney LA, Parvez B, Muhammad R, Roden DM, Darbar D, Thorleifsson G, Walters GB, Kong A, Thorsteinsdottir U, Arnar DO, Stefansson K. A rare variant in MYH6 is associated with high risk of sick sinus syndrome. Nat Genet 2011;43:316–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Gudbjartsson DF, Holm H, Sulem P, Masson G, Oddsson A, Magnusson OT, Saemundsdottir J, Helgadottir HT, Helgason H, Johannsdottir H, Gretarsdottir S, Gudjonsson SA, Njølstad I, Løchen M-L, Baum L, Ma RCW, Sigfusson G, Kong A, Thorgeirsson G, Sverrisson JT, Thorsteinsdottir U, Stefansson K, Arnar DO. A frameshift deletion in the sarcomere gene MYL4 causes early-onset familial atrial fibrillation. Eur Heart J 2017;38:27–34. [DOI] [PubMed] [Google Scholar]
- 23. Orr N, Arnaout R, Gula LJ, Spears DA, Leong-Sit P, Li Q, Tarhuni W, Reischauer S, Chauhan VS, Borkovich M, Uppal S, Adler A, Coughlin SR, Stainier DYR, Gollob MH. A mutation in the atrial-specific myosin light chain gene (MYL4) causes familial atrial fibrillation. Nat Commun 2016;7:11303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Peng W, Li M, Li H, Tang K, Zhuang J, Zhang J, Xiao J, Jiang H, Li D, Yu Y, Sham PC, Nattel S, Xu Y. Dysfunction of myosin light-chain 4 (MYL4) leads to heritable atrial cardiomyopathy with electrical, contractile, and structural components: evidence from genetically-engineered rats. J A Heart Assoc 2017;6:e007030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Ghazizadeh Z, Kiviniemi T, Olafsson S, Plotnick D, Beerens ME, Zhang K, Gillon L, Steinbaugh MJ, Barrera V, Sui SH, Werdich AA, Kapur S, Eranti A, Gunn J, Jalkanen J, Airaksinen J, Kleber AG, Hollmen M, MacRae CA. Metastable atrial state underlies the primary genetic substrate for MYL4 mutation-associated atrial fibrillation. Circulation 2020;141:301–312. [DOI] [PubMed] [Google Scholar]
- 26. Choi SH, Weng LC, Roselli C, Lin H, Haggerty CM, Shoemaker MB, Barnard J, Arking DE, Chasman DI, Albert CM, Chaffin M, Tucker NR, Smith JD, Gupta N, Gabriel S, Margolin L, Shea MA, Shaffer CM, Yoneda ZT, Boerwinkle E, Smith NL, Silverman EK, Redline S, Vasan RS, Burchard EG, Gogarten SM, Laurie C, Blackwell TW, Abecasis G, Carey DJ, Fornwalt BK, Smelser DT, Baras A, Dewey FE, Jaquish CE, Papanicolaou GJ, Sotoodehnia N, Van Wagoner DR, Psaty BM, Kathiresan S, Darbar D, Alonso A, Heckbert SR, Chung MK, Roden DM, Benjamin EJ, Murray MF, Lunetta KL, Lubitz SA, Ellinor PT; DiscovEHR study and the NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium. Association between titin loss-of-function variants and early-onset atrial fibrillation. JAMA 2018;320:2354–2364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Goodyer WR, Dunn K, Caleshu C, Jackson M, Wylie J, Moscarello T, Platt J, Reuter C, Smith A, Trela A, Ceresnak SR, Motonaga KS, Yang AE, Dubin P, Perez AM. Broad genetic testing in a clinical setting uncovers a high prevalence of titin loss-of-function variants in very early onset atrial fibrillation. Circ Genom Precis Med 2019;12:e002713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Huang K, Trinder M, Roston TM, Laksman ZW, Brunham LR. The interplay between titin, polygenic risk and modifiable cardiovascular risk factors in atrial fibrillation. Can J Cardiol 2020. [DOI] [PubMed] [Google Scholar]
- 29. Darbar D, Roden DM. Genetic mechanisms of atrial fibrillation: impact on response to treatment. Nat Rev Cardiol 2013;10:317–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Thorolfsdottir RB, Sveinbjornsson G, Sulem P, Helgadottir A, Gretarsdottir S, Benonisdottir S, Magnusdottir A, Davidsson OB, Rajamani S, Roden DM, Darbar D, Pedersen TR, Sabatine MS, Jonsdottir I, Arnar DO, Thorsteinsdottir U, Gudbjartsson DF, Holm H, Stefansson K. A missense variant in PLEC increases risk of atrial fibrillation. J Am Coll Cardiol 2017;70:2157–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Ahlberg G, Refsgaard L, Lundegaard PR, Andreasen L, Ranthe MF, Linscheid N, Nielsen JB, Melbye M, Haunsø S, Sajadieh A, Camp L, Olesen S-P, Rasmussen S, Lundby A, Ellinor PT, Holst AG, Svendsen JH, Olesen MS. Rare truncating variants in the sarcomeric protein titin associate with familial and early-onset atrial fibrillation. Nat Commun 2018;9:4316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Choi SH, Jurgens SJ, Weng L-C, Pirruccello JP, Roselli C, Chaffin M, Lee CJ-Y, Hall AW, Khera AV, Lunetta KL, Lubitz SA, Ellinor PT. Monogenic and polygenic contributions to atrial fibrillation risk. Circ Res 2020;126:200–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Herman DS, Lam L, Taylor MRG, Wang L, Teekakirikul P, Christodoulou D, Conner L, DePalma SR, McDonough B, Sparks E, Teodorescu DL, Cirino AL, Banner NR, Pennell DJ, Graw S, Merlo M, Di Lenarda A, Sinagra G, Bos JM, Ackerman MJ, Mitchell RN, Murry CE, Lakdawala NK, Ho CY, Barton PJR, Cook SA, Mestroni L, Seidman JG, Seidman CE. Truncations of titin causing dilated cardiomyopathy. N Engl J Med 2012;366:619–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Akinrinade O, Ollila L, Vattulainen S, Tallila J, Gentile M, Salmenperä P, Koillinen H, Kaartinen M, Nieminen MS, Myllykangas S, Alastalo T-P, Koskenvuo JW, Heliö T. Genetics and genotype–phenotype correlations in Finnish patients with dilated cardiomyopathy. Eur Heart J 2015;36:2327–2337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Tayal U, Newsome S, Buchan R, Whiffin N, Walsh R, Barton PJ, Ware JS, Cook SA, Prasad SK. Truncating variants in titin independently predict early arrhythmias in patients with dilated cardiomyopathy. J Am Coll Cardiol 2017;69:2466–2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Ware JS, Cook SA. Role of titin in cardiomyopathy: from DNA variants to patient stratification. Nat Rev Cardiol 2018;15:241–252. [DOI] [PubMed] [Google Scholar]
- 37. Linschoten M, Teske AJ, Baas AF, Vink A, Dooijes D, Baars HF, Asselbergs FW. Truncating Titin (TTN) variants in chemotherapy-induced cardiomyopathy. J Card Fail 2017;23:476–479. [DOI] [PubMed] [Google Scholar]
- 38. Ware JS, Li J, Mazaika E, Yasso CM, DeSouza T, Cappola TP, Tsai EJ, Hilfiker-Kleiner D, Kamiya CA, Mazzarotto F, Cook SA, Halder I, Prasad SK, Pisarcik J, Hanley-Yanez K, Alharethi R, Damp J, Hsich E, Elkayam U, Sheppard R, Kealey A, Alexis J, Ramani G, Safirstein J, Boehmer J, Pauly DF, Wittstein IS, Thohan V, Zucker MJ, Liu P, Gorcsan J, McNamara DM, Seidman CE, Seidman JG, Arany Z. Shared genetic predisposition in peripartum and dilated cardiomyopathies. N Engl J Med 2016;374:233–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Osmancik P, Herman D, Neuzil P, Hala P, Taborsky M, Kala P, Poloczek M, Stasek J, Haman L, Branny M, Chovancik J, Cervinka P, Holy J, Kovarnik T, Zemanek D, Havranek S, Vancura V, Opatrny J, Peichl P, Tousek P, Lekesova V, Jarkovsky J, Novackova M, Benesova K, Widimsky P, Reddy VY. Left atrial appendage closure versus direct oral anticoagulants in high-risk patients with atrial fibrillation. J Am Coll Cardiol 2020;75:3122–3135. [DOI] [PubMed] [Google Scholar]
- 40. Collins MM, Ahlberg G, Hansen CV, Guenther S, Marín-Juez R, Sokol AM, El-Sammak H, Piesker J, Hellsten Y, Olesen MS, Stainier DYR, Lundegaard PR. Early sarcomere and metabolic defects in a zebrafish pitx2c cardiac arrhythmia model. Proc Natl Acad Sci USA 2019;116:24115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ellinor PT, Lunetta KL, Glazer NL, Pfeufer A, Alonso A, Chung MK, Sinner MF, de Bakker PIW, Mueller M, Lubitz SA, Fox E, Darbar D, Smith NL, Smith JD, Schnabel RB, Soliman EZ, Rice KM, Van Wagoner DR, Beckmann B-M, van Noord C, Wang K, Ehret GB, Rotter JI, Hazen SL, Steinbeck G, Smith AV, Launer LJ, Harris TB, Makino S, Nelis M, Milan DJ, Perz S, Esko T, Köttgen A, Moebus S, Newton-Cheh C, Li M, Möhlenkamp S, Wang TJ, Linda Kao WH, Vasan RS, Nöthen MM, MacRae CA, Ch Stricker BH, Hofman A, Uitterlinden AG, Levy D, Boerwinkle E, Metspalu A, Topol EJ, Chakravarti A, Gudnason V, Psaty BM, Roden DM, Meitinger T, Wichmann HE, Witteman JCM, Barnard J, Arking DE, Benjamin EJ, Heckbert SR, Kääb S. Common variants in KCNN3 are associated with lone atrial fibrillation. Nat Genet 2010;42:240–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Christophersen IE, Rienstra M, Roselli C, Yin X, Geelhoed B, Barnard J, Lin H, Arking DE, Smith AV, Albert CM, Chaffin M, Tucker NR, Li M, Klarin D, Bihlmeyer NA, Low S-K, Weeke PE, Müller-Nurasyid M, Smith JG, Brody JA, Niemeijer MN, Dörr M, Trompet S, Huffman J, Gustafsson S, Schurmann C, Kleber ME, Lyytikäinen L-P, Seppälä I, Malik R, Horimoto ARVR, Perez M, Sinisalo J, Aeschbacher S, Thériault S, Yao J, Radmanesh F, Weiss S, Teumer A, Choi SH, Weng L-C, Clauss S, Deo R, Rader DJ, Shah SH, Sun A, Hopewell JC, Debette S, Chauhan G, Yang Q, Worrall BB, Paré G, Kamatani Y, Hagemeijer YP, Verweij N, Siland JE, Kubo M, Smith JD, Van Wagoner DR, Bis JC, Perz S, Psaty BM, Ridker PM, Magnani JW, Harris TB, Launer LJ, Shoemaker MB, Padmanabhan S, Haessler J, Bartz TM, Waldenberger M, Lichtner P, Arendt M, Krieger JE, Kähönen M, Risch L, Mansur AJ, Peters A, Smith BH, Lind L, Scott SA, Lu Y, Bottinger EB, Hernesniemi J, Lindgren CM, Wong JA, Huang J, Eskola M, Morris AP, Ford I, Reiner AP, Delgado G, Chen LY, Chen Y-DI, Sandhu RK, Li M, Boerwinkle E, Eisele L, Lannfelt L, Rost N, Anderson CD, Taylor KD, Campbell A, Magnusson PK, Porteous D, Hocking LJ, Vlachopoulou E, Pedersen NL, Nikus K, Orho-Melander M, Hamsten A, Heeringa J, Denny JC, Kriebel J, Darbar D, Newton-Cheh C, Shaffer C, Macfarlane PW, Heilmann-Heimbach S, Almgren P, Huang PL, Sotoodehnia N, Soliman EZ, Uitterlinden AG, Hofman A, Franco OH, Völker U, Jöckel K-H, Sinner MF, Lin HJ, Guo X; METASTROKE Consortium of the ISGC, Neurology Working Group of the CHARGE Consortium, Dichgans M, Ingelsson E, Kooperberg C, Melander O, Loos RJF, Laurikka J, Conen D, Rosand J, van der Harst P, Lokki M-L, Kathiresan S, Pereira A, Jukema JW, Hayward C, Rotter JI, März W, Lehtimäki T, Stricker BH, Chung MK, Felix SB, Gudnason V, Alonso A, Roden DM, Kääb S, Chasman DI, Heckbert SR, Benjamin EJ, Tanaka T, Lunetta KL, Lubitz SA, Ellinor PT; AFGen Consortium. Large-scale analyses of common and rare variants identify 12 new loci associated with atrial fibrillation. Nat Genet 2017;49:946–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Skibsbye L, Poulet C, Diness JG, Bentzen BH, Yuan L, Kappert U, Matschke K, Wettwer E, Ravens U, Grunnet M, Christ T, Jespersen T. Small-conductance calcium-activated potassium (SK) channels contribute to action potential repolarization in human atria. Cardiovasc Res 2014;103:156–167. [DOI] [PubMed] [Google Scholar]
- 44. Hill MC, Kadow ZA, Li L, Tran TT, Wythe JD, Martin JF. A cellular atlas of Pitx2-dependent cardiac development. Development 2019;146:dev180398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Mommersteeg MTM, Brown NA, Prall OWJ, Vries CG-D, Harvey RP, Moorman AFM, Christoffels VM. Pitx2c and Nkx2-5 are required for the formation and identity of the pulmonary myocardium. Circ Res 2007;101:902–909. [DOI] [PubMed] [Google Scholar]
- 46. Syeda F, Kirchhof P, Fabritz L. PITX2-dependent gene regulation in atrial fibrillation and rhythm control. J Physiol 2017;595:4019–4026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Kirchhof P, Kahr PC, Kaese S, Piccini I, Vokshi I, Scheld H-H, Rotering H, Fortmueller L, Laakmann S, Verheule S, Schotten U, Fabritz L, Brown NA. PITX2c is expressed in the adult left atrium, and reducing Pitx2c expression promotes atrial fibrillation inducibility and complex changes in gene expression. Circ Cardiovasc Genet 2011;4:123–133. [DOI] [PubMed] [Google Scholar]
- 48. Kääb S, Darbar D, van Noord C, Dupuis J, Pfeufer A, Newton-Cheh C, Schnabel R, Makino S, Sinner MF, Kannankeril PJ, Beckmann BM, Choudry S, Donahue BS, Heeringa J, Perz S, Lunetta KL, Larson MG, Levy D, MacRae CA, Ruskin JN, Wacker A, Schömig A, Wichmann H-E, Steinbeck G, Meitinger T, Uitterlinden AG, Witteman JCM, Roden DM, Benjamin EJ, Ellinor PT. Large scale replication and meta-analysis of variants on chromosome 4q25 associated with atrial fibrillation. Eur Heart J 2009;30:813–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Benjamin EJ, Rice KM, Arking DE, Pfeufer A, van Noord C, Smith AV, Schnabel RB, Bis JC, Boerwinkle E, Sinner MF, Dehghan A, Lubitz SA, D'Agostino RB Sr, Lumley T, Ehret GB, Heeringa J, Aspelund T, Newton-Cheh C, Larson MG, Marciante KD, Soliman EZ, Rivadeneira F, Wang TJ, Eiríksdottir G, Levy D, Psaty BM, Li M, Chamberlain AM, Hofman A, Vasan RS, Harris TB, Rotter JI, Kao WHL, Agarwal SK, Stricker BHC, Wang K, Launer LJ, Smith NL, Chakravarti A, Uitterlinden AG, Wolf PA, Sotoodehnia N, Köttgen A, van Duijn CM, Meitinger T, Mueller M, Perz S, Steinbeck G, Wichmann HE, Lunetta KL, Heckbert SR, Gudnason V, Alonso A, Kääb S, Ellinor PT, Witteman JCM. Variants in ZFHX3 are associated with atrial fibrillation in individuals of European ancestry. Nat Genet 2009;41:879–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Low S-K, Takahashi A, Ebana Y, Ozaki K, Christophersen IE, Ellinor PT, Ogishima S, Yamamoto M, Satoh M, Sasaki M, Yamaji T, Iwasaki M, Tsugane S, Tanaka K, Naito M, Wakai K, Tanaka H, Furukawa T, Kubo M, Ito K, Kamatani Y, Tanaka T, Consortium AF. Identification of six new genetic loci associated with atrial fibrillation in the Japanese population. Nat Genet 2017;49:953–958. [DOI] [PubMed] [Google Scholar]
- 51. Lee J-Y, Kim T-H, Yang P-S, Lim HE, Choi E-K, Shim J, Shin E, Uhm J-S, Kim J-S, Joung B, Oh S, Lee M-H, Kim Y-H, Pak H-N. Korean atrial fibrillation network genome-wide association study for early-onset atrial fibrillation identifies novel susceptibility loci. Eur Heart J 2017;38:2586–2594. [DOI] [PubMed] [Google Scholar]
- 52. Nielsen JB, Thorolfsdottir RB, Fritsche LG, Zhou W, Skov MW, Graham SE, Herron TJ, McCarthy S, Schmidt EM, Sveinbjornsson G, Surakka I, Mathis MR, Yamazaki M, Crawford RD, Gabrielsen ME, Skogholt AH, Holmen OL, Lin M, Wolford BN, Dey R, Dalen H, Sulem P, Chung JH, Backman JD, Arnar DO, Thorsteinsdottir U, Baras A, O’Dushlaine C, Holst AG, Wen X, Hornsby W, Dewey FE, Boehnke M, Kheterpal S, Mukherjee B, Lee S, Kang HM, Holm H, Kitzman J, Shavit JA, Jalife J, Brummett CM, Teslovich TM, Carey DJ, Gudbjartsson DF, Stefansson K, Abecasis GR, Hveem K, Willer CJ. Biobank-driven genomic discovery yields new insight into atrial fibrillation biology. Nat Genet 2018;50:1234–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Roselli C, Chaffin MD, Weng L-C, Aeschbacher S, Ahlberg G, Albert CM, Almgren P, Alonso A, Anderson CD, Aragam KG, Arking DE, Barnard J, Bartz TM, Benjamin EJ, Bihlmeyer NA, Bis JC, Bloom HL, Boerwinkle E, Bottinger EB, Brody JA, Calkins H, Campbell A, Cappola TP, Carlquist J, Chasman DI, Chen LY, Chen Y-DI, Choi E-K, Choi SH, Christophersen IE, Chung MK, Cole JW, Conen D, Cook J, Crijns HJ, Cutler MJ, Damrauer SM, Daniels BR, Darbar D, Delgado G, Denny JC, Dichgans M, Dörr M, Dudink EA, Dudley SC, Esa N, Esko T, Eskola M, Fatkin D, Felix SB, Ford I, Franco OH, Geelhoed B, Grewal RP, Gudnason V, Guo X, Gupta N, Gustafsson S, Gutmann R, Hamsten A, Harris TB, Hayward C, Heckbert SR, Hernesniemi J, Hocking LJ, Hofman A, Horimoto ARVR, Huang J, Huang PL, Huffman J, Ingelsson E, Ipek EG, Ito K, Jimenez-Conde J, Johnson R, Jukema JW, Kääb S, Kähönen M, Kamatani Y, Kane JP, Kastrati A, Kathiresan S, Katschnig-Winter P, Kavousi M, Kessler T, Kietselaer BL, Kirchhof P, Kleber ME, Knight S, Krieger JE, Kubo M, Launer LJ, Laurikka J, Lehtimäki T, Leineweber K, Lemaitre RN, Li M, Lim HE, Lin HJ, Lin H, Lind L, Lindgren CM, Lokki M-L, London B, Loos RJF, Low S-K, Lu Y, Lyytikäinen L-P, Macfarlane PW, Magnusson PK, Mahajan A, Malik R, Mansur AJ, Marcus GM, Margolin L, Margulies KB, März W, McManus DD, Melander O, Mohanty S, Montgomery JA, Morley MP, Morris AP, Müller-Nurasyid M, Natale A, Nazarian S, Neumann B, Newton-Cheh C, Niemeijer MN, Nikus K, Nilsson P, Noordam R, Oellers H, Olesen MS, Orho-Melander M, Padmanabhan S, Pak H-N, Paré G, Pedersen NL, Pera J, Pereira A, Porteous D, Psaty BM, Pulit SL, Pullinger CR, Rader DJ, Refsgaard L, Ribasés M, Ridker PM, Rienstra M, Risch L, Roden DM, Rosand J, Rosenberg MA, Rost N, Rotter JI, Saba S, Sandhu RK, Schnabel RB, Schramm K, Schunkert H, Schurman C, Scott SA, Seppälä I, Shaffer C, Shah S, Shalaby AA, Shim J, Shoemaker MB, Siland JE, Sinisalo J, Sinner MF, Slowik A, Smith AV, Smith BH, Smith JG, Smith JD, Smith NL, Soliman EZ, Sotoodehnia N, Stricker BH, Sun A, Sun H, Svendsen JH, Tanaka T, Tanriverdi K, Taylor KD, Teder-Laving M, Teumer A, Thériault S, Trompet S, Tucker NR, Tveit A, Uitterlinden AG, Van Der Harst P, Van Gelder IC, Van Wagoner DR, Verweij N, Vlachopoulou E, Völker U, Wang B, Weeke PE, Weijs B, Weiss R, Weiss S, Wells QS, Wiggins KL, Wong JA, Woo D, Worrall BB, Yang P-S, Yao J, Yoneda ZT, Zeller T, Zeng L, Lubitz SA, Lunetta KL, Ellinor PT. Multi-ethnic genome-wide association study for atrial fibrillation. Nat Genet 2018;50:1225–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lubitz SA, Lunetta KL, Lin H, Arking DE, Trompet S, Li G, Krijthe BP, Chasman DI, Barnard J, Kleber ME, Dörr M, Ozaki K, Smith AV, Müller-Nurasyid M, Walter S, Agarwal SK, Bis JC, Brody JA, Chen LY, Everett BM, Ford I, Franco OH, Harris TB, Hofman A, Kääb S, Mahida S, Kathiresan S, Kubo M, Launer LJ, Macfarlane PW, Magnani JW, McKnight B, McManus DD, Peters A, Psaty BM, Rose LM, Rotter JI, Silbernagel G, Smith JD, Sotoodehnia N, Stott DJ, Taylor KD, Tomaschitz A, Tsunoda T, Uitterlinden AG, Wagoner DRV, Völker U, Völzke H, Murabito JM, Sinner MF, Gudnason V, Felix SB, März W, Chung M, Albert CM, Stricker BH, Tanaka T, Heckbert SR, Jukema JW, Alonso A, Benjamin EJ, Ellinor PT. Novel genetic markers associate with atrial fibrillation risk in Europeans and Japanese. J Am Coll Cardiol 2014;63:1200–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Lubitz SA, Yin X, Lin HJ, Kolek M, Smith JG, Trompet S, Rienstra M, Rost NS, Teixeira PL, Almgren P, Anderson CD, Chen LY, Engström G, Ford I, Furie KL, Guo X, Larson MG, Lunetta KL, Macfarlane PW, Psaty BM, Soliman EZ, Sotoodehnia N, Stott DJ, Taylor KD, Weng L-C, Yao J, Geelhoed B, Verweij N, Siland JE, Kathiresan S, Roselli C, Roden DM, Harst P, Darbar D, Jukema JW, Melander O, Rosand J, Rotter JI, Heckbert SR, Ellinor PT, Alonso A, Benjamin EJ. Genetic risk prediction of atrial fibrillation. Circulation 2017;135:1311–1320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Pulit SL, Weng L-C, McArdle PF, Trinquart L, Choi SH, Mitchell BD, Rosand J, de Bakker PIW, Benjamin EJ, Ellinor PT, Kittner SJ, Lubitz SA, Anderson CD. Atrial fibrillation genetic risk differentiates cardioembolic stroke from other stroke subtypes. Neurol Genet 2018;4:e293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Marston NA, Patel PN, Kamanu FK, Nordio F, Melloni GM, Roselli C, Gurmu Y, Weng L-C, Bonaca MP, Giugliano RP, Scirica BM, O’Donoghue ML, Cannon CP, Anderson CD, Bhatt DL, Steg PG, Cohen M, Storey RF, Sever P, Keech AC, Raz I, Mosenzon O, Antman EM, Braunwald E, Ellinor PT, Lubitz SA, Sabatine MS, Ruff CT. Clinical application of a novel genetic risk score for ischemic stroke in patients with cardiometabolic disease. Circulation 2021;143:470–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Weng L-C, Preis SR, Hulme OL, Larson MG, Choi SH, Wang B, Trinquart L, McManus DD, Staerk L, Lin H, Lunetta KL, Ellinor PT, Benjamin EJ, Lubitz SA. Genetic predisposition, clinical risk factor burden, and lifetime risk of atrial fibrillation. Circulation 2018;137:1027–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH, Natarajan P, Lander ES, Lubitz SA, Ellinor PT, Kathiresan S. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet 2018;50:1219–1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Haïssaguerre M, Jaïs P, Shah DC, Takahashi A, Hocini M, Quiniou G, Garrigue S, Le Mouroux A, Le Métayer P, Clémenty J. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998;339:659–666. [DOI] [PubMed] [Google Scholar]
- 61. Hindricks G, Potpara T, Dagres N, Arbelo E, Bax JJ, Blomstrom-Lundqvist C, Boriani G, Castella M, Dan GA, Dilaveris PE, Fauchier L, Filippatos G, Kalman JM, La Meir M, Lane DA, Lebeau JP, Lettino M, Lip GYH, Pinto FJ, Thomas GN, Valgimigli M, Van Gelder IC, Van Putte BP, Watkins CL; ESC Scientific Document Group. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association of Cardio-Thoracic Surgery (EACTS). Eur Heart J 2021;42:373–498. [DOI] [PubMed] [Google Scholar]
- 62. Kuck K-H, Brugada J, Fürnkranz A, Metzner A, Ouyang F, Chun KRJ, Elvan A, Arentz T, Bestehorn K, Pocock SJ, Albenque J-P, Tondo C. Cryoballoon or radiofrequency ablation for paroxysmal atrial fibrillation. N Engl J Med 2016;374:2235–2245. [DOI] [PubMed] [Google Scholar]
- 63. Kany S, Brachmann J, Lewalter T, Kuck KH, Andresen D, Willems S, Hoffmann E, Eckardt L, Thomas D, Hochadel M, Senges J, Metzner A, Rillig A. Safety and patient-reported outcomes in index ablation versus repeat ablation in atrial fibrillation: insights from the German Ablation Registry. Clin Res Cardiol 2020;110:841–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Packer DL, Mark DB, Robb RA, Monahan KH, Bahnson TD, Poole JE, Noseworthy PA, Rosenberg YD, Jeffries N, Mitchell LB, Flaker GC, Pokushalov E, Romanov A, Bunch TJ, Noelker G, Ardashev A, Revishvili A, Wilber DJ, Cappato R, Kuck KH, Hindricks G, Davies DW, Kowey PR, Naccarelli GV, Reiffel JA, Piccini JP, Silverstein AP, Al-Khalidi HR, Lee KL; CABANA Investigators. Effect of catheter ablation vs antiarrhythmic drug therapy on mortality, stroke, bleeding, and cardiac arrest among patients with atrial fibrillation: the CABANA randomized clinical trial. JAMA 2019;321:1261–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Blomstrom-Lundqvist C, Gizurarson S, Schwieler J, Jensen SM, Bergfeldt L, Kenneback G, Rubulis A, Malmborg H, Raatikainen P, Lonnerholm S, Hoglund N, Mortsell D. Effect of catheter ablation vs antiarrhythmic medication on quality of life in patients with atrial fibrillation: the CAPTAF randomized clinical trial. JAMA 2019;321:1059–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Willems S, Meyer C, de Bono J, Brandes A, Eckardt L, Elvan A, van Gelder I, Goette A, Gulizia M, Haegeli L, Heidbuchel H, Haeusler KG, Kautzner J, Mont L, Ng GA, Szumowski L, Themistoclakis S, Wegscheider K, Kirchhof P. Cabins, castles, and constant hearts: rhythm control therapy in patients with atrial fibrillation. Eur Heart J 2019;40:3793–3799c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Kornej J, Börschel CS, Benjamin EJ, Schnabel RB. Epidemiology of atrial fibrillation in the 21st century. Circ Res 2020;127:4–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Kuck K-H, Albenque J-P, Chun KRJ, Fürnkranz A, Busch M, Elvan A, Schlüter M, Braegelmann KM, Kueffer FJ, Hemingway L, Arentz T, Tondo C, Brugada J, Kuck K-H, Metzner A, Fink T, Lemes C, Chun J, Chen S, Schmidt B, Bologna F, Bordignon S, Elvan A, Mouden M, Arentz T, Müller-Eschenborn B, Jadidi A, Kühne M, Sticherling C, Busch M, Krüger A, Abdiou E, Mont L, Martín EMB, Sanz FA, Neuzil P, Dujka L, Ruiz-Granell R, Barrera A, Salas AR, Albenque J-P, Boveda S, Kühlkamp V, Stanciu B, Pérez-Castellano N, Villacastín JP, Godoy VC. Repeat ablation for atrial fibrillation recurrence post cryoballoon or radiofrequency ablation in the FIRE AND ICE trial. Circ Arrhythm Electrophysiol 2019;12:e007247. [DOI] [PubMed] [Google Scholar]
- 69. Verma A, Jiang C-y, Betts TR, Chen J, Deisenhofer I, Mantovan R, Macle L, Morillo CA, Haverkamp W, Weerasooriya R, Albenque J-P, Nardi S, Menardi E, Novak P, Sanders P. Approaches to catheter ablation for persistent atrial fibrillation. N Engl J Med 2015;372:1812–1822. [DOI] [PubMed] [Google Scholar]
- 70. Kirchhof P, Camm AJ, Goette A, Brandes A, Eckardt L, Elvan A, Fetsch T, van Gelder IC, Haase D, Haegeli LM, Hamann F, Heidbuchel H, Hindricks G, Kautzner J, Kuck KH, Mont L, Ng GA, Rekosz J, Schoen N, Schotten U, Suling A, Taggeselle J, Themistoclakis S, Vettorazzi E, Vardas P, Wegscheider K, Willems S, Crijns H, Breithardt G; EAST-AFNET 4 Trial Investigators. Early rhythm-control therapy in patients with atrial fibrillation. N Engl J Med 2020;383:1305–1316. [DOI] [PubMed] [Google Scholar]
- 71. Alboni P, Botto GL, Baldi N, Luzi M, Russo V, Gianfranchi L, Marchi P, Calzolari M, Solano A, Baroffio R, Gaggioli G. Outpatient treatment of recent-onset atrial fibrillation with the “pill-in-the-pocket” approach. N Engl J Med 2004;351:2384–2391. [DOI] [PubMed] [Google Scholar]
- 72. Parvez B, Chopra N, Rowan S, Vaglio JC, Muhammad R, Roden DM, Darbar D. A common β1-adrenergic receptor polymorphism predicts favorable response to rate-control therapy in atrial fibrillation. J Am Coll Cardiol 2012;59:49–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Doki K, Sekiguchi Y, Kuga K, Aonuma K, Homma M. β1-Adrenergic receptor Arg389Gly polymorphism affects the antiarrhythmic efficacy of flecainide in patients with coadministration of β-blockers. Pharmacogenet Genomics 2016;26:481–485. [DOI] [PubMed] [Google Scholar]
- 74. Aleong RG, Sauer WH, Davis G, Murphy GA, Port JD, Anand IS, Fiuzat M, O'Connor CM, Abraham WT, Liggett SB, Bristow MR. Prevention of atrial fibrillation by bucindolol is dependent on the beta1389 Arg/Gly adrenergic receptor polymorphism. JACC: Heart Failure 2013;1:338–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Piccini JP, Abraham WT, Dufton C, Carroll IA, Healey JS, van Veldhuisen DJ, Sauer WH, Anand IS, White M, Wilton SB, Aleong R, Rienstra M, Krueger SK, Ayala-Paredes F, Khaykin Y, Merkely B, Miloradović V, Wranicz JK, Ilkhanoff L, Ziegler PD, Davis G, Emery LL, Marshall D, Kao DP, Bristow MR, Connolly SJ. Bucindolol for the maintenance of sinus rhythm in a genotype-defined HF population: the GENETIC-AF trial. JACC Heart Fail 2019;7:586–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Kodama I, Kamiya K, Toyama J. Cellular electropharmacology of amiodarone. Cardiovasc Res 1997;35:13–29. [DOI] [PubMed] [Google Scholar]
- 77. Singh BN, Singh SN, Reda DJ, Tang XC, Lopez B, Harris CL, Fletcher RD, Sharma SC, Atwood JE, Jacobson AK, Lewis HD, Raisch DW, Ezekowitz MD. Amiodarone versus sotalol for atrial fibrillation. N Engl J Med 2005;352:1861–1872. [DOI] [PubMed] [Google Scholar]
- 78. Roy D, Talajic M, Nattel S, Wyse DG, Dorian P, Lee KL, Bourassa MG, Arnold JMO, Buxton AE, Camm AJ, Connolly SJ, Dubuc M, Ducharme A, Guerra PG, Hohnloser SH, Lambert J, Le Heuzey J-Y, O'Hara G, Pedersen OD, Rouleau J-L, Singh BN, Stevenson LW, Stevenson WG, Thibault B, Waldo AL. Rhythm control versus rate control for atrial fibrillation and heart failure. N Engl J Med 2008;358:2667–2677. [DOI] [PubMed] [Google Scholar]
- 79. Zimetbaum P. Amiodarone for atrial fibrillation. N Engl J Med 2007;356:935–941. [DOI] [PubMed] [Google Scholar]
- 80. Tardos JG, Ronk CJ, Patel MY, Koren A, Kim MH. US Antiarrhythmic drug treatment for patients with atrial fibrillation: an insurance claims-based report. J Am Heart Assoc 2021;10:e016792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Barnett AS, Kim S, Fonarow GC, Thomas LE, Reiffel JA, Allen LA, Freeman JV, Naccarelli G, Mahaffey KW, Go AS, Kowey PR, Ansell JE, Gersh BJ, Hylek EM, Peterson ED, Piccini JP. Treatment of atrial fibrillation and concordance with the American Heart Association/American College of Cardiology/Heart Rhythm Society Guidelines. Circ Arrhythm Electrophysiol 2017;10:e005051. [DOI] [PubMed] [Google Scholar]
- 82. Mason JW, Winkle RA. Accuracy of the ventricular tachycardia-induction study for predicting long-term efficacy and inefficacy of antiarrhythmic drugs. N Engl J Med 1980;303:1073–1077. [DOI] [PubMed] [Google Scholar]
- 83. Mitchell LB, Duff HJ, Manyari DE, Wyse DG. A randomized clinical trial of the noninvasive and invasive approaches to drug therapy of ventricular tachycardia. N Engl J Med 1987;317:1681–1687. [DOI] [PubMed] [Google Scholar]
- 84. Njoku A, Kannabhiran M, Arora R, Reddy P, Gopinathannair R, Lakkireddy D, Dominic P. Left atrial volume predicts atrial fibrillation recurrence after radiofrequency ablation: a meta-analysis. Europace 2017;20:33–42. [DOI] [PubMed] [Google Scholar]
- 85. Berruezo A, Tamborero D, Mont L, Benito B, Tolosana JM, Sitges M, Vidal B, Arriagada G, Méndez F, Matiello M, Molina I, Brugada J. Pre-procedural predictors of atrial fibrillation recurrence after circumferential pulmonary vein ablation. Eur Heart J 2007;28:836–841. [DOI] [PubMed] [Google Scholar]
- 86. Ornelas-Loredo A, Kany S, Abraham V, Alzahrani Z, Darbar FA, Sridhar A, Ahmed M, Alamar I, Menon A, Zhang M, Chen Y, Hong L, Konda S, Darbar D. Association between obesity-mediated atrial fibrillation and therapy with sodium channel blocker antiarrhythmic drugs. JAMA Cardiol 2020;5:57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. McCauley MD, Hong L, Sridhar A, Menon A, Perike S, Zhang M, Silva IBd, Yan J, Bonini MG, Ai X, Rehman J, Darbar D. Ion channel and structural remodeling in obesity-mediated atrial fibrillation. Circ Arrhythm Electrophysiol 2020;13:e008296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Darbar D, Motsinger AA, Ritchie MD, Gainer JV, Roden DM. Polymorphism modulates symptomatic response to antiarrhythmic drug therapy in patients with lone atrial fibrillation. Heart Rhythm 2007;4:743–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Ocaranza MP, Díaz-Araya G, Carreño JE, Muñoz D, Riveros JP, Jalil JE, Lavandero S. Polymorphism in gene coding for ACE determines different development of myocardial fibrosis in rats. Am J Physiol Heart Circ Physiol 2004;286:H498–H506. [DOI] [PubMed] [Google Scholar]
- 90. Parvez B, Vaglio J, Rowan S, Muhammad R, Kucera G, Stubblefield T, Carter S, Roden D, Darbar D. Symptomatic response to antiarrhythmic drug therapy is modulated by a common single nucleotide polymorphism in atrial fibrillation. J Am Coll Cardiol 2012;60:539–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Syeda F, Holmes AP, Yu TY, Tull S, Kuhlmann SM, Pavlovic D, Betney D, Riley G, Kucera JP, Jousset F, de Groot JR, Rohr S, Brown NA, Fabritz L, Kirchhof P. PITX2 modulates atrial membrane potential and the antiarrhythmic effects of sodium-channel blockers. J Am Coll Cardiol 2016;68:1881–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Husser D, Adams V, Piorkowski C, Hindricks G, Bollmann A. Chromosome 4q25 variants and atrial fibrillation recurrence after catheter ablation. J Am Coll Cardiol 2010;55:747–753. [DOI] [PubMed] [Google Scholar]
- 93. Ueberham L, Bollmann A, Shoemaker MB, Arya A, Adams V, Hindricks G, Husser D. Genetic ACE I/D polymorphism and recurrence of atrial fibrillation after catheter ablation. Circ Arrhythm Electrophysiol 2013;6:732–737. [DOI] [PubMed] [Google Scholar]
- 94. Lee H-C, Lu T, Weintraub NL, VanRollins M, Spector AA, Shibata EF. Effects of epoxyeicosatrienoic acids on the cardiac sodium channels in isolated rat ventricular myocytes. J Physiol 1999;519:153–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Wutzler A, Kestler C, Perrot A, Loehr L, Huemer M, Parwani AS, Attanasio P, Özcelik C, Schunck W-H, Gollasch M, Haverkamp W, Boldt L-H. Variations in the human soluble epoxide hydrolase gene and recurrence of atrial fibrillation after catheter ablation. Int J Cardiol 2013;168:3647–3651. [DOI] [PubMed] [Google Scholar]
- 96. Fatini C, Sticchi E, Genuardi M, Sofi F, Gensini F, Gori AM, Lenti M, Michelucci A, Abbate R, Gensini GF. Analysis of minK and eNOS genes as candidate loci for predisposition to non-valvular atrial fibrillation. Eur Heart J 2006;27:1712–1718. [DOI] [PubMed] [Google Scholar]
- 97. Shim J, Park JH, Lee J-Y, Uhm J-S, Joung B, Lee M-H, Ellinor PT, Pak H-N. eNOS3 genetic polymorphism is related to post-ablation early recurrence of atrial fibrillation. Yonsei Med J 2015;56:1244–1250. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 98. Wu G, Cheng M, Huang H, Yang B, Jiang H, Huang C. A variant of IL6R is associated with the recurrence of atrial fibrillation after catheter ablation in a Chinese Han population. PLos One 2014;9:e99623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Hu Y-F, Lee K-T, Wang H-H, Ueng K-C, Yeh H-I, Chao T-F, Liao J-N, Lin Y-J, Chang S-L, Lo L-W, Tuan T-C, Li C-H, Chung F-P, Hsu C-P, Chang H-H, Huang C-H, Chen S-A. The association between heme oxygenase-1 gene promoter polymorphism and the outcomes of catheter ablation of atrial fibrillation. PLos One 2013;8:e56440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Wang Q, Hu X, Li S, Wang X, Wang J, Zhang R, Sun J, Zhang P, Feng X, Li Y-G. Association of the angiotensinogen M235T polymorphism with recurrence after catheter ablation of acquired atrial fibrillation. J Renin Angiotensin Aldosterone Syst 2015;16:888–897. [DOI] [PubMed] [Google Scholar]
- 101. Wu H, Xu J, Chen S, Zhou G, Qi B, Wei Y, Hu E, Tang D, Chen G, Li H, Zhao L, Shi Y, Liu S. Association of SCN10A polymorphisms with the recurrence of atrial fibrillation after catheter ablation in a Chinese Han population. Sci Rep 2017;7:44003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Husser D, Ueberham L, Hindricks G, Büttner P, Ingram C, Weeke P, Shoemaker MB, Adams V, Arya A, Sommer P, Darbar D, Roden DM, Bollmann A. Rare variants in genes encoding the cardiac sodium channel and associated compounds and their impact on outcome of catheter ablation of atrial fibrillation. PLos One 2017;12:e0183690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Shoemaker MB, Bollmann A, Lubitz SA, Ueberham L, Saini H, Montgomery J, Edwards T, Yoneda Z, Sinner MF, Arya A, Sommer P, Delaney J, Goyal SK, Saavedra P, Kanagasundram A, Whalen SP, Roden DM, Hindricks G, Ellis CR, Ellinor PT, Darbar D, Husser D. Common genetic variants and response to atrial fibrillation ablation. Circ Arrhythm Electrophysiol 2015;8:296–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Choi EK, Park JH, Lee JY, Nam CM, Hwang MK, Uhm JS, Joung B, Ko YG, Lee MH, Lubitz SA, Ellinor PT, Pak HN. Korean atrial fibrillation (AF) network: genetic variants for AF do not predict ablation success. J Am Heart Assoc 2015;4:e002046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Choe W-S, Kang JH, Choi E-K, Shin SY, Lubitz SA, Ellinor PT, Oh S, Lim HE. A genetic risk score for atrial fibrillation predicts the response to catheter ablation. Korean Circ J 2019;49:338–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Amioka M, Nakano Y, Ochi H, Onohara Y, Sairaku A, Tokuyama T, Motoda C, Matsumura H, Tomomori S, Hironobe N, Okubo Y, Okamura S, Chayama K, Kihara Y. Ser96Ala genetic variant of the human histidine-rich calcium-binding protein is a genetic predictor of recurrence after catheter ablation in patients with paroxysmal atrial fibrillation. PLos One 2019;14:e0213208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Shoemaker MB, Husser D, Roselli C, Jazairi MA, Chrispin J, Kühne M, Neumann B, Knight S, Sun H, Mohanty S, Shaffer C, Thériault S, Rinke LL, Siland JE, Crawford DM, Ueberham L, Zardkoohi O, Büttner P, Geelhoed B, Blum S, Aeschbacher S, Smith JD, Wagoner DRV, Freudling R, Müller-Nurasyid M, Montgomery J, Yoneda Z, Wells Q, Issa T, Weeke P, Jacobs V, Gelder ICV, Hindricks G, Barnard J, Calkins H, Darbar D, Michaud G, Kääb S, Ellinor P, Natale A, Chung M, Nazarian S, Cutler MJ, Sinner MF, Conen D, Rienstra M, Bollmann A, Roden DM, Lubitz S. Genetic susceptibility for atrial fibrillation in patients undergoing atrial fibrillation ablation. Circ Arrhythm Electrophysiol 2020;13:e007676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Goyal SK, Wang L, Upender R, Darbar D, Monahan K. Severity of obstructive sleep apnea influences the effect of genotype on response to anti-arrhythmic drug therapy for atrial fibrillation. J Clin Sleep Med 2014;10:503–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Fry A, Littlejohns TJ, Sudlow C, Doherty N, Adamska L, Sprosen T, Collins R, Allen NE. Comparison of sociodemographic and health-related characteristics of UK Biobank participants with those of the general population. Am J Epidemiol 2017;186:1026–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Martin AR, Kanai M, Kamatani Y, Okada Y, Neale BM, Daly MJ. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat Genet 2019;51:584–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Svennberg E, Engdahl J, Al-Khalili F, Friberg L, Frykman V, Rosenqvist M. Mass screening for untreated atrial fibrillation. Circulation 2015;131:2176–2184. [DOI] [PubMed] [Google Scholar]
- 112. Khurshid S, Kartoun U, Ashburner JM, Trinquart L, Philippakis A, Khera AV, Ellinor PT, Ng K, Lubitz SA. Performance of atrial fibrillation risk prediction models in over 4 million individuals. Circ Arrhythm Electrophysiol 2021;14:e008997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Andrade JG, Champagne J, Dubuc M, Deyell MW, Verma A, Macle L, Leong-Sit P, Novak P, Badra-Verdu M, Sapp J, Mangat I, Khoo C, Steinberg C, Bennett MT, Tang ASL, Khairy P, Parkash R, Guerra P, Dyrda K, Rivard L, Racine N, Sterns L, Leather R, Seifer C, Jolly U, Raymond J-M, Roux J-F, Nault I, Sarrazin J-F, Ramanathan K, Cheung C, Fordyce C, McKinney J, Luong C, Rizkallah J, Angaran P, Ha A, Glover B, Skanes A, Gula L. Cryoballoon or radiofrequency ablation for atrial fibrillation assessed by continuous monitoring. Circulation 2019;140:1779–1788. [DOI] [PubMed] [Google Scholar]


