Table of contents

  • 1. Preamble 3319

  • 2. Introduction 3321

  •  2.1. What is new 3322

  • 3. Definitions and clinical impact 3326

  •  3.1. Definition and classification of AF 3326

  •  3.2. Diagnostic criteria for AF 3327

  •  3.3. Symptoms attributable to AF 3328

  •  3.4. Diagnostic evaluation of new AF 3328

  •  3.5. Adverse events associated with AF 3329

  •  3.6. Atrial flutter 3330

  • 4. Patient pathways and management of AF 3330

  •  4.1. Patient-centred, multidisciplinary AF management 3330

  •   4.1.1. The patient at the heart of care 3330

  •   4.1.2. Education and shared decision-making 3331

  •   4.1.3. Education of healthcare professionals 3332

  •   4.1.4. Inclusive management of AF 3332

  •  4.2. Principles of AF-CARE 3332

  • 5. [C] Comorbidity and risk factor management 3338

  •  5.1. Hypertension 3339

  •  5.2. Heart failure 3339

  •  5.3. Type 2 diabetes mellitus 3340

  •  5.4. Obesity 3340

  •  5.5. Obstructive sleep apnoea 3340

  •  5.6. Physical inactivity 3340

  •  5.7. Alcohol excess 3341

  • 6. [A] Avoid stroke and thromboembolism 3341

  •  6.1. Initiating oral anticoagulation 3341

  •   6.1.1. Decision support for anticoagulation in AF 3341

  •  6.2. Oral anticoagulants 3343

  •   6.2.1. Direct oral anticoagulants 3344

  •   6.2.2. Vitamin K antagonists 3345

  •   6.2.3. Clinical vs. device-detected subclinical AF 3345

  •  6.3. Antiplatelet drugs and combinations with anticoagulants 3346

  •  6.4. Residual ischaemic stroke risk despite anticoagulation 3346

  •  6.5. Percutaneous left atrial appendage occlusion 3346

  •  6.6. Surgical left atrial appendage occlusion 3347

  •  6.7. Bleeding risk 3348

  •   6.7.1. Assessment of bleeding risk 3348

  •   6.7.2. Management of bleeding on anticoagulant therapy 3348

  • 7. [R] Reduce symptoms by rate and rhythm control 3351

  •  7.1. Management of heart rate in patients with AF 3351

  •   7.1.1. Indications and target heart rate 3352

  •   7.1.2. Heart rate control in the acute setting 3352

  •   7.1.3. Long-term heart rate control 3352

  •   7.1.4. Atrioventricular node ablation and pacemaker implantation 3353

  •  7.2. Rhythm control strategies in patients with AF 3353

  •   7.2.1. General principles and anticoagulation 3353

  •   7.2.2. Electrical cardioversion 3356

  •   7.2.3. Pharmacological cardioversion 3356

  •   7.2.4. Antiarrhythmic drugs 3357

  •   7.2.5. Catheter ablation 3358

  •   7.2.6. Anticoagulation in patients undergoing catheter ablation 3359

  •   7.2.7. Endoscopic and hybrid AF ablation 3360

  •   7.2.8. AF ablation during cardiac surgery 3361

  •   7.2.9. Atrial tachycardia after pulmonary vein isolation 3361

  • 8. [E] Evaluation and dynamic reassessment 3361

  •  8.1. Implementation of dynamic care 3362

  •  8.2. Improving treatment adherence 3362

  •  8.3. Cardiac imaging 3362

  •  8.4. Patient-reported outcome measures 3363

  • 9. The AF-CARE pathway in specific clinical settings 3364

  •  9.1. AF-CARE in unstable patients 3364

  •  9.2. AF-CARE in acute and chronic coronary syndromes 3364

  •  9.3. AF-CARE in vascular disease 3366

  •  9.4. AF-CARE in acute stroke or intracranial haemorrhage 3366

  •   9.4.1. Management of acute ischaemic stroke 3366

  •   9.4.2. Introduction or re-introduction of anticoagulation after ischaemic stroke 3367

  •   9.4.3. Introduction or re-introduction of anticoagulation after haemorrhagic stroke 3367

  •  9.5. AF-CARE for trigger-induced AF 3367

  •  9.6. AF-CARE in post-operative patients 3368

  •  9.7. AF-CARE in embolic stroke of unknown source 3368

  •  9.8. AF-CARE during pregnancy 3369

  •  9.9. AF-CARE in congenital heart disease 3370

  •  9.10. AF-CARE in endocrine disorders 3370

  •  9.11. AF-CARE in inherited cardiomyopathies and primary arrhythmia syndromes 3370

  •  9.12. AF-CARE in cancer 3371

  •  9.13. AF-CARE in older, multimorbid, or frail patients 3371

  •  9.14. AF-CARE in atrial flutter 3371

  • 10. Screening and prevention of AF 3371

  •  10.1. Epidemiology of AF 3371

  •  10.2. Screening tools for AF 3372

  •  10.3. Screening strategies for AF 3373

  •   10.3.1. Single timepoint screening ‘snapshot’ 3374

  •   10.3.2. Prolonged screening 3374

  •  10.4. Factors associated with incident AF 3375

  •  10.5. Primary prevention of AF 3375

  •   10.5.1. Hypertension 3376

  •   10.5.2. Heart failure 3376

  •   10.5.3. Type 2 diabetes mellitus 3376

  •   10.5.4. Obesity 3376

  •   10.5.5. Sleep apnoea syndrome 3376

  •   10.5.6. Physical activity 3376

  •   10.5.7. Alcohol intake 3377

  • 11. Key messages 3377

  • 12. Gaps in evidence 3377

  • 13. ‘What to do’ and ‘What not to do’ messages from the guidelines 3379

  • 14. Evidence tables 3382

  • 15. Data availability statement 3382

  • 16. Author information 3382

  • 17. Appendix 3383

  • 18. References 3384

Tables of Recommendations

  • Recommendation Table 1 — Recommendations for the diagnosis of AF (see also Evidence Table 1) 3328

  • Recommendation Table 2 — Recommendations for symptom evaluation in patients with AF (see also Evidence Table 2) 3328

  • Recommendation Table 3 — Recommendations for diagnostic evaluation in patients with new AF (see also Evidence Table 3) 3328

  • Recommendation Table 4 — Recommendations for patient-centred care and education (see also Evidence Table 4) 3332

  • Recommendation Table 5 — Recommendations for comorbidity and risk factor management in AF (see also Evidence Table 5) 3339

  • Recommendation Table 6 — Recommendations to assess and manage thromboembolic risk in AF (see also Evidence Table 6) 3342

  • Recommendation Table 7 — Recommendations for oral anticoagulation in AF (see also Evidence Table 7) 3344

  • Recommendation Table 8 — Recommendations for combining antiplatelet drugs with anticoagulants for stroke prevention (see also Evidence Table 8) 3346

  • Recommendation Table 9 — Recommendations for thromboembolism despite anticoagulation (see also Evidence Table 9) 3346

  • Recommendation Table 10 — Recommendations for percutaneous left atrial appendage occlusion (see also Evidence Table 10) 3347

  • Recommendation Table 11 — Recommendations for surgical left atrial appendage occlusion (see also Evidence Table 11) 3348

  • Recommendation Table 12 — Recommendations for assessment of bleeding risk (see also Evidence Table 12) 3348

  • Recommendation Table 13 — Recommendations for management of bleeding in anticoagulated patients (see also Evidence Table 13) 3351

  • Recommendation Table 14 — Recommendations for heart rate control in patients with AF (see also Evidence Table 14) 3351

  • Recommendation Table 15 — Recommendations for general concepts in rhythm control (see also Evidence Table 15) 3355

  • Recommendation Table 16 — Recommendations for electrical cardioversion of AF (see also Evidence Table 16) 3356

  • Recommendation Table 17 — Recommendations for pharmacological cardioversion of AF (see also Evidence Table 17) 3356

  • Recommendation Table 18 — Recommendations for antiarrhythmic drugs for long-term maintenance of sinus rhythm (see also Evidence Table 18) 3358

  • Recommendation Table 19 — Recommendations for catheter ablation of AF (see also Evidence Table 19) 3359

  • Recommendation Table 20 — Recommendations for anticoagulation in patients undergoing catheter ablation (see also Evidence Table 20) 3360

  • Recommendation Table 21 — Recommendations for endoscopic and hybrid AF ablation (see also Evidence Table 21) 3360

  • Recommendation Table 22 — Recommendations for AF ablation during cardiac surgery (see also Evidence Table 22) 3361

  • Recommendation Table 23 — Recommendations to improve patient experience (see also Evidence Table 23) 3364

  • Recommendation Table 24 — Recommendations for patients with acute coronary syndromes or undergoing percutaneous intervention (see also Evidence Table 24) 3366

  • Recommendation Table 25 — Recommendations for trigger-induced AF (see also Evidence Table 25) 3368

  • Recommendation Table 26 — Recommendations for management of post-operative AF (see also Evidence Table 26) 3368

  • Recommendation Table 27 — Recommendations for patients with embolic stroke of unknown source (see also Evidence Table 27) 3369

  • Recommendation Table 28 — Recommendations for patients with AF during pregnancy (see also Evidence Table 28) 3369

  • Recommendation Table 29 — Recommendations for patients with AF and congenital heart disease (see also Evidence Table 29) 3370

  • Recommendation Table 30 — Recommendations for prevention of thromboembolism in atrial flutter (see also Evidence Table 30) 3371

  • Recommendation Table 31 — Recommendations for screening for AF (see also Evidence Table 31) 3374

  • Recommendation Table 32 — Recommendations for primary prevention of AF (see also Evidence Table 32) 3376

List of tables

  • Table 1 Classes of recommendations 3320

  • Table 2 Levels of evidence 3320

  • Table 3 New recommendations 3322

  • Table 4 Revised recommendations 3325

  • Table 5 Definitions and classifications for the temporal pattern of AF 3327

  • Table 6 Other clinical concepts relevant to AF 3327

  • Table 7 The modified European Heart Rhythm Association (mEHRA) symptom classification 3329

  • Table 8 Diagnostic work-up for patients with AF 3330

  • Table 9 Achieving patient-centred AF management 3331

  • Table 10 Updated definitions for the CHA2DS2-VA score 3342

  • Table 11 Recommended doses for direct oral anticoagulant therapy 3345

  • Table 12 Drugs for rate control in AF 3352

  • Table 13 Antiarrhythmic drugs for sinus rhythm restoration 3357

  • Table 14 Non-cardiac conditions associated with trigger-induced AF 3367

  • Table 15 Tools for AF screening 3373

  • Table 16 Factors associated with incident AF 3375

  • Table 17 ‘What to do’ and ‘what not to do’ 3379

List of figures

  • Figure 1 Impacts and outcomes associated with clinical AF. AF, atrial fibrillation 3329

  • Figure 2 Multidisciplinary approach to AF management 3331

  • Figure 3 Central illustration. Patient pathway for AF-CARE (see Figures 4, 5, 6, and 7 for the [R] pathways for first-diagnosed, paroxysmal, persistent and permanent AF) 3333

  • Figure 4 [R] Pathway for patients with first-diagnosed AF 3334

  • Figure 5 [R] Pathway for patients with paroxysmal AF 3335

  • Figure 6 [R] Pathway for patients with persistent AF 3336

  • Figure 7 [R] Pathway for patients with permanent AF 3337

  • Figure 8 Management of key comorbidities to reduce AF recurrence 3338

  • Figure 9 Common drug interactions with oral anticoagulants 3343

  • Figure 10 Modifying the risk of bleeding associated with OAC 3349

  • Figure 11 Management of oral anticoagulant-related bleeding in patients with AF 3350

  • Figure 12 Approaches for cardioversion in patients with AF 3354

  • Figure 13 Relevance of echocardiography in the AF-CARE pathway 3363

  • Figure 14 Antithrombotic therapy in patients with AF and acute or chronic coronary syndromes 3365

  • Figure 15 Non-invasive diagnostic methods for AF screening 3372

  • Figure 16 Approaches to screening for AF 3374

Abbreviations and acronyms

     
  • AAD

    Antiarrhythmic drugs

  •  
  • ACE

    Angiotensin-converting enzyme

  •  
  • ACEi

    Angiotensin-converting enzyme inhibitor

  •  
  • ACS

    Acute coronary syndromes

  •  
  • ACTIVE W

    Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events (trial)

  •  
  • AF

    Atrial fibrillation

  •  
  • AF-CARE

    Atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment

  •  
  • AFEQT

    Atrial Fibrillation Effect on QualiTy-of-Life (questionnaire)

  •  
  • AFFIRM

    Atrial Fibrillation Follow-up Investigation of Rhythm Management (trial)

  •  
  • AFL

    Atrial flutter

  •  
  • AFQLQ

    Atrial Fibrillation Quality of Life Questionnaire

  •  
  • AF-QoL

    Atrial Fibrillation Quality of Life (questionnaire)

  •  
  • AFSS

    Atrial Fibrillation Severity Scale

  •  
  • AI

    Artificial intelligence

  •  
  • APACHE-AF

    Apixaban After Anticoagulation-associated Intracerebral Haemorrhage in Patients With Atrial Fibrillation (trial)

  •  
  • APAF-CRT

    Ablate and Pace for Atrial Fibrillation—cardiac resynchronization therapy

  •  
  • ARB

    Angiotensin receptor blocker

  •  
  • ARTESiA

    Apixaban for the Reduction of Thromboembolism in Patients With Device-Detected Sub-Clinical Atrial Fibrillation (trial)

  •  
  • AT

    Atrial tachycardia

  •  
  • ATHENA

    A Placebo-Controlled, Double-Blind, Parallel Arm Trial to Assess the Efficacy of Dronedarone 400 mg twice daily for the Prevention of Cardiovascular Hospitalization or Death from Any Cause in Patients with Atrial Fibrillation/Atrial Flutter (trial)

  •  
  • AUGUSTUS

    An open-label, 2 × 2 factorial, randomized controlled, clinical trial to evaluate the safety of apixaban vs. vitamin k antagonist and aspirin vs. aspirin placebo in patients with atrial fibrillation and acute coronary syndrome or percutaneous coronary intervention

  •  
  • AVERROES

    Apixaban Versus Acetylsalicylic Acid to Prevent Stroke in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment (trial)

  •  
  • AVN

    Atrioventricular node

  •  
  • b.p.m.

    Beats per minute

  •  
  • BMI

    Body mass index

  •  
  • BNP

    B-type natriuretic peptide

  •  
  • BP

    Blood pressure

  •  
  • C2HEST

    Coronary artery disease or chronic obstructive pulmonary disease (1 point each); hypertension (1 point); elderly (age ≥75 years, 2 points); systolic heart failure (2 points); thyroid disease (hyperthyroidism, 1 point)

  •  
  • CABANA

    Catheter Ablation versus Anti-arrhythmic Drug Therapy for Atrial Fibrillation (trial)

  •  
  • CAD

    Coronary artery disease

  •  
  • CASTLE-AF

    Catheter Ablation versus Standard Conventional Treatment in Patients With Left Ventricle (LV) Dysfunction and AF (trial)

  •  
  • CASTLE-HTx

    Catheter Ablation for Atrial Fibrillation in Patients With End-Stage Heart Failure and Eligibility for Heart Transplantation (trial)

  •  
  • CCS

    Chronic coronary syndrome

  •  
  • CHADS2

    Congestive heart failure, hypertension, age >75 years, diabetes; previous stroke (2 points)

  •  
  • CHA2DS2-VA

    Congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years (score)

  •  
  • CHA2DS2-VASc

    Congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke or TIA or thromboembolism (2 points), vascular disease, age 65–74 years, sex category

  •  
  • CKD

    Chronic kidney disease

  •  
  • CMR

    Cardiac magnetic resonance

  •  
  • COMPASS

    Cardiovascular Outcomes for People Using Anticoagulation Strategies (trial)

  •  
  • CPAP

    Continuous positive airway pressure

  •  
  • CrCl

    Creatinine clearance

  •  
  • CRT

    Cardiac resynchronization therapy

  •  
  • CT

    Computed tomography

  •  
  • CTA

    Computed tomography angiography

  •  
  • CTI

    Cavo-tricuspid isthmus

  •  
  • DAPT

    Dual antiplatelet therapy

  •  
  • DOAC

    Direct oral anticoagulant

  •  
  • EAST-AFNET 4

    Early treatment of Atrial fibrillation for Stroke prevention Trial

  •  
  • ECG

    Electrocardiogram

  •  
  • ECV

    Electrical cardioversion

  •  
  • EHRA

    European Heart Rhythm Association

  •  
  • ELAN

    Early versus Late initiation of direct oral Anticoagulants in post-ischaemic stroke patients with atrial fibrillatioN (trial)

  •  
  • ESUS

    Embolic stroke of undetermined source

  •  
  • FFP

    Fresh frozen plasma

  •  
  • GI

    Gastrointestinal

  •  
  • GWAS

    Genome-wide association studies

  •  
  • HAS-BLED

    Hypertension, Abnormal renal/liver function, Stroke, Bleeding history or predisposition, Labile international normalized ratio, Elderly (>65 years), Drugs/alcohol concomitantly (score)

  •  
  • HAVOC

    Hypertension, age, valvular heart disease, peripheral vascular disease, obesity, congestive heart failure, and coronary artery disease

  •  
  • HbA1c

    Haemoglobin A1c (glycated or glycosylated haemoglobin)

  •  
  • HCM

    Hypertrophic cardiomyopathy

  •  
  • HF

    Heart failure

  •  
  • HFmrEF

    Heart failure with mildly reduced ejection fraction

  •  
  • HFpEF

    Heart failure with preserved ejection fraction

  •  
  • HFrEF

    Heart failure with reduced ejection fraction

  •  
  • HR

    Hazard ratio

  •  
  • i.v.

    Intravenous

  •  
  • ICH

    Intracranial haemorrhage

  •  
  • ICHOM

    International Consortium for Health Outcomes Measurement

  •  
  • IMT

    Intima-media thickness

  •  
  • INR

    International normalized ratio (of prothrombin time)

  •  
  • LA

    Left atrium

  •  
  • LAA

    Left atrial appendage

  •  
  • LAAO

    Left atrial appendage occlusion

  •  
  • LAAOS III

    Left Atrial Appendage Occlusion Study

  •  
  • LEGACY

    Long-Term Effect of Goal directed weight management on Atrial Fibrillation Cohort: a 5 Year follow-up study

  •  
  • LMWH

    Low molecular weight heparin

  •  
  • LOOP

    Atrial Fibrillation Detected by Continuous ECG Monitoring (trial)

  •  
  • LV

    Left ventricle

  •  
  • LVEF

    Left ventricular ejection fraction

  •  
  • LVH

    Left ventricular hypertrophy

  •  
  • mEHRA

    Modified European Heart Rhythm Association score

  •  
  • MI

    Myocardial infarction

  •  
  • MRI

    Magnetic resonance imaging

  •  
  • NOAH

    Non-vitamin K Antagonist Oral Anticoagulants in Patients With Atrial High Rate Episodes (trial)

  •  
  • NSAID

    Non-steroidal anti-inflammatory drug

  •  
  • NT-proBNP

    N-terminal pro-B-type natriuretic peptide

  •  
  • NYHA

    New York Heart Association

  •  
  • OAC

    Oral anticoagulant(s)

  •  
  • OR

    Odds ratio

  •  
  • OSA

    Obstructive sleep apnoea

  •  
  • PAD

    Peripheral arterial disease

  •  
  • PCC

    Prothrombin complex concentrate

  •  
  • PCI

    Percutaneous intervention

  •  
  • PFO

    Patent foramen ovale

  •  
  • POAF

    Post-operative atrial fibrillation

  •  
  • PPG

    Photoplethysmography

  •  
  • PROM

    Patient-reported outcome measure

  •  
  • PVD

    Peripheral vascular disease

  •  
  • PVI

    Pulmonary vein isolation

  •  
  • QLAF

    Quality of Life in Atrial Fibrillation (questionnaire)

  •  
  • QRS

    Q wave, R wave, and S wave, the ‘QRS complex’ represents ventricular depolarization

  •  
  • RACE 7 ACWAS

    Rate Control versus Electrical Cardioversion Trial 7—Acute Cardioversion versus Wait and See (trial)

  •  
  • RACE I

    RAte Control versus Electrical cardioversion study

  •  
  • RACE II

    Rate Control Efficacy in Permanent Atrial Fibrillation (trial)

  •  
  • RACE 3

    Routine versus Aggressive upstream rhythm Control for prevention of Early AF in heart failure (trial)

  •  
  • RACE 4

    IntegRAted Chronic Care Program at Specialized AF Clinic Versus Usual CarE in Patients with Atrial Fibrillation (trial)

  •  
  • RATE-AF

    RAte control Therapy Evaluation in permanent Atrial Fibrillation (trial)

  •  
  • RCT

    Randomized controlled trial

  •  
  • RR

    Relative risk

  •  
  • SAVE

    Sleep Apnea cardioVascular Endpoints (trial)

  •  
  • SBP

    Systolic blood pressure

  •  
  • SGLT2

    Sodium-glucose cotransporter-2

  •  
  • SIC-AF

    Successful Intravenous Cardioversion for Atrial Fibrillation

  •  
  • SORT-AF

    Supervised Obesity Reduction Trial for AF Ablation Patients (trial)

  •  
  • SoSTART

    Start or STop Anticoagulants Randomised Trial

  •  
  • SR

    Sinus rhythm

  •  
  • STEEER-AF

    Stroke prevention and rhythm control Therapy: Evaluation of an Educational programme of the European Society of Cardiology in a cluster-Randomised trial in patients with Atrial Fibrillation (trial)

  •  
  • STEMI

    ST-segment elevation myocardial infarction

  •  
  • STROKESTOP

    Systematic ECG Screening for Atrial Fibrillation Among 75 Year Old Subjects in the Region of Stockholm and Halland, Sweden (trial)

  •  
  • TE

    Thromboembolism

  •  
  • TIA

    Transient ischaemic attack

  •  
  • TIMING

    Timing of Oral Anticoagulant Therapy in Acute Ischemic Stroke With Atrial Fibrillation (trial)

  •  
  • TOE

    Transoesophageal echocardiography

  •  
  • TSH

    Thyroid-stimulating hormone

  •  
  • TTE

    Transthoracic echocardiogram

  •  
  • TTR

    Time in therapeutic range

  •  
  • UFH

    Unfractionated heparin

  •  
  • VKA

    Vitamin K antagonist

1. Preamble

Guidelines evaluate and summarize available evidence with the aim of assisting health professionals in proposing the best diagnostic or therapeutic approach for an individual patient with a given condition. Guidelines are intended for use by health professionals and the European Society of Cardiology (ESC) makes its Guidelines freely available.

ESC Guidelines do not override the individual responsibility of health professionals to make appropriate and accurate decisions in consideration of each patient's health condition and in consultation with that patient or the patient's caregiver where appropriate and/or necessary. It is also the health professional's responsibility to verify the rules and regulations applicable in each country to drugs and devices at the time of prescription and to respect the ethical rules of their profession.

ESC Guidelines represent the official position of the ESC on a given topic and are regularly updated when warranted by new evidence. ESC Policies and Procedures for formulating and issuing ESC Guidelines can be found on the ESC website (https://www.escardio.org/Guidelines/Clinical-Practice-Guidelines/Guidelines-development/Writing-ESC-Guidelines). This guideline updates and replaces the previous version from 2020.

The Members of this task force were selected by the ESC to include professionals involved with the medical care of patients with this pathology as well as patient representatives and methodologists. The selection procedure included an open call for authors and aimed to include members from across the whole of the ESC region and from relevant ESC Subspecialty Communities. Consideration was given to diversity and inclusion, notably with respect to gender and country of origin. The task force performed a critical review and evaluation of the published literature on diagnostic and therapeutic approaches including assessment of the risk–benefit ratio. The strength of every recommendation and the level of evidence supporting them were weighed and scored according to predefined scales as outlined in Tables 1 and 2 below. Patient-reported outcome measures (PROMs) and patient-reported experience measures were also evaluated as the basis for recommendations and/or discussion in these guidelines. The task force followed ESC voting procedures and all approved recommendations were subject to a vote and achieved at least 75% agreement among voting members. Members of the task force with declared interests on specific topics were asked to abstain from voting on related recommendations.

The experts of the writing and reviewing panels provided declaration of interest forms for all relationships that might be perceived as real or potential sources of conflicts of interest. Their declarations of interest were reviewed according to the ESC declaration of interest rules which can be found on the ESC website (http://www.escardio.org/guidelines) and have been compiled in a report published in a supplementary document with the guidelines. Funding for the development of ESC Guidelines is derived entirely from the ESC with no involvement of the healthcare industry.

Table 1

Classes of recommendations

Table 1

Classes of recommendations

Table 2

Levels of evidence

Table 2

Levels of evidence

The ESC Clinical Practice Guidelines (CPG) Committee supervises and co-ordinates the preparation of new guidelines and is responsible for the approval process. In addition to review by the CPG Committee, ESC Guidelines undergo multiple rounds of double-blind peer review by external experts, including members from across the whole of the ESC region, all National Cardiac Societies of the ESC and from relevant ESC Subspecialty Communities. After appropriate revisions, the guidelines are signed off by all the experts in the task force. The finalized document is signed off by the CPG Committee for publication in the European Heart Journal.

ESC Guidelines are based on analyses of published evidence, chiefly on clinical trials and meta-analyses of trials, but potentially including other types of studies. Evidence tables summarizing key information from relevant studies are generated early in the guideline development process to facilitate the formulation of recommendations, to enhance comprehension of recommendations after publication, and reinforce transparency in the guidelines development process. The tables are published in their own section of ESC Guidelines and reference specific recommendation tables.

Off-label use of medication may be presented in this guideline if a sufficient level of evidence shows that it can be considered medically appropriate for a given condition. However, the final decisions concerning an individual patient must be made by the responsible health professional giving special consideration to:

  • The specific situation of the patient. Unless otherwise provided for by national regulations, off-label use of medication should be limited to situations where it is in the patient's interest with regard to the quality, safety, and efficacy of care, and only after the patient has been informed and has provided consent.

  • Country-specific health regulations, indications by governmental drug regulatory agencies, and the ethical rules to which health professionals are subject, where applicable.

2. Introduction

Atrial fibrillation (AF) is one of the most commonly encountered heart conditions, with a broad impact on all health services across primary and secondary care. The prevalence of AF is expected to double in the next few decades as a consequence of the ageing population, an increasing burden of comorbidities, improved awareness, and new technologies for detection.

The effects of AF are variable across individual patients; however, morbidity from AF remains highly concerning. Patients with AF can suffer from a variety of symptoms and poor quality of life. Stroke and heart failure as consequences of AF are now well appreciated by healthcare professionals, but AF is also linked to a range of other thromboembolic outcomes. These include subclinical cerebral damage (potentially leading to vascular dementia), and thromboembolism to every other organ, all of which contribute to the higher risk of mortality associated with AF.

The typical drivers of AF onset and progression are a range of comorbidities and associated risk factors. To achieve optimal care for patients with AF, it is now widely accepted that these comorbidities and risk factors must be managed early and in a dynamic way. Failure to do so contributes to recurrent cycles of AF, treatment failure, poor patient outcomes, and a waste of healthcare resources. In this iteration of the European Society of Cardiology (ESC) practice guidelines on AF, the task force has consolidated and evolved past approaches to develop the AF-CARE framework (Atrial Fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment). Comorbidities and risk factors is placed as the initial and central component of patient management. This should be considered first as it applies to all patients with AF, regardless of their thromboembolic risk factors or any symptoms that might warrant intervention. This is followed by considering how best to [A] avoid stroke and thromboembolism, and then the options available to reduce symptoms, and in some cases improve prognosis, through [R] rate and rhythm control. [E] Evaluation and reassessment should be individualized for every patient, with a dynamic approach that accounts for how AF and its associated conditions change over time.

Patient empowerment is critical in any long-term medical problem to achieve better outcomes, encourage adherence, and to seek timely guidance on changes in clinical status. A patient-centred, shared decision-making approach will facilitate the choice of management that suits each individual patient, particularly in AF where some therapies and interventions improve clinical outcomes, and others are focused on addressing symptoms and quality of life. Education and awareness are essential, not only for patients but also healthcare professionals in order to constrain the impact of AF on patients and healthcare services.

With this in mind, the task force have created a range of patient pathways that cover the major aspects of AF-CARE. At present, these remain based on the time-orientated classification of AF (first-diagnosed, paroxysmal, persistent, and permanent), but ongoing research may allow for pathology-based classifications and a future of personalized medicine. Clinical practice guidelines can only cover common scenarios with an evidence base, and so there remains a need for healthcare professionals to care for patients within a local multidisciplinary team. While guideline-adherent care has repeatedly been shown to improve patient outcomes, the actual implementation of guidelines is often poor in many healthcare settings. This has been demonstrated in the ESC's first randomized controlled trial (RCT), STEEER-AF (Stroke prevention and rhythm control Therapy: Evaluation of an Educational programme of the European Society of Cardiology in a cluster-Randomised trial in patients with Atrial Fibrillation), which has sought to improve guideline adherence in parallel to guideline production. The task force developing the 2024 AF Guidelines have made implementation a key goal by focusing on the underpinning evidence and using a consistent writing style for each recommendation (the intervention proposed, the population it should be applied to, and the potential value to the patient, followed by any exceptions). Tables 3 and 4 below outline new recommendations and those with important revisions. These initiatives have been designed to make the 2024 ESC Guidelines for the management of AF easier to read, follow, and implement, with the aim of improving the lives of patients with AF. A patient version of these guidelines is also available at http://www.escardio.org/Guidelines/guidelines-for-patients.

2.1. What is new

Table 3

New recommendations

Table 3

New recommendations

Table 4

Revised recommendations

Table 4

Revised recommendations

3. Definitions and clinical impact

3.1. Definition and classification of AF

Atrial fibrillation is one of the most common heart rhythm disorders. A supraventricular arrhythmia with uncoordinated atrial activation, AF results in a loss of effective atrial contraction (see Supplementary data online for pathophysiology). AF is reflected on the surface electrocardiogram (ECG) by the absence of discernible and regular P waves, and irregular activation of the ventricles. This results in no specific pattern to RR intervals, in the absence of an atrioventricular block. The definition of AF by temporal pattern is presented in Table 5. It should be noted that these categories reflect observed episodes of AF and do not suggest the underlying pathophysiological process. Some patients may progress consecutively through these categories, while others may need periodic reclassification due to their individual clinical status. Over time, some patients with AF develop atrial and ventricular damage, which can make attempts at rhythm control futile. For this reason, or when patients and physicians make a joint decision for rate control, AF is classified as permanent (the most common ‘type’ of AF in historical registries).1 Despite many limitations, this task force have retained this temporal approach because most trials in patients with AF have used these definitions. Classifying AF by underlying drivers could inform management, but the evidence in support of the clinical use of such classification is currently lacking.

Table 5

Definitions and classifications for the temporal pattern of AF

Temporal classificationDefinition
First-diagnosed AFAF that has not been diagnosed before, regardless of symptom status, temporal pattern, or duration.
Paroxysmal AFAF which terminates spontaneously within 7 days or with the assistance of an intervention. Evidence suggests that most self-terminating paroxysms last <48 h.2
Persistent AFAF episodes which are not self-terminating. Many intervention trials have used 7 days as a cut-off for defining persistent AF.3,4 Long-standing persistent AF is arbitrarily defined as continuous AF of at least 12 months' duration but where rhythm control is still a treatment option in selected patients, distinguishing it from permanent AF.
Permanent AFAF for which no further attempts at restoration of sinus rhythm are planned, after a shared decision between the patient and physician.
Temporal classificationDefinition
First-diagnosed AFAF that has not been diagnosed before, regardless of symptom status, temporal pattern, or duration.
Paroxysmal AFAF which terminates spontaneously within 7 days or with the assistance of an intervention. Evidence suggests that most self-terminating paroxysms last <48 h.2
Persistent AFAF episodes which are not self-terminating. Many intervention trials have used 7 days as a cut-off for defining persistent AF.3,4 Long-standing persistent AF is arbitrarily defined as continuous AF of at least 12 months' duration but where rhythm control is still a treatment option in selected patients, distinguishing it from permanent AF.
Permanent AFAF for which no further attempts at restoration of sinus rhythm are planned, after a shared decision between the patient and physician.

AF, atrial fibrillation.

© ESC 2024
Table 5

Definitions and classifications for the temporal pattern of AF

Temporal classificationDefinition
First-diagnosed AFAF that has not been diagnosed before, regardless of symptom status, temporal pattern, or duration.
Paroxysmal AFAF which terminates spontaneously within 7 days or with the assistance of an intervention. Evidence suggests that most self-terminating paroxysms last <48 h.2
Persistent AFAF episodes which are not self-terminating. Many intervention trials have used 7 days as a cut-off for defining persistent AF.3,4 Long-standing persistent AF is arbitrarily defined as continuous AF of at least 12 months' duration but where rhythm control is still a treatment option in selected patients, distinguishing it from permanent AF.
Permanent AFAF for which no further attempts at restoration of sinus rhythm are planned, after a shared decision between the patient and physician.
Temporal classificationDefinition
First-diagnosed AFAF that has not been diagnosed before, regardless of symptom status, temporal pattern, or duration.
Paroxysmal AFAF which terminates spontaneously within 7 days or with the assistance of an intervention. Evidence suggests that most self-terminating paroxysms last <48 h.2
Persistent AFAF episodes which are not self-terminating. Many intervention trials have used 7 days as a cut-off for defining persistent AF.3,4 Long-standing persistent AF is arbitrarily defined as continuous AF of at least 12 months' duration but where rhythm control is still a treatment option in selected patients, distinguishing it from permanent AF.
Permanent AFAF for which no further attempts at restoration of sinus rhythm are planned, after a shared decision between the patient and physician.

AF, atrial fibrillation.

© ESC 2024

Several other classifications have been applied to patients with AF, many of which have limited evidence to support them. The definition of AF is a developing field and ongoing research may allow for pathology-based strategies that could facilitate personalized management in the future. Table 6 presents some commonly used concepts in current clinical practice. Due to the lack of supporting evidence (particularly for the time periods stated), this task force have edited and updated these definitions by consensus.

Table 6

Other clinical concepts relevant to AF

Clinical conceptDefinition
Clinical AFSymptomatic or asymptomatic AF that is clearly documented by an ECG (12-lead ECG or other ECG devices). The minimum duration to establish the diagnosis of clinical AF for ambulatory ECG is not clear and depends on the clinical context. Periods of 30 s or more may indicate clinical concern, and trigger further monitoring or risk stratification for thromboembolism.
Device-detected subclinical AFDevice-detected subclinical AF refers to asymptomatic episodes of AF detected on continuous monitoring devices. These devices include implanted cardiac electronic devices, for which most atrial high-rate episodesa may be AF, as well as consumer-based wearable monitors. Confirmation is needed by a competent professional reviewing intracardiac electrograms or an ECG-recorded rhythm.5,6 Device-detected subclinical AF is a predictor of future clinical AF.7
AF burdenThe overall time spent in AF during a clearly specified and reported period of monitoring, expressed as a percentage of time.
Recent-onset AFThere is accumulating data on the value of the term recent-onset AF in decision-making for acute pharmacological or electrical cardioversion of AF. The cut-off time interval to define this entity has not yet been established.8–10
Trigger-induced AFNew AF episode in close proximity to a precipitating and potentially reversible factor.11–14
Early AFThe time since diagnosis that qualifies for early AF is dissociated from any underlying atrial cardiomyopathy and is not well defined, broadly ranging from 3 to 24 months.15–17 The definition of early AF also does not necessarily determine early timing of intervention.
Self-terminating AFParoxysmal AF which terminates spontaneously.2 This definition may be of value for decisions on acute rhythm control taken jointly by the patient and healthcare provider.
Non-self-terminating AFAtrial fibrillation which does not terminate spontaneously and, if needed, termination can be achieved only with an intervention.
Atrial cardiomyopathyA combination of structural, electrical, or functional changes in the atria that leads to clinical impact (e.g. progression/recurrence of AF, limited effectiveness of AF therapy, and/or development of heart failure).18,19 Atrial cardiomyopathy includes inflammatory and prothrombotic remodelling of the atria, neurohormonal activation (thereby affecting the ventricles), and fibrosis of myocardial tissue.20
Clinical conceptDefinition
Clinical AFSymptomatic or asymptomatic AF that is clearly documented by an ECG (12-lead ECG or other ECG devices). The minimum duration to establish the diagnosis of clinical AF for ambulatory ECG is not clear and depends on the clinical context. Periods of 30 s or more may indicate clinical concern, and trigger further monitoring or risk stratification for thromboembolism.
Device-detected subclinical AFDevice-detected subclinical AF refers to asymptomatic episodes of AF detected on continuous monitoring devices. These devices include implanted cardiac electronic devices, for which most atrial high-rate episodesa may be AF, as well as consumer-based wearable monitors. Confirmation is needed by a competent professional reviewing intracardiac electrograms or an ECG-recorded rhythm.5,6 Device-detected subclinical AF is a predictor of future clinical AF.7
AF burdenThe overall time spent in AF during a clearly specified and reported period of monitoring, expressed as a percentage of time.
Recent-onset AFThere is accumulating data on the value of the term recent-onset AF in decision-making for acute pharmacological or electrical cardioversion of AF. The cut-off time interval to define this entity has not yet been established.8–10
Trigger-induced AFNew AF episode in close proximity to a precipitating and potentially reversible factor.11–14
Early AFThe time since diagnosis that qualifies for early AF is dissociated from any underlying atrial cardiomyopathy and is not well defined, broadly ranging from 3 to 24 months.15–17 The definition of early AF also does not necessarily determine early timing of intervention.
Self-terminating AFParoxysmal AF which terminates spontaneously.2 This definition may be of value for decisions on acute rhythm control taken jointly by the patient and healthcare provider.
Non-self-terminating AFAtrial fibrillation which does not terminate spontaneously and, if needed, termination can be achieved only with an intervention.
Atrial cardiomyopathyA combination of structural, electrical, or functional changes in the atria that leads to clinical impact (e.g. progression/recurrence of AF, limited effectiveness of AF therapy, and/or development of heart failure).18,19 Atrial cardiomyopathy includes inflammatory and prothrombotic remodelling of the atria, neurohormonal activation (thereby affecting the ventricles), and fibrosis of myocardial tissue.20

AF, atrial fibrillation; b.p.m., beats per minute; ECG, electrocardiogram.

aAtrial high-rate episodes are defined as episodes generally lasting more than 5 min with an atrial lead rate ≥170 b.p.m.,7,21–24 detected by implanted cardiac devices that allow for automated continuous monitoring and storage of atrial rhythm. Atrial high-rate episodes need to be visually inspected because some may be electrical artefacts or false positives.

© ESC 2024
Table 6

Other clinical concepts relevant to AF

Clinical conceptDefinition
Clinical AFSymptomatic or asymptomatic AF that is clearly documented by an ECG (12-lead ECG or other ECG devices). The minimum duration to establish the diagnosis of clinical AF for ambulatory ECG is not clear and depends on the clinical context. Periods of 30 s or more may indicate clinical concern, and trigger further monitoring or risk stratification for thromboembolism.
Device-detected subclinical AFDevice-detected subclinical AF refers to asymptomatic episodes of AF detected on continuous monitoring devices. These devices include implanted cardiac electronic devices, for which most atrial high-rate episodesa may be AF, as well as consumer-based wearable monitors. Confirmation is needed by a competent professional reviewing intracardiac electrograms or an ECG-recorded rhythm.5,6 Device-detected subclinical AF is a predictor of future clinical AF.7
AF burdenThe overall time spent in AF during a clearly specified and reported period of monitoring, expressed as a percentage of time.
Recent-onset AFThere is accumulating data on the value of the term recent-onset AF in decision-making for acute pharmacological or electrical cardioversion of AF. The cut-off time interval to define this entity has not yet been established.8–10
Trigger-induced AFNew AF episode in close proximity to a precipitating and potentially reversible factor.11–14
Early AFThe time since diagnosis that qualifies for early AF is dissociated from any underlying atrial cardiomyopathy and is not well defined, broadly ranging from 3 to 24 months.15–17 The definition of early AF also does not necessarily determine early timing of intervention.
Self-terminating AFParoxysmal AF which terminates spontaneously.2 This definition may be of value for decisions on acute rhythm control taken jointly by the patient and healthcare provider.
Non-self-terminating AFAtrial fibrillation which does not terminate spontaneously and, if needed, termination can be achieved only with an intervention.
Atrial cardiomyopathyA combination of structural, electrical, or functional changes in the atria that leads to clinical impact (e.g. progression/recurrence of AF, limited effectiveness of AF therapy, and/or development of heart failure).18,19 Atrial cardiomyopathy includes inflammatory and prothrombotic remodelling of the atria, neurohormonal activation (thereby affecting the ventricles), and fibrosis of myocardial tissue.20
Clinical conceptDefinition
Clinical AFSymptomatic or asymptomatic AF that is clearly documented by an ECG (12-lead ECG or other ECG devices). The minimum duration to establish the diagnosis of clinical AF for ambulatory ECG is not clear and depends on the clinical context. Periods of 30 s or more may indicate clinical concern, and trigger further monitoring or risk stratification for thromboembolism.
Device-detected subclinical AFDevice-detected subclinical AF refers to asymptomatic episodes of AF detected on continuous monitoring devices. These devices include implanted cardiac electronic devices, for which most atrial high-rate episodesa may be AF, as well as consumer-based wearable monitors. Confirmation is needed by a competent professional reviewing intracardiac electrograms or an ECG-recorded rhythm.5,6 Device-detected subclinical AF is a predictor of future clinical AF.7
AF burdenThe overall time spent in AF during a clearly specified and reported period of monitoring, expressed as a percentage of time.
Recent-onset AFThere is accumulating data on the value of the term recent-onset AF in decision-making for acute pharmacological or electrical cardioversion of AF. The cut-off time interval to define this entity has not yet been established.8–10
Trigger-induced AFNew AF episode in close proximity to a precipitating and potentially reversible factor.11–14
Early AFThe time since diagnosis that qualifies for early AF is dissociated from any underlying atrial cardiomyopathy and is not well defined, broadly ranging from 3 to 24 months.15–17 The definition of early AF also does not necessarily determine early timing of intervention.
Self-terminating AFParoxysmal AF which terminates spontaneously.2 This definition may be of value for decisions on acute rhythm control taken jointly by the patient and healthcare provider.
Non-self-terminating AFAtrial fibrillation which does not terminate spontaneously and, if needed, termination can be achieved only with an intervention.
Atrial cardiomyopathyA combination of structural, electrical, or functional changes in the atria that leads to clinical impact (e.g. progression/recurrence of AF, limited effectiveness of AF therapy, and/or development of heart failure).18,19 Atrial cardiomyopathy includes inflammatory and prothrombotic remodelling of the atria, neurohormonal activation (thereby affecting the ventricles), and fibrosis of myocardial tissue.20

AF, atrial fibrillation; b.p.m., beats per minute; ECG, electrocardiogram.

aAtrial high-rate episodes are defined as episodes generally lasting more than 5 min with an atrial lead rate ≥170 b.p.m.,7,21–24 detected by implanted cardiac devices that allow for automated continuous monitoring and storage of atrial rhythm. Atrial high-rate episodes need to be visually inspected because some may be electrical artefacts or false positives.

© ESC 2024

3.2. Diagnostic criteria for AF

In many patients, the diagnosis of AF is straightforward, e.g. typical symptoms associated with characteristic features on a standard 12-lead ECG that indicate the need for AF management. Diagnosis becomes more challenging in the context of asymptomatic episodes or AF detected on longer-term monitoring devices, particularly those that do not provide an ECG (see Section 10). To guard against inappropriate diagnosis of AF, this task force continues to recommend that ECG documentation is required to initiate risk stratification and AF management. In current practice, ECG confirmation can include multiple options: not only where AF persists across a standard 12-lead ECG, but also single- and multiple-lead devices that provide an ECG (see Supplementary data online, Additional Evidence  Table S1). This does not include non-ECG wearables and other devices that typically use photoplethysmography. Note that many pivotal AF trials required two or more ECGs documenting AF, or an established AF diagnosis before randomization.25–29 The time period of AF required for diagnosis on monitoring devices is not clear cut. A standard 12-lead ECG measures 10 s, while 30 s or more on single-lead or multiple-lead ECG devices has generally been the consensus opinion, albeit with limited evidence.

Recommendation Table 1

Recommendations for the diagnosis of AF (see also Evidence Table 1)

Recommendation Table 1

Recommendations for the diagnosis of AF (see also Evidence Table 1)

3.3. Symptoms attributable to AF

Symptoms related to episodes of AF are variable and broad, and not just typical palpitations (Figure 1). Asymptomatic episodes of AF can occur,30 although 90% of patients with AF describe symptoms with variable severity.31 Even in symptomatic patients, some episodes of AF may remain asymptomatic.32,33 The presence or absence of symptoms is not related to incident stroke, systemic embolism, or mortality.34 However, symptoms do impact on patient quality of life.35,36 Cardiac-specific AF symptoms such as palpitations are less common than non-specific symptoms such as fatigue, but they significantly impair quality of life.36,37 Although women are often underrepresented in clinical trials of AF,38–40 the available literature suggests that women with AF appear to be more symptomatic and have poorer quality of life.41,42 Patients with AF report a higher burden of anxiety and severity of depression (odds ratio [OR], 1.08; 95% confidence interval [CI], 1.02–1.15; P = .009) as compared with the general population,43,44 with higher prevalence of these symptoms in women with AF.45

Impacts and outcomes associated with clinical AF. AF, atrial fibrillation.
Figure 1

Impacts and outcomes associated with clinical AF. AF, atrial fibrillation.

Assessment of AF-related symptoms should be recorded initially, after a change in treatment, or before and after intervention. The modified European Heart Rhythm Association score (mEHRA) symptom classification (Table 7) is similar to the New York Heart Association (NYHA) functional class for heart failure. It correlates with quality of life scores in clinical trials, is associated with clinical progress and events, and may be a valuable starting point in routine practice to assess the burden and impact of symptoms together with the patient.46–48 Note that symptoms may also relate to associated comorbidities and not just the AF component. The patient-related effects of symptoms from AF over time can alternatively be evaluated using patient-reported outcome measures (see Section 8.4).

Table 7

The modified European Heart Rhythm Association (mEHRA) symptom classification

ScoreSymptomsDescription
1NoneAF does not cause any symptoms
2aMildNormal daily activity not affected by symptoms related to AF
2bModerateNormal daily activity not affected by symptoms related to AF, but patient troubled by symptoms
3SevereNormal daily activity affected by symptoms related to AF
4DisablingNormal daily activity discontinued
ScoreSymptomsDescription
1NoneAF does not cause any symptoms
2aMildNormal daily activity not affected by symptoms related to AF
2bModerateNormal daily activity not affected by symptoms related to AF, but patient troubled by symptoms
3SevereNormal daily activity affected by symptoms related to AF
4DisablingNormal daily activity discontinued

AF, atrial fibrillation.

© ESC 2024
Table 7

The modified European Heart Rhythm Association (mEHRA) symptom classification

ScoreSymptomsDescription
1NoneAF does not cause any symptoms
2aMildNormal daily activity not affected by symptoms related to AF
2bModerateNormal daily activity not affected by symptoms related to AF, but patient troubled by symptoms
3SevereNormal daily activity affected by symptoms related to AF
4DisablingNormal daily activity discontinued
ScoreSymptomsDescription
1NoneAF does not cause any symptoms
2aMildNormal daily activity not affected by symptoms related to AF
2bModerateNormal daily activity not affected by symptoms related to AF, but patient troubled by symptoms
3SevereNormal daily activity affected by symptoms related to AF
4DisablingNormal daily activity discontinued

AF, atrial fibrillation.

© ESC 2024
Recommendation Table 2

Recommendations for symptom evaluation in patients with AF (see also Evidence Table 2)

Recommendation Table 2

Recommendations for symptom evaluation in patients with AF (see also Evidence Table 2)

3.4. Diagnostic evaluation of new AF

All patients with AF should be offered a comprehensive diagnostic assessment and review of medical history to identify risk factors and/or comorbidities needing active treatment. Table 8 displays the essential diagnostic work-up for a patient with AF.

Table 8

Diagnostic work-up for patients with AF

All patientsSelected patients
  • Medical history to determine AF pattern, relevant family history, and comorbidities, and to assess risk factors for thromboembolism and bleeding

  • Ambulatory ECG monitoring for assessing AF burden and ventricular rate control

  • Exercise ECG to evaluate rate control or effects of class IC antiarrhythmic drugs

  • 12-lead ECG

  • Further blood tests for investigation of cardiovascular disease and refinement of stroke/bleeding risk (e.g. NT-proBNP, troponin)

  • Assess symptoms and functional impairment

  • Transoesophageal echocardiography for left atrial thrombus and valvular disease assessment

  • Collect generic or AF-specific patient-reported outcome measures

  • Coronary CT, angiography, or ischaemia imaging for suspected CAD

  • Blood tests (full blood count, kidney function, serum electrolytes, liver function, glucose/HbA1c, and thyroid function)

  • CMR for evaluation of atrial and ventricular cardiomyopathies, and to plan interventional procedures

  • Transthoracic echocardiography where this will guide AF-CARE management decisions

  • Brain imaging and cognitive function assessment for cerebrovascular disease and dementia risk

All patientsSelected patients
  • Medical history to determine AF pattern, relevant family history, and comorbidities, and to assess risk factors for thromboembolism and bleeding

  • Ambulatory ECG monitoring for assessing AF burden and ventricular rate control

  • Exercise ECG to evaluate rate control or effects of class IC antiarrhythmic drugs

  • 12-lead ECG

  • Further blood tests for investigation of cardiovascular disease and refinement of stroke/bleeding risk (e.g. NT-proBNP, troponin)

  • Assess symptoms and functional impairment

  • Transoesophageal echocardiography for left atrial thrombus and valvular disease assessment

  • Collect generic or AF-specific patient-reported outcome measures

  • Coronary CT, angiography, or ischaemia imaging for suspected CAD

  • Blood tests (full blood count, kidney function, serum electrolytes, liver function, glucose/HbA1c, and thyroid function)

  • CMR for evaluation of atrial and ventricular cardiomyopathies, and to plan interventional procedures

  • Transthoracic echocardiography where this will guide AF-CARE management decisions

  • Brain imaging and cognitive function assessment for cerebrovascular disease and dementia risk

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; CAD, coronary artery disease; CMR, cardiac magnetic resonance; CT, computed tomography; CTA, computed tomography angiography; ECG, electrocardiogram; HbA1c, glycated haemoglobin; NT-proBNP, N-terminal pro-B-type natriuretic peptide.

© ESC 2024
Table 8

Diagnostic work-up for patients with AF

All patientsSelected patients
  • Medical history to determine AF pattern, relevant family history, and comorbidities, and to assess risk factors for thromboembolism and bleeding

  • Ambulatory ECG monitoring for assessing AF burden and ventricular rate control

  • Exercise ECG to evaluate rate control or effects of class IC antiarrhythmic drugs

  • 12-lead ECG

  • Further blood tests for investigation of cardiovascular disease and refinement of stroke/bleeding risk (e.g. NT-proBNP, troponin)

  • Assess symptoms and functional impairment

  • Transoesophageal echocardiography for left atrial thrombus and valvular disease assessment

  • Collect generic or AF-specific patient-reported outcome measures

  • Coronary CT, angiography, or ischaemia imaging for suspected CAD

  • Blood tests (full blood count, kidney function, serum electrolytes, liver function, glucose/HbA1c, and thyroid function)

  • CMR for evaluation of atrial and ventricular cardiomyopathies, and to plan interventional procedures

  • Transthoracic echocardiography where this will guide AF-CARE management decisions

  • Brain imaging and cognitive function assessment for cerebrovascular disease and dementia risk

All patientsSelected patients
  • Medical history to determine AF pattern, relevant family history, and comorbidities, and to assess risk factors for thromboembolism and bleeding

  • Ambulatory ECG monitoring for assessing AF burden and ventricular rate control

  • Exercise ECG to evaluate rate control or effects of class IC antiarrhythmic drugs

  • 12-lead ECG

  • Further blood tests for investigation of cardiovascular disease and refinement of stroke/bleeding risk (e.g. NT-proBNP, troponin)

  • Assess symptoms and functional impairment

  • Transoesophageal echocardiography for left atrial thrombus and valvular disease assessment

  • Collect generic or AF-specific patient-reported outcome measures

  • Coronary CT, angiography, or ischaemia imaging for suspected CAD

  • Blood tests (full blood count, kidney function, serum electrolytes, liver function, glucose/HbA1c, and thyroid function)

  • CMR for evaluation of atrial and ventricular cardiomyopathies, and to plan interventional procedures

  • Transthoracic echocardiography where this will guide AF-CARE management decisions

  • Brain imaging and cognitive function assessment for cerebrovascular disease and dementia risk

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; CAD, coronary artery disease; CMR, cardiac magnetic resonance; CT, computed tomography; CTA, computed tomography angiography; ECG, electrocardiogram; HbA1c, glycated haemoglobin; NT-proBNP, N-terminal pro-B-type natriuretic peptide.

© ESC 2024

A 12-lead ECG is warranted in all AF patients to confirm rhythm, determine ventricular rate, and look for signs of structural heart disease, conduction defects, or ischaemia.56 Blood tests should be carried out (kidney function, serum electrolytes, liver function, full blood count, glucose/glycated haemoglobin [HbA1c], and thyroid tests) to detect any concomitant conditions that may exacerbate AF or increase the risk of bleeding and/or thromboembolism.57,58

Other investigations will depend on individualized assessment and the planned treatment strategy.59–65 A transthoracic echocardiogram (TTE) should be carried out in the initial work-up, where this will guide management decisions, or in patients where there is a change in cardiovascular signs or symptoms. The task force recognizes that accessibility to TTE might be limited or delayed in the primary care setting, but this should not delay initiation of oral anticoagulation (OAC) or other components of AF-CARE where indicated.66 Further details on TTE and reassessment (e.g. if elevated heart rate limits diagnostic imaging, or where there is a change in clinical status) are presented in Section 8.3. Additional imaging using different modalities may be required to assist with comorbidity and AF-related management (see Supplementary data online, Figure S1).

Recommendation Table 3

Recommendations for diagnostic evaluation in patients with new AF (see also Evidence Table 3)

Recommendation Table 3

Recommendations for diagnostic evaluation in patients with new AF (see also Evidence Table 3)

3.5. Adverse events associated with AF

Atrial fibrillation is associated with a range of serious adverse events (Figure 1) (see Supplementary data online, Additional Evidence Table S2). Patients with AF also have high rates of hospitalization and complications from coexisting medical conditions. The most common non-fatal outcome in those with AF is heart failure, occurring in around half of patients over time. Patients with AF have a four- to five-fold increase in the relative risk (RR) of heart failure compared with those without AF, as demonstrated in two meta-analyses (RR, 4.62; 95% CI, 3.13–6.83 and RR, 4.99; 95% CI, 3.0–8.22).68,69 The next most common adverse impacts from AF are ischaemic stroke (RR, 2.3; 95% CI, 1.84–2.94), ischaemic heart disease (RR, 1.61; 95% CI, 1.38–1.87), and other thromboembolic events.69–71 The latter typically include arterial thromboembolic events (preferred to the term systemic), although venous thromboembolism is also associated with AF.72,73 Patients with AF also have an increased risk of cognitive impairment (adjusted hazard ratio [HR], 1.39; 95% CI, 1.25–1.53)74 and dementia (OR, 1.6; 95% CI, 1.3–2.0).75–77 It should be noted that most of the observational studies on adverse events have a mix of patients taking and not taking OAC. When carefully controlling for the confounding effects of stroke, comorbidities, and OAC, AF exposure was still significantly associated with vascular dementia (HR, 1.68; 95% CI, 1.33–2.12; P < .001), but not Alzheimer's disease (HR, 0.85; 95% CI, 0.70–1.03; P = .09).78

Hospital admission rates due to AF vary widely depending on the population studied, and may be skewed by selection bias. In a Dutch RCT including first-diagnosed AF patients (mean age 64 years), cardiovascular hospitalization rates were 7.0% to 9.4% per year.79 An Australian study identified 473 501 hospitalizations for AF during 15 years of follow-up (300 million person-years), with a relative increase in AF hospitalizations of 203% over the study period, in contrast to an increase for all hospitalizations of 71%. The age-specific incidence of hospital admission increased particularly in the older age groups.80

Atrial fibrillation is also associated with increased mortality. In 2017, AF contributed to over 250 000 deaths globally, with an age-standardized mortality rate of 4.0 per 100 000 people (95% uncertainty interval 3.9–4.2).81 The most frequent cause of death in patients with AF is heart failure related,70 with complex relationships to cardiovascular and non-cardiovascular disease.82 There is up to a two-fold increased risk of all-cause mortality (RR, 1.95; 95% CI, 1.50–2.54),68 and cardiovascular mortality (RR, 2.03; 95% CI, 1.79–2.30)69 in AF compared with sinus rhythm. Even in the absence of major thromboembolic risk factors, the incidence of death is 15.5 per 1000 person-years in those with AF exposure, compared with 9.4 per 1000 person-years without (adjusted HR, 1.44; 95% CI, 1.38–1.50; P < .001).78 Patients with OAC-related bleeding have higher mortality, including both minor and major bleeding (as defined by the International Society on Thrombosis and Haemostasis scale).83 Despite OAC, patients with AF remain at high residual risk of death, highlighting the importance of attention to concomitant disease.84

3.6. Atrial flutter

Atrial flutter (AFL) is the among the most common atrial tachyarrhythmias, with an overall incidence rate of 88 per 100 000 person-years, rising to 317 per 100 000 person-years in people over 50 years of age.85 Risk factors for AFL and AF are similar, and more than half of all patients with AFL will develop AF.85 Observational studies suggest that thromboembolic risk is elevated in AFL.86 In direct comparison of AFL with AF, some studies suggest a similar risk of stroke and others a lower risk in AFL,87–90 possibly due to different comorbidity burdens and the impact of confounders such as AFL/AF ablation and anticoagulation (more frequently stopped in AFL).91

4. Patient pathways and management of AF

4.1. Patient-centred, multidisciplinary AF management

4.1.1. The patient at the heart of care

A patient-centred and integrated approach to AF management means working with a model of care that respects the patient's experience, values, needs, and preferences for planning, co-ordination, and delivery of care. A central component of this model is the therapeutic relationship between the patient and the multidisciplinary team of healthcare professionals (Figure 2). In patient-centred AF management, patients are seen not as passive recipients of health services, but as active participants who work as partners alongside healthcare professionals. Patient-centred AF management requires integration of all aspects of AF management. This includes symptom control, lifestyle recommendations, psychosocial support, and management of comorbidities, alongside optimal medical treatment consisting of pharmacotherapy, cardioversion, and interventional or surgical ablation (Table 9). Services should be designed to ensure that all patients have access to an organized model of AF management, including tertiary care specialist services when indicated (see Supplementary data online, Table S1, Evidence Table 4 and Additional Evidence Table S3). It is equally important to maintain pathways for patients to promptly re-engage with specialist services when their condition alters.

Multidisciplinary approach to AF management.
Figure 2

Multidisciplinary approach to AF management.

Principal caregivers are involved in the community and hospital settings to provide optimal, patient-centred care for patients living with AF. AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment.

Table 9

Achieving patient-centred AF management

Components of patient-centred AF management:
  • Optimal treatment according to the AF-CARE pathway, which includes:

  ∘ [C] Comorbidity and risk factor management
  ∘ [A] Avoid stroke and thromboembolism
  ∘ [R] Reduce symptoms by rate and rhythm control
  ∘ [E] Evaluation and dynamic reassessment
  • Lifestyle recommendations

  • Psychosocial support

  • Education and awareness for patients, family members, and caregivers

  • Seamless co-ordination between primary care and specialized AF care

How to implement patient-centred AF management:
  • Shared decision-making

  • Multidisciplinary team approach

  • Patient education and empowerment, with emphasis on self-care

  • Structured educational programmes for healthcare professionals

  • Technology support (e-Health, m-Health, telemedicine)a

Components of patient-centred AF management:
  • Optimal treatment according to the AF-CARE pathway, which includes:

  ∘ [C] Comorbidity and risk factor management
  ∘ [A] Avoid stroke and thromboembolism
  ∘ [R] Reduce symptoms by rate and rhythm control
  ∘ [E] Evaluation and dynamic reassessment
  • Lifestyle recommendations

  • Psychosocial support

  • Education and awareness for patients, family members, and caregivers

  • Seamless co-ordination between primary care and specialized AF care

How to implement patient-centred AF management:
  • Shared decision-making

  • Multidisciplinary team approach

  • Patient education and empowerment, with emphasis on self-care

  • Structured educational programmes for healthcare professionals

  • Technology support (e-Health, m-Health, telemedicine)a

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment.

ae-Health refers to healthcare services provided using electronic methods; m-Health, refers to healthcare services supported by mobile devices; and telemedicine refers to remote diagnosis or treatment supported by telecommunications technology.

© ESC 2024
Table 9

Achieving patient-centred AF management

Components of patient-centred AF management:
  • Optimal treatment according to the AF-CARE pathway, which includes:

  ∘ [C] Comorbidity and risk factor management
  ∘ [A] Avoid stroke and thromboembolism
  ∘ [R] Reduce symptoms by rate and rhythm control
  ∘ [E] Evaluation and dynamic reassessment
  • Lifestyle recommendations

  • Psychosocial support

  • Education and awareness for patients, family members, and caregivers

  • Seamless co-ordination between primary care and specialized AF care

How to implement patient-centred AF management:
  • Shared decision-making

  • Multidisciplinary team approach

  • Patient education and empowerment, with emphasis on self-care

  • Structured educational programmes for healthcare professionals

  • Technology support (e-Health, m-Health, telemedicine)a

Components of patient-centred AF management:
  • Optimal treatment according to the AF-CARE pathway, which includes:

  ∘ [C] Comorbidity and risk factor management
  ∘ [A] Avoid stroke and thromboembolism
  ∘ [R] Reduce symptoms by rate and rhythm control
  ∘ [E] Evaluation and dynamic reassessment
  • Lifestyle recommendations

  • Psychosocial support

  • Education and awareness for patients, family members, and caregivers

  • Seamless co-ordination between primary care and specialized AF care

How to implement patient-centred AF management:
  • Shared decision-making

  • Multidisciplinary team approach

  • Patient education and empowerment, with emphasis on self-care

  • Structured educational programmes for healthcare professionals

  • Technology support (e-Health, m-Health, telemedicine)a

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment.

ae-Health refers to healthcare services provided using electronic methods; m-Health, refers to healthcare services supported by mobile devices; and telemedicine refers to remote diagnosis or treatment supported by telecommunications technology.

© ESC 2024

4.1.2. Education and shared decision-making

Clear advice about the rationale for treatments, the possibility of treatment modification, and shared decision-making can help patients live with AF (see Supplementary data online, Table S2).92 An open and effective relationship between the patient and the healthcare professional is critical, with shared decision-making found to improve outcomes for OAC and arrhythmia management.93,94 In using a shared approach, both the clinician and patient are involved in the decision-making process (to the extent that the patient prefers). Information is shared in both directions. Furthermore, both the clinician and the patient express their preferences and discuss the options. Of the potential treatment decisions, no treatment is also a possibility.95 There are several toolkits available to facilitate this, although most are focused on anticoagulation decisions. For example, the Shared Decision-Making Toolkit (http://afibguide.com, http://afibguide.com/clinician) and the Successful Intravenous Cardioversion for Atrial Fibrillation (SIC-AF) score have been shown to reduce decisional conflict compared with usual care in patients with AF.93,94 Patient-support organizations can also make an important contribution to providing understandable and actionable knowledge about AF and its treatments (e.g. local support groups and international charities, such as http://afa-international.org). As AF is a chronic or recurrent disease in most patients, education is central to empower patients, their families, and caregivers.

4.1.3. Education of healthcare professionals

Gaps in knowledge and skills across all domains of AF care are consistently described among cardiologists, neurologists, internal medicine specialists, emergency physicians, general practitioners, nurses, and allied health practitioners.96–98 Healthcare professionals involved in multidisciplinary AF management should have a knowledge of all available options for diagnosis and treatment.99–101 In the STEEER-AF trial,99 real-world adherence to clinical practice guidelines for AF across six ESC countries was poor. These findings highlight the critical need for appropriate training and education of healthcare professionals.102

Specifically targeted education for healthcare professionals can increase knowledge and lead to more appropriate use of OAC for prevention of thromboembolism.103 However, educational interventions for healthcare providers are often not enough to sustainably impact behaviour.104 Other tools may be needed, such as active feedback,103 clinical decision support tools,105 expert consultation,106 or e-Health learning.107

4.1.4. Inclusive management of AF

Evidence is growing on differences in AF incidence, prevalence, risk factors, comorbidities, and outcomes according to gender.108 Women diagnosed with AF are generally older, have more hypertension and heart failure with preserved ejection fraction (HFpEF), and have less diagnosed coronary artery disease (CAD).109 Registry studies have reported differences in outcomes, with higher morbidity and mortality in women, although these may be confounded by age and comorbidity burden.110–112 Women with AF may be more symptomatic, and report a lower quality of life.41,113 It is unclear whether this is related to delayed medical assessment in women, or whether there are genuine sex differences. Despite a higher symptom load, women are less likely to undergo AF ablation than men, even though antiarrhythmic drug therapy seems to be associated with more proarrhythmic events in women.109 These observations call for more research on gender differences in order to prevent disparities and inequality in care. Other diversity aspects such as age, race, ethnicity, and transgender issues, as well as social determinants (including socioeconomic status, disability, education level, health literacy, and rural/urban location) are important contributors to inequality that should be actively considered to improve patient outcomes.114

4.2. Principles of AF-CARE

The 2024 ESC Guidelines for the management of AF have compiled and evolved past approaches to create principles of management to aid implementation of these guidelines, and hence improve patient care and outcomes. There is growing evidence that clinical support tools115–118 can aid best-practice management, with the caveat that any tool is a guide only, and that all patients require personalized attention. The AF-CARE approach covers many established principles in the management of AF, but does so in a systematic, time-orientated format with four essential treatment pillars (Figure 3; central illustration). Joint management with each patient forms the starting point of the AF-CARE approach. Notably, it takes account of the growing evidence base that therapies for AF are most effective when associated health conditions are addressed. A careful search for these comorbidities and risk factors [C] is critical and should be applied in all patients with a diagnosis of AF. Avoidance of stroke and thromboembolism [A] in patients with risk factors is considered next, focused on appropriate use of anticoagulant therapy. Reducing AF-related symptoms and morbidity by effective use of heart rate and rhythm control [R] is then applied, which in selected patients may also reduce hospitalization or improve prognosis. The potential benefit of rhythm control, accompanied by consideration of all risks involved, should be considered in all patients at each contact point with healthcare professionals. As AF, and its related comorbidities, changes over time, different levels of evaluation [E] and re-evaluation are required in each patient, and these approaches should be dynamic. Due to the wide variability in response to therapy, and the changing pathophysiology of AF as age and comorbidities advance, reassessment should be built into the standard care pathway to prevent adverse outcomes for patients and improve population health.

Central illustration. Patient pathway for AF-CARE (see Figures 4, 5, 6, and 7 for the [R] pathways for first-diagnosed, paroxysmal, persistent and permanent AF).
Figure 3

Central illustration. Patient pathway for AF-CARE (see Figures 4, 5, 6, and 7 for the [R] pathways for first-diagnosed, paroxysmal, persistent and permanent AF).

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; CCS, chronic coronary syndrome; CHA2DS2-VA, congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years; DOAC, direct oral anticoagulant; ECG, electrocardiogram; HFrEF, heart failure with reduced ejection fraction; INR, international normalized ratio of prothrombin time; OAC, oral anticoagulant; OSA, obstructive sleep apnoea; PVD, peripheral vascular disease; SGLT2, sodium-glucose cotransporter-2; VKA, vitamin K antagonist. aAs part of a comprehensive management of cardiometabolic risk factors.

AF-CARE builds upon prior ESC Guidelines, e.g. the five-step outcome-focused integrated approach in the 2016 ESC Guidelines for the management of AF,119 and the AF Better Care (ABC) pathway in the 2020 ESC Guidelines for the diagnosis and management of AF.120 The reorganization into AF-CARE was based on the parallel developments in new approaches and technologies (in particular for rhythm control), with new evidence consistently suggesting that all aspects of AF management are more effective when comorbidities and risk factors have been considered. This includes management relating to symptom benefit, improving prognosis, prevention of thromboembolism, and the response to rate and rhythm control strategies. AF-CARE makes explicit the need for individualized evaluation and follow-up in every patient, with an active approach that accounts for how patients, their AF, and associated comorbidities change over time. The AF-CARE principles have been applied to different patient pathways for ease of implementation into routine clinical care. This includes the management of first-diagnosed AF (Figure 4), paroxysmal AF (Figure 5), persistent AF (Figure 6), and permanent AF (Figure 7).

[R] Pathway for patients with first-diagnosed AF.
Figure 4

[R] Pathway for patients with first-diagnosed AF.

AF, atrial fibrillation; AF-CARE, Atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; LVEF, left ventricular ejection fraction. After following the pathway for first-diagnosed AF, patients with recurrent AF should enter the AF-CARE [R] pathway for paroxysmal, persistent, or permanent AF, depending on the type of their AF.

[R] Pathway for patients with paroxysmal AF.
Figure 5

[R] Pathway for patients with paroxysmal AF.

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; b.p.m., beats per minute; HFmrEF, heart failure with mildly reduced ejection fraction; HFrEF, Heart failure with reduced ejection fraction; LVEF, left ventricular ejection fraction. aIn patients with HFrEF: Class I if high probability of tachycardia-induced cardiomyopathy; and Class IIa in selected patients to improve prognosis.

[R] Pathway for patients with persistent AF.
Figure 6

[R] Pathway for patients with persistent AF.

AF, atrial fibrillation; AF-CARE, Atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; b.p.m., beats per minute; HFmrEF, heart failure with mildly reduced ejection fraction; HFrEF, heart failure with reduced ejection fraction; LVEF, left ventricular ejection fraction. aIn patients with HFrEF: Class I if high probability of tachycardia-induced cardiomyopathy; and Class IIa in selected patients to improve prognosis.

[R] Pathway for patients with permanent AF.
Figure 7

[R] Pathway for patients with permanent AF.

AF, atrial fibrillation; AF-CARE, Atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; b.p.m., beats per minute; CRT, cardiac resynchronization therapy; HF, heart failure; LVEF, left ventricular ejection fraction. Permanent AF is a shared decision made between the patient and physician that no further attempts at restoration of sinus rhythm are planned. aNote that the combination of beta-blockers with diltiazem or verapamil should only be used under specialist advice, and monitored with an ambulatory ECG to check for bradycardia.

Recommendation Table 4

Recommendations for patient-centred care and education (see also Evidence Table 4)

Recommendation Table 4

Recommendations for patient-centred care and education (see also Evidence Table 4)

5. [C] Comorbidity and risk factor management

A broad array of comorbidities are associated with the recurrence and progression of AF. Managing comorbidities is also central to the success of other aspects of care for patients with AF, with evidence available for hypertension, heart failure, diabetes mellitus, obesity, and sleep apnoea, along with lifestyle changes that improve physical activity and reduce alcohol intake (see Supplementary data online, Additional Evidence Table S4). Identification and treatment of these comorbidities and clusters of risk factors form an important part of effective AF-CARE (Figure 8), with the evidence outlined in the rest of this section highlighting where management can improve patient outcomes or prevent AF recurrence. Many of these factors (and more) are also associated with incident AF (see Section 10).

Management of key comorbidities to reduce AF recurrence.
Figure 8

Management of key comorbidities to reduce AF recurrence.

LVEF, left ventricular ejection fraction; SGLT2, sodium-glucose cotransporter-2.

Recommendation Table 5

Recommendations for comorbidity and risk factor management in AF (see also Evidence Table 5)

Recommendation Table 5

Recommendations for comorbidity and risk factor management in AF (see also Evidence Table 5)

5.1. Hypertension

Hypertension in patients with AF is associated with an increased risk of stroke, heart failure, major bleeding, and cardiovascular mortality.158–161 The target for treated systolic blood pressure (BP) in most adults is 120–129 mmHg. Where BP-lowering treatment is poorly tolerated, clinically significant frailty exists or the patient's age is 85 years or older, a more lenient target of <140 mmHg is acceptable or ‘as low as reasonably achievable’. On-treatment diastolic BP should ideally be 70–79 mmHg.162 In an individual participant data meta-analysis of 22 randomized trials reporting baseline AF, a 5 mmHg reduction in systolic BP reduced the risk of major cardiovascular events by 9% (HR, 0.91; 95% CI, 0.83–1.00), with identical effect in patients with AF or sinus rhythm.129

In individuals with AF, hypertension often coexists with other modifiable and non-modifiable risk factors that all contribute to recurrence of AF, readmission to hospital, and ongoing symptoms after rhythm control.163–171 Optimal control of blood pressure should be considered an essential component of treating AF and undertaken within a strategy of comprehensive risk factor management.126–128 Although the majority of research has focused on clinical outcomes, limited comparative data on hypertension medication suggests that use of angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARB) may be superior for prevention of recurrent AF.172–175

5.2. Heart failure

Heart failure is a key determinant of prognosis in patients with AF, as well as an important factor associated with recurrence and progression of AF.176,177 During 30 years of follow-up in the Framingham cohort, 57% of those with new heart failure had concomitant AF, and 37% of those with new AF had heart failure.178 Numerous cardiovascular and non-cardiovascular conditions impact the development of both AF and heart failure, leading to the common pathway of atrial cardiomyopathy.18 In patients with acute heart failure attending the emergency department, AF is one of the most prevalent triggering factors of the episode.179 The development of heart failure in patients with AF is associated with a two-fold increase in stroke and thromboembolism,180 even after anticoagulation,181 and 25% higher all-cause mortality.178 Prognosis may be affected by left ventricular ejection fraction (LVEF), with the rate of death highest with the combination of AF and heart failure with reduced ejection fraction (HFrEF) (LVEF ≤ 40%), as compared with AF and HFpEF (LVEF ≥ 50%). However, rates of stroke and incident heart failure hospitalization are similar regardless of LVEF.182 Due to how common concomitant AF and heart failure are in clinical practice, strategies to improve outcomes in these patients are detailed within each component of the AF-CARE pathway. However, it is also critical that heart failure itself is managed appropriately in patients with AF to prevent avoidable adverse events.

Optimization of heart failure management should follow current ESC Guidelines: 2023 Focused Update183 of the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure.137 Achieving euvolaemia with diuretics is an important first step that not only manages the heart failure component, but can also facilitate better control of heart rate in AF. For HFrEF, it should be highlighted that many older guideline-recommended therapies lack specific evidence for benefit in patients with coexisting AF. No trial data are available in this context for ACE inhibitors, there are conflicting data on ARBs,132,184 and an individual patient-level analysis of RCTs found no difference between beta-blockers and placebo for all-cause mortality in HFrEF with AF.133 However, these drugs have clear proof of safety and there may be other indications for these therapies beyond prognosis, including comorbidity management and symptom improvement. These and other therapies may also have dual functions, for example, beta-blockers or digoxin for rate control of AF, in addition to improving heart failure metrics and reducing hospitalization.48,185,186 More recent additions to HFrEF management, such as eplerenone, sacubitril-valsartan, and sodium-glucose cotransporter-2 (SGLT2) inhibitors, had substantial numbers of patients with AF enrolled in RCTs, with no evidence that AF status affected their ability to reduce cardiovascular mortality/heart failure hospitalization.134–136 Cardiac resynchronization therapy (CRT) in the context of HFrEF and AF is discussed in detail in the 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy, with an important focus on ensuring effective biventricular pacing (with a low threshold for considering atrioventricular node ablation).187 Patients who have heart failure with mildly reduced ejection fraction (HFmrEF) (LVEF 41%–49%) and AF should generally be treated according to guidance for HFrEF,137 albeit with limited evidence to date in AF.188–190 For treatment of HFpEF and AF,191 pre-specified subgroup data on AF from multiple large trials show that the SGLT2 inhibitors dapagliflozin, empaglifozin, and sotagliflozin are effective in improving prognosis.138–140

Appropriate management of heart failure has the potential to reduce recurrence of AF, e.g. by reducing adverse atrial and ventricular myocardial remodelling, but there are limited data for specific therapies. In the Routine versus Aggressive upstream rhythm Control for prevention of Early AF in heart failure (RACE 3) trial, combined management of mild-to-moderate heart failure with ACE inhibitors/ARBs, mineralocorticoid receptor antagonists, statins, and cardiac rehabilitation increased the maintenance of sinus rhythm on ambulatory monitoring at 12 months.39 This benefit was not preserved at the 5 year follow-up, although this may have been confounded by the lack of ongoing intervention beyond the initial 12 months.192

5.3. Type 2 diabetes mellitus

Diabetes mellitus is present in around 25% of patients with AF.193–195 Patients with both diabetes and AF have a worse prognosis,196 with increased healthcare utilization and excess mortality and cardiovascular events. The prevalence and incidence of AF and type 2 diabetes are widely increasing, thus making the association of these two conditions a public health challenge.195,197 Moreover, diabetes is a major factor influencing thromboembolic risk.198,199 Following catheter ablation of AF, diabetes and higher HbA1c are associated with increased length of stay and a greater recurrence of AF.200–203

In cohort studies, the management of diabetes mellitus as part of comprehensive risk factor management has been associated with reduced AF symptoms, burden, reversal of the type of AF (from persistent to paroxysmal or no AF), and improved maintenance of sinus rhythm.126–128 However, robust evidence is limited, and individual glucose-lowering medications have had variable effects on AF.204–206 There are emerging data of the use of SGLT2 and glucagon-like peptide-1 antagonists in patients with diabetes and AF that may impact on treatment choice in the near future. Importantly, diabetes frequently coexists with multiple risk factors in patients with AF, and a comprehensive approach to management is required. Further details are provided in the 2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes.207

5.4. Obesity

Obesity frequently coexists with other risk factors that have been independently associated with the development of AF.208,209 Obesity (body mass index [BMI] ≥30 kg/m2) and being overweight (BMI >25 kg/m2) are associated with a greater risk of recurrent atrial arrhythmias after AF ablation (13% increase for every 5 kg/m2 higher BMI).210–212 In the setting of comprehensive risk factor management, weight loss of ≥10% in overweight and obese individuals with AF has been associated with reduced AF symptoms and AF burden in an RCT (aiming for BMI <27 kg/m2).125 Cohort studies have also shown a graded response to maintenance of sinus rhythm,126 improved ablation outcomes,128 and reversal of the type of AF127 commensurate with the degree of weight loss and risk factor management. However, in the Supervised Obesity Reduction Trial for AF Ablation Patients (SORT-AF) randomized trial in AF ablation patients, a sole weight loss intervention that achieved 4% loss in weight over 12 months did not impact ablation outcomes.213 This is consistent with the findings in LEGACY (Long-Term Effect of Goal directed weight management on Atrial Fibrillation Cohort: a 5 Year follow-up study) that showed that weight loss of ≤3% had no impact on AF recurrence.126 Observational studies have raised the possibility of a point of no return in terms of the benefit of weight loss,214 but also the possibility that bariatric surgery can improve symptoms and reduce AF recurrence.215–217

5.5. Obstructive sleep apnoea

Obstructive sleep apnoea (OSA) is a highly prevalent condition, particularly in patients with AF.157,218 Optimal screening tools in the AF population are still under evaluation, although it may be reasonable to screen for OSA in patients where a rhythm control strategy is being pursued. Polysomnography or home sleep apnoea testing are suggested in preference to screening questionnaires.155–157,219 Questionnaires assessing daytime sleepiness are poor predictors of moderate-to-severe OSA.155 Which parameter should be used to focus on risk of AF in patients with OSA, and to guide OSA treatment in patients with AF, is still unclear.220,221

Observational studies have suggested that individuals with OSA not treated with continuous positive airway pressure (CPAP) respond poorly to treatments for AF, with an increased risk of recurrence after cardioversion or ablation.222 Conversely, OSA patients treated with CPAP seem to mitigate their propensity toward developing AF.148–153,222–224 A small randomized trial of CPAP vs. no therapy demonstrated reversal of atrial remodelling in individuals with moderate OSA.154 However, other small RCTs have failed to show a benefit of CPAP therapy on ablation outcomes225 or post-cardioversion.226 Data on the cardiovascular mortality benefit of CPAP therapy in OSA are inconclusive.227–230

5.6. Physical inactivity

Reduced cardiorespiratory fitness frequently coexists with other modifiable risk factors and has been associated with a greater recurrence of AF after catheter ablation.141 Better cardiorespiratory fitness has a demonstrated inverse relationship to AF burden in both middle-aged and elderly people.141 Small RCTs, meta-analyses, and observational cohorts have shown that regular aerobic exercise may also improve AF-related symptoms, quality of life, and exercise capacity.142,143 Better cardiorespiratory fitness and a gain in cardiorespiratory fitness over time are associated with a greater reduction in AF burden and improved maintenance of sinus rhythm.141–145

5.7. Alcohol excess

Alcohol consumption can increase the risk of adverse events in patients with AF, such as thromboembolism, death, or AF-related hospitalization.231,232 Alcohol is associated with an increased risk of ischaemic stroke in patients with newly diagnosed AF, and alcohol abstinence after AF diagnosis can reduce the risk of ischaemic stroke.233 In patients receiving OAC, alcohol excess is associated with a greater risk of bleeding,234 mediated by poor adherence, alcohol–drug interactions, liver disease, and variceal bleeding.

Alcohol consumption is associated with a dose-dependent increase in the recurrence of AF after catheter ablation.147,235 In an RCT among regular non-binge drinkers with AF, the goal of abstinence led to a significant reduction in AF recurrence and burden; alcohol intake was reduced from 16.8 to 2.1 standard drinks per week (≤30 grams or 3 standard drinks of alcohol) in the intervention arm, with 61% attaining abstinence.147 In observational data of patients undergoing catheter ablation, reduction of consumption to ≤7 standard drinks (≤70 grams of alcohol) per week was associated with improved maintenance of sinus rhythm.128,235

6. [A] Avoid stroke and thromboembolism

6.1. Initiating oral anticoagulation

Atrial fibrillation is a major risk factor for thromboembolism, irrespective of whether it is paroxysmal, persistent, or permanent.236,237 Left untreated, and dependent on other patient-specific factors, the risk of ischaemic stroke in AF is increased five-fold, and one in every five strokes is associated with AF.238 The default approach should therefore be to provide OAC to all eligible patients, except those at low risk of incident stroke or thromboembolism. The effectiveness of OAC to prevent ischaemic stroke in patients with AF is well established.239,240 Antiplatelet drugs alone (aspirin, or aspirin in combination with clopidogrel) are not recommended for stroke prevention in AF.241,242

6.1.1. Decision support for anticoagulation in AF

Tools have been developed to enable easier implementation of OAC in patients with clinical AF. The majority of OAC clinical trials have used variations of the CHADS2 score to indicate those at risk (with points for chronic heart failure, hypertension, age, diabetes, and 2 points for prior stroke/transient ischaemic attack [TIA]). Although most available stroke risk scores are simple and practical, the predictive value of scores is generally modest (see Supplementary data online, Table S3).243–245 Classification and discrimination of adverse events is relatively poor for all scores and hence the benefit of using them to select patients for OAC is unclear. There is also considerable variation in the definition of risk factors across countries,246 and a lack of evidence from clinical trials on the ability of stroke risk scoring to enhance clinical practice.243 This guideline continues to provide a Class IA recommendation for the use of OAC in patients at risk of thromboembolism. However, in the absence of strong evidence for how to apply risk scores in real-world patients, this has been separated from the use of any particular risk score. This is also in line with regulatory approvals for direct oral anticoagulants (DOACs), which do not stipulate risk scores or numerical thresholds.25–28,245

Substantive changes have occurred in the decades since these risk scores were developed in regards to population-level risk factor profiles, therapies, and targets.198 Historical scores do not take into account parameters that have been associated with thromboembolism in contemporary cohorts, such as cancer, chronic kidney disease (CKD), ethnicity, and a range of circulating biomarkers (including troponin and B-type natriuretic peptide [BNP]). As an example, for CKD there is a correlation between decreasing glomerular filtration rate and proteinuria with stroke risk,247–250 and cohort data suggest a two-fold increased risk of ischaemic stroke and mortality in AF patients with CKD vs. without.251 Other factors, such as atrial enlargement, hyperlipidaemia, smoking, and obesity, have been identified in specific cohort studies as additional risk factors for ischaemic stroke in AF.70,252,253 Biomarkers, such as troponin, natriuretic peptides, growth differentiation factor-15, cystatin C, and interleukin-6, can also indicate residual stroke risk among anticoagulated AF patients.254,255 Biomarker-guided stroke prevention is currently being evaluated in an ongoing RCT (NCT03753490). Until further validation within RCTs is available, this task force continues to support using simple clinical classification for implementation of OAC. Clinicians should use tools that have been validated in their local population and take an individualized approach to thromboembolic risk stratification that considers the full range of each patient's specific risk factors. The absolute risk level at which to start OAC in individual patients cannot be estimated from population-level studies. It will vary depending on how those factors interact with other medical issues, and the degree of risk acceptable or tolerated by that person. In general, most of the available risk scores have a threshold of 0.6%–1.0% per annum of thromboembolic events for clinical AF to warrant OAC prescription.

Across Europe, the most popular risk score is CHA2DS2–VASc, giving points for congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/TIA/thromboembolism (2 points), vascular disease, age 65–74 years and female sex. However, implementation has varied in terms of gender. Female sex is an age-dependent stroke risk modifier rather than a risk factor per se.112,256,257 The inclusion of gender complicates clinical practice both for healthcare professionals and patients.258 It also omits individuals who identify as non-binary, transgender, or are undergoing sex hormone therapy. Previous guidelines from the ESC (and globally) have not actually used CHA2DS2-VASc; instead providing different score levels for women and men with AF to qualify for OAC. Hence, CHA2DS2-VA (excluding gender) has effectively been in place (Table 10).78 This task force proposes, in the absence of other locally validated alternatives, that clinicians and patients should use the CHA2DS2-VA score to assist in decisions on OAC therapy (i.e. without a criterion for birth sex or gender). Pending further trials in lower risk patients (NCT04700826,259 NCT02387229260), OAC are recommended in those with a CHA2DS2-VA score of 2 or more and should be considered in those with a CHA2DS2-VA score of 1, following a patient-centred and shared care approach. Healthcare professionals should take care to assess for other thromboembolic risk factors that may also indicate the need for OAC prescription.

Table 10

Updated definitions for the CHA2DS2-VA score

CHA2DS2-VA componentDefinition and commentsPoints awardeda
CChronic heart failureSymptoms and signs of heart failure (irrespective of LVEF, thus including HFpEF, HFmrEF, and HFrEF), or the presence of asymptomatic LVEF ≤40%.261–2631
HHypertensionResting blood pressure >140/90 mmHg on at least two occasions, or current antihypertensive treatment. The optimal BP target associated with lowest risk of major cardiovascular events is 120–129/70–79 mmHg (or keep as low as reasonably achievable).162,2641
AAge 75 years or aboveAge is an independent determinant of ischaemic stroke risk.265 Age-related risk is a continuum, but for reasons of practicality, two points are given for age ≥75 years.2
DDiabetes mellitusDiabetes mellitus (type 1 or type 2), as defined by currently accepted criteria,266 or treatment with glucose lowering therapy.1
SPrior stroke, TIA, or arterial thromboembolismPrevious thromboembolism is associated with highly elevated risk of recurrence and therefore weighted 2 points.2
VVascular diseaseCoronary artery disease, including prior myocardial infarction, angina, history of coronary revascularization (surgical or percutaneous), and significant CAD on angiography or cardiac imaging.267
OR
Peripheral vascular disease, including: intermittent claudication, previous revascularization for PVD, percutaneous or surgical intervention on the abdominal aorta, and complex aortic plaque on imaging (defined as features of mobility, ulceration, pedunculation, or thickness ≥4 mm).268,269
1
AAge 65–74 years1 point is given for age between 65 and 74 years.1
CHA2DS2-VA componentDefinition and commentsPoints awardeda
CChronic heart failureSymptoms and signs of heart failure (irrespective of LVEF, thus including HFpEF, HFmrEF, and HFrEF), or the presence of asymptomatic LVEF ≤40%.261–2631
HHypertensionResting blood pressure >140/90 mmHg on at least two occasions, or current antihypertensive treatment. The optimal BP target associated with lowest risk of major cardiovascular events is 120–129/70–79 mmHg (or keep as low as reasonably achievable).162,2641
AAge 75 years or aboveAge is an independent determinant of ischaemic stroke risk.265 Age-related risk is a continuum, but for reasons of practicality, two points are given for age ≥75 years.2
DDiabetes mellitusDiabetes mellitus (type 1 or type 2), as defined by currently accepted criteria,266 or treatment with glucose lowering therapy.1
SPrior stroke, TIA, or arterial thromboembolismPrevious thromboembolism is associated with highly elevated risk of recurrence and therefore weighted 2 points.2
VVascular diseaseCoronary artery disease, including prior myocardial infarction, angina, history of coronary revascularization (surgical or percutaneous), and significant CAD on angiography or cardiac imaging.267
OR
Peripheral vascular disease, including: intermittent claudication, previous revascularization for PVD, percutaneous or surgical intervention on the abdominal aorta, and complex aortic plaque on imaging (defined as features of mobility, ulceration, pedunculation, or thickness ≥4 mm).268,269
1
AAge 65–74 years1 point is given for age between 65 and 74 years.1

BP, blood pressure; CAD, coronary artery disease; CHA2DS2-VA, chronic heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years; HFmrEF, heart failure with mildly reduced ejection fraction; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; LVEF, left ventricular ejection fraction; PVD, peripheral vascular disease.

aIn addition to these factors, other markers that modify an individual's risk for stroke and thromboembolism should be considered, including cancer, chronic kidney disease, ethnicity (black, Hispanic, Asian), biomarkers (troponin and BNP), and in specific groups, atrial enlargement, hyperlipidaemia, smoking, and obesity.

© ESC 2024
Table 10

Updated definitions for the CHA2DS2-VA score

CHA2DS2-VA componentDefinition and commentsPoints awardeda
CChronic heart failureSymptoms and signs of heart failure (irrespective of LVEF, thus including HFpEF, HFmrEF, and HFrEF), or the presence of asymptomatic LVEF ≤40%.261–2631
HHypertensionResting blood pressure >140/90 mmHg on at least two occasions, or current antihypertensive treatment. The optimal BP target associated with lowest risk of major cardiovascular events is 120–129/70–79 mmHg (or keep as low as reasonably achievable).162,2641
AAge 75 years or aboveAge is an independent determinant of ischaemic stroke risk.265 Age-related risk is a continuum, but for reasons of practicality, two points are given for age ≥75 years.2
DDiabetes mellitusDiabetes mellitus (type 1 or type 2), as defined by currently accepted criteria,266 or treatment with glucose lowering therapy.1
SPrior stroke, TIA, or arterial thromboembolismPrevious thromboembolism is associated with highly elevated risk of recurrence and therefore weighted 2 points.2
VVascular diseaseCoronary artery disease, including prior myocardial infarction, angina, history of coronary revascularization (surgical or percutaneous), and significant CAD on angiography or cardiac imaging.267
OR
Peripheral vascular disease, including: intermittent claudication, previous revascularization for PVD, percutaneous or surgical intervention on the abdominal aorta, and complex aortic plaque on imaging (defined as features of mobility, ulceration, pedunculation, or thickness ≥4 mm).268,269
1
AAge 65–74 years1 point is given for age between 65 and 74 years.1
CHA2DS2-VA componentDefinition and commentsPoints awardeda
CChronic heart failureSymptoms and signs of heart failure (irrespective of LVEF, thus including HFpEF, HFmrEF, and HFrEF), or the presence of asymptomatic LVEF ≤40%.261–2631
HHypertensionResting blood pressure >140/90 mmHg on at least two occasions, or current antihypertensive treatment. The optimal BP target associated with lowest risk of major cardiovascular events is 120–129/70–79 mmHg (or keep as low as reasonably achievable).162,2641
AAge 75 years or aboveAge is an independent determinant of ischaemic stroke risk.265 Age-related risk is a continuum, but for reasons of practicality, two points are given for age ≥75 years.2
DDiabetes mellitusDiabetes mellitus (type 1 or type 2), as defined by currently accepted criteria,266 or treatment with glucose lowering therapy.1
SPrior stroke, TIA, or arterial thromboembolismPrevious thromboembolism is associated with highly elevated risk of recurrence and therefore weighted 2 points.2
VVascular diseaseCoronary artery disease, including prior myocardial infarction, angina, history of coronary revascularization (surgical or percutaneous), and significant CAD on angiography or cardiac imaging.267
OR
Peripheral vascular disease, including: intermittent claudication, previous revascularization for PVD, percutaneous or surgical intervention on the abdominal aorta, and complex aortic plaque on imaging (defined as features of mobility, ulceration, pedunculation, or thickness ≥4 mm).268,269
1
AAge 65–74 years1 point is given for age between 65 and 74 years.1

BP, blood pressure; CAD, coronary artery disease; CHA2DS2-VA, chronic heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years; HFmrEF, heart failure with mildly reduced ejection fraction; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; LVEF, left ventricular ejection fraction; PVD, peripheral vascular disease.

aIn addition to these factors, other markers that modify an individual's risk for stroke and thromboembolism should be considered, including cancer, chronic kidney disease, ethnicity (black, Hispanic, Asian), biomarkers (troponin and BNP), and in specific groups, atrial enlargement, hyperlipidaemia, smoking, and obesity.

© ESC 2024
Recommendation Table 6

Recommendations to assess and manage thromboembolic risk in AF (see also Evidence Table 6)

Recommendation Table 6

Recommendations to assess and manage thromboembolic risk in AF (see also Evidence Table 6)

6.2. Oral anticoagulants

Vitamin K antagonists (VKA), predominantly warfarin but also other coumarin and indandione derivatives, have been the principal drugs to prevent thromboembolic events in the context of AF. As with any anticoagulant, a balance must be reached between preventing thromboembolism and preserving physiological haemostasis, with VKA-associated intracranial and other major haemorrhage the most critical limitation for acceptance of OAC. The global switch to DOACs as first-line therapy has changed this risk–benefit balance, allowing more widespread prescription with no need for routine monitoring (see Supplementary data online, Additional Evidence Tables S5–S7). This component of AF management may see substantive changes in the coming years, with a number of factor XI inhibitors in various stages of clinical evaluation. A phase 2 trial of abelacimab in patients with AF has shown lower rates of bleeding compared with rivaroxaban286; however, a phase 3 trial of asundexian was terminated early due to lack of efficacy against apixaban (NCT05643573), despite favourable phase 2 results.287 Regardless of the type of OAC prescribed, healthcare teams should be aware of the potential for interactions with other drugs, foods, and supplements, and incorporate this information into the education provided to patients and their carers. The list of potential interactions with VKA is broad,288,289 but there are also some common cardiovascular and non-cardiovascular drugs that interact with DOACs.290,291  Figure 9 highlights common and major interactions to consider for VKAs and DOACs.

Common drug interactions with oral anticoagulants.
Figure 9

Common drug interactions with oral anticoagulants.

INR, international normalized ratio of prothrombin time; NSAID, non-steroidal anti-inflammatory drug. This figure depicts only common or major interactions and is not an exhaustive list of all potential interactions. Please see the European Medicines Agency website or your local formulary for more information.

Recommendation Table 7

Recommendations for oral anticoagulation in AF (see also Evidence Table 7)

Recommendation Table 7

Recommendations for oral anticoagulation in AF (see also Evidence Table 7)

6.2.1. Direct oral anticoagulants

The DOACs (apixaban, dabigatran, edoxaban, and rivaroxaban) have all demonstrated at least non-inferior efficacy compared with warfarin for the prevention of thromboembolism, but with the added benefit of a 50% reduction in intracranial haemorrhage (ICH).25–28 Meta-analyses of individual data from 71 683 RCT patients showed that standard, full-dose DOAC treatment compared with warfarin reduces the risk of stroke or systemic embolism (HR, 0.81; 95% CI, 0.73–0.91), all-cause mortality (HR, 0.90; 95% CI, 0.85–0.95), and intracranial bleeding (HR, 0.48; 95% CI, 0.39–0.59), with no significant difference in other major bleeding (HR, 0.86; 95% CI, 0.73–1.00) and little or no between-trial heterogeneity.292 Post-marketing observational data on the effectiveness and safety of dabigatran,313,314 rivaroxaban,315,316 apixaban,317 and edoxaban318 vs. warfarin show general consistency with the respective phase 3 RCTs.

For patients undergoing cardioversion, three underpowered trials showed non-significantly lower rates of cardiovascular events with DOACs compared with warfarin.319–321 In meta-analysis of these 5203 patients predominantly undergoing electrical cardioversion, the composite of stroke, systemic embolism, myocardial infarction (MI), and cardiovascular death was significantly lower at 0.42% in patients randomized to a DOAC vs. 0.98% in those allocated VKA (risk ratio, 0.42; 95% CI, 0.21–0.86; P = .017), with no heterogeneity between trials and no significant difference in major bleeding.293

Specific patient subgroups show consistent benefit with DOACs vs. VKAs. For heart failure, major thromboembolic events were lower in DOAC-treated patients vs. warfarin in subgroup analysis of landmark RCTs,322 confirmed in large-scale real-world data.323 In a retrospective cohort of patients aged over 80 years, DOAC use was associated with a lower risk of ischaemic stroke, dementia, mortality, and major bleeding than warfarin,324 but this may be confounded by prescription bias.

Direct oral anticoagulants retain their efficacy and safety over VKAs in patients with mild-to-moderate CKD (creatinine clearance [CrCl] >30 mL/min),325 although specific dosing adjustments apply.25–28,326 In Europe, reduced doses of rivaroxaban, apixaban, and edoxaban are approved in patients with severe CKD (CrCl 15–29 mL/min), although limited numbers of patients were included in the major RCTs against VKA.327 Dabigatran is more dependent on renal elimination and so is contraindicated with an estimated glomerular filtration rate <30 mL/min/1.73 m2. Small trials have been performed in patients on haemodialysis, with two finding no difference between apixaban 2.5 mg twice daily and VKA for efficacy or safety outcomes,328,329 and one trial showing that rivaroxaban 10 mg led to significantly lower rates of cardiovascular events and major bleeding compared with VKA.330 Careful institution and regular follow-up are advised when instituting anticoagulants in any patient with impaired renal function (See Supplementary data online, Additional Evidence Table 8).326

Direct oral anticoagulants as a class should be avoided in specific patient groups, such as those with mechanical heart valves or moderate-to-severe mitral stenosis. In patients with mechanical heart valves, an excess of thromboembolic and major bleeding events among patients on dabigatran therapy vs. VKA was observed, with an RCT terminated prematurely.331 A trial of apixaban vs. VKA after implantation of a mechanical aortic valve was also stopped due to excess thromboembolic events in the apixaban group.332 The restriction on DOAC use does not apply to bioprosthetic heart valves (including mitral) or after transcatheter aortic valve implantation, where DOACs can be used and trial data show non-inferiority for clinical events compared with VKAs.304,333,334 With regards to mitral stenosis, the DOAC vs. VKA trials excluded patients with moderate-to-severe disease. In 4531 randomized patients with rheumatic heart disease and AF, VKAs led to a lower rate of composite cardiovascular events and death than rivaroxaban, without a higher rate of bleeding.294 Eighty-two per cent of the patients included had a mitral valve area ≤2 cm, supporting the restriction of DOAC use in patients with moderate-to-severe mitral stenosis. Note that patients with other types of valve disease (mitral regurgitation and others) should preferentially be prescribed a DOAC, and the term ‘valvular’ AF is obsolete and should be avoided.

Inappropriate dose reductions for DOACs are frequent in clinical practice,311 but need to be avoided as they increase the risk of stroke without decreasing bleeding risk.310 Hence, DOAC therapy should be instituted according to the standard full dose as tested in phase 3 RCTs and approved by regulators (Table 11). The prescribed dosage should consider the individual patient's profile.335 Drug interactions need to be considered in all patients taking or planned for DOACs (see Figure 9 for common drug interactions).336 There is insufficient evidence currently to advise on routine laboratory testing for DOAC levels. However, in certain situations, measurement of DOAC levels (where available) may be helpful, such as severe bleeding, the need for urgent surgery, or thromboembolic events despite apparent DOAC compliance.337,338 Patients should always be involved in decision-making on anticoagulation,339 leading to better alignment with personal preferences that can help to increase understanding and adherence.

Table 11

Recommended doses for direct oral anticoagulant therapy

DOACStandard full doseCriteria for dose reductionReduced dose only if criteria met
Apixaban5 mg twice dailyTwo out of three needed for dose reduction:
(i) age ≥80 years
(ii) body weight ≤60 kg
(iii) serum creatinine ≥133 mmol/L.
2.5 mg twice daily
Dabigatran150 mg twice dailyDose reduction recommended if any apply:
(i) age ≥80 years
(ii) receiving concomitant verapamil.
Dose reduction considered on an individual basis if any apply:
(i) age 75–80
(ii) moderate renal impairment (creatinine clearance 30–50 mL/min)
(iii) patients with gastritis, oesophagitis, or gastro-oesophageal reflux
(iv) others at increased risk of bleeding.
110 mg twice daily
Edoxaban60 mg once dailyDose reduction if any apply:
(i) moderate or severe renal impairment (creatinine clearance 15–50 mL/min)
(ii) body weight ≤60 kg
(iii) concomitant use of ciclosporin, dronedarone, erythromycin, or ketoconazole.
30 mg once daily
Rivaroxaban20 mg once dailyCreatinine clearance 15–49 mL/min.15 mg once daily
DOACStandard full doseCriteria for dose reductionReduced dose only if criteria met
Apixaban5 mg twice dailyTwo out of three needed for dose reduction:
(i) age ≥80 years
(ii) body weight ≤60 kg
(iii) serum creatinine ≥133 mmol/L.
2.5 mg twice daily
Dabigatran150 mg twice dailyDose reduction recommended if any apply:
(i) age ≥80 years
(ii) receiving concomitant verapamil.
Dose reduction considered on an individual basis if any apply:
(i) age 75–80
(ii) moderate renal impairment (creatinine clearance 30–50 mL/min)
(iii) patients with gastritis, oesophagitis, or gastro-oesophageal reflux
(iv) others at increased risk of bleeding.
110 mg twice daily
Edoxaban60 mg once dailyDose reduction if any apply:
(i) moderate or severe renal impairment (creatinine clearance 15–50 mL/min)
(ii) body weight ≤60 kg
(iii) concomitant use of ciclosporin, dronedarone, erythromycin, or ketoconazole.
30 mg once daily
Rivaroxaban20 mg once dailyCreatinine clearance 15–49 mL/min.15 mg once daily

DOAC, direct oral anticoagulant.

Dose and dose adjustments are taken from the European Medicines Association Summary of Product Characteristics for each DOAC. There may be other patient-specific reasons for providing a reduced dose, but, in general, the standard full dose should be used to provide optimal prevention of thromboembolism related to AF. Note that antiplatelet agents should be stopped in most patients when commencing a DOAC (see Section 6.3). A number of drug interactions exist with each DOAC and should be taken into consideration (see Figure 9).

© ESC 2024
Table 11

Recommended doses for direct oral anticoagulant therapy

DOACStandard full doseCriteria for dose reductionReduced dose only if criteria met
Apixaban5 mg twice dailyTwo out of three needed for dose reduction:
(i) age ≥80 years
(ii) body weight ≤60 kg
(iii) serum creatinine ≥133 mmol/L.
2.5 mg twice daily
Dabigatran150 mg twice dailyDose reduction recommended if any apply:
(i) age ≥80 years
(ii) receiving concomitant verapamil.
Dose reduction considered on an individual basis if any apply:
(i) age 75–80
(ii) moderate renal impairment (creatinine clearance 30–50 mL/min)
(iii) patients with gastritis, oesophagitis, or gastro-oesophageal reflux
(iv) others at increased risk of bleeding.
110 mg twice daily
Edoxaban60 mg once dailyDose reduction if any apply:
(i) moderate or severe renal impairment (creatinine clearance 15–50 mL/min)
(ii) body weight ≤60 kg
(iii) concomitant use of ciclosporin, dronedarone, erythromycin, or ketoconazole.
30 mg once daily
Rivaroxaban20 mg once dailyCreatinine clearance 15–49 mL/min.15 mg once daily
DOACStandard full doseCriteria for dose reductionReduced dose only if criteria met
Apixaban5 mg twice dailyTwo out of three needed for dose reduction:
(i) age ≥80 years
(ii) body weight ≤60 kg
(iii) serum creatinine ≥133 mmol/L.
2.5 mg twice daily
Dabigatran150 mg twice dailyDose reduction recommended if any apply:
(i) age ≥80 years
(ii) receiving concomitant verapamil.
Dose reduction considered on an individual basis if any apply:
(i) age 75–80
(ii) moderate renal impairment (creatinine clearance 30–50 mL/min)
(iii) patients with gastritis, oesophagitis, or gastro-oesophageal reflux
(iv) others at increased risk of bleeding.
110 mg twice daily
Edoxaban60 mg once dailyDose reduction if any apply:
(i) moderate or severe renal impairment (creatinine clearance 15–50 mL/min)
(ii) body weight ≤60 kg
(iii) concomitant use of ciclosporin, dronedarone, erythromycin, or ketoconazole.
30 mg once daily
Rivaroxaban20 mg once dailyCreatinine clearance 15–49 mL/min.15 mg once daily

DOAC, direct oral anticoagulant.

Dose and dose adjustments are taken from the European Medicines Association Summary of Product Characteristics for each DOAC. There may be other patient-specific reasons for providing a reduced dose, but, in general, the standard full dose should be used to provide optimal prevention of thromboembolism related to AF. Note that antiplatelet agents should be stopped in most patients when commencing a DOAC (see Section 6.3). A number of drug interactions exist with each DOAC and should be taken into consideration (see Figure 9).

© ESC 2024

6.2.2. Vitamin K antagonists

Vitamin K antagonist therapy reduces stroke risk by 64% and mortality by 26% in patients with AF at elevated thromboembolic risk (mostly warfarin in trials, compared with placebo or no treatment).239 Vitamin K antagonists are still used in many patients worldwide, but prescriptions have declined sharply since the introduction of DOACs.340,341 Vitamin K antagonists are currently the only treatment option in AF patients with mechanical heart valves or moderate-to-severe mitral valve stenosis.294,331 The use of VKAs is not only limited by numerous drug and food interactions (Figure 9), but also a narrow therapeutic range. This requires frequent monitoring and dose adjustment according to the prothrombin time expressed as the international normalized ratio (INR).342 If the time in therapeutic range (TTR) is maintained for long periods (e.g. >70% with INR 2.0–3.0), then VKA can be effective for thromboembolic protection with an acceptable safety profile.295–297,343 However, VKAs are associated with higher rates of intracranial bleeding,299,300 and also higher rates of other types of bleeding compared with DOACs.83

In view of the potential safety benefits, switching from VKAs to a DOAC is justified where there are concerns about intracranial bleeding or for patient-choice reasons, and a switch is recommended where patients have failed to maintain an adequate TTR (<70%). This depends on patients fulfilling eligibility criteria for DOACs and should take into account other correctable reasons for poor INR control. There is limited data on switching OAC in older patients (≥75 years) with polypharmacy or other markers of frailty. A recent trial in this patient group prematurely stopped for futility showed that switching from VKAs to DOACs led to a higher primary outcome rate of major or clinically relevant non-major bleeding events compared with continuing with INR-guided VKA (17.8 vs. 10.5 per 100 patient-years, driven by non-major bleeds).309 Hence, in such patients who are clinically stable with good TTR, VKAs may be continued rather than switching to a DOAC after an open discussion with the patient and shared decision-making.

6.2.3. Clinical vs. device-detected subclinical AF

The known benefit of anticoagulation applies to clinical AF. Two RCTs have been published assessing the value of DOAC therapy in device-detected subclinical AF. The ARTESiA trial (Apixaban for the Reduction of Thromboembolism in Patients With Device-Detected Sub-Clinical Atrial Fibrillation) was completed with 4012 patients with device-detected subclinical AF and a mean follow-up of 3.5 years.282 The primary efficacy outcome of stroke or systemic embolism was significantly less in those randomized to apixaban compared with aspirin (HR, 0.63; 95% CI, 0.45–0.88; P = .007). In the intention-to-treat analysis, the primary safety outcome of major bleeding was higher with apixaban (HR, 1.36; 95% CI, 1.01–1.82; P = .04). The NOAH trial (Non-vitamin K Antagonist Oral Anticoagulants in Patients With Atrial High Rate Episodes) was stopped prematurely due to safety concerns and futility for the efficacy of edoxaban, and hence provides limited information.281 The analysis of 2536 patients with device-detected atrial high-rate episodes and a median follow-up of 21 months identified no difference in a composite of cardiovascular death, stroke, or embolism comparing edoxaban and placebo (HR, 0.81; 95% CI, 0.60–1.08; P = .15). Those randomized to edoxaban had a higher rate of the composite of death or major bleeding than placebo (HR, 1.31; 95% CI, 1.02–1.67; P = .03). Patients had a low burden of device-detected subclinical AF in both trials (median duration 1.5 h and 2.8 h, respectively), with lower rates of thromboembolism (around 1% per patient-year) than would be expected for an equivalent cohort of patients with clinical AF and a CHA2DS2-VASc score of 4.

Considering the trade-off between potential benefit and the risk of major bleeding, this task force concludes that DOAC therapy may be considered in subgroups of patients with asymptomatic device-detected subclinical AF who have high estimated stroke risk and an absence of major bleeding risk factors (see Section 6.7). The duration and burden of subclinical AF that could indicate potential benefit from OAC remains uncertain.344 Regardless of the initial decision on OAC, patients with subclinical AF should receive management and follow-up for all aspects of AF-CARE as the risk of developing clinical AF is high (6%–9% per year).

6.3. Antiplatelet drugs and combinations with anticoagulants

Antiplatelet drugs, such as aspirin and clopidogrel, are not an alternative to OAC. They should not be used for stroke prevention, and can lead to potential harm (especially among elderly patients with AF).345–347 In ACTIVE W (Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events), dual antiplatelet therapy (DAPT) with aspirin and clopidogrel was less effective than warfarin for the prevention of stroke, systemic embolism, MI, or vascular death (annual risk of events 5.6% vs. 3.9%, respectively; P = .0003), with similar rates of major bleeding.348 The AVERROES (Apixaban Versus Acetylsalicylic Acid to Prevent Stroke in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment) trial demonstrated a lower rate of stroke or systemic embolism with apixaban compared with aspirin (HR, 0.45; 95% CI, 0.32–0.62; P < .001), with no significant difference in major bleeding (there were 11 cases of intracranial bleeding with apixaban and 13 with aspirin).242

The combination of OAC with antiplatelet agents (especially aspirin) without an adequate indication occurs frequently in clinical practice (see Supplementary data online, Additional Evidence Table S9).349,350 Bleeding events are more common when antithrombotic agents are combined, and no clear benefit has been observed in terms of prevention of stroke or death.349 In general, combining antiplatelet drugs with anticoagulants (DOACs or VKAs) should only occur in selected patients with acute vascular disease (e.g. acute coronary syndromes; see Section 9.2). The combination of low-dose rivaroxaban (2.5 mg) with aspirin reduced the risk of stroke in patients with chronic vascular disease in a subanalysis of the COMPASS (Cardiovascular Outcomes for People Using Anticoagulation Strategies) trial,351,352 but this cannot be generalized to AF patients because those with an indication for full-dose anticoagulants were excluded.

Recommendation Table 8

Recommendations for combining antiplatelet drugs with anticoagulants for stroke prevention (see also Evidence Table 8)

Recommendation Table 8

Recommendations for combining antiplatelet drugs with anticoagulants for stroke prevention (see also Evidence Table 8)

6.4. Residual ischaemic stroke risk despite anticoagulation

Although OAC significantly reduces the risk of ischaemic stroke in patients with AF, there remains a residual risk.252,354 One-third of patients with AF presenting with an ischaemic stroke are already on anticoagulation,355 with heterogeneous aetiology.356 This may include non-AF-related competing stroke mechanisms (such as large artery and small vessel diseases), non-adherence to therapy, an inappropriately low dose of anticoagulant, or thromboembolism despite sufficient anticoagulation.357 Laboratory measurement of INR or DOAC levels may contribute to revealing an amenable cause of the stroke. Regardless of anticoagulation status, patients with ischaemic stroke are more likely to have cardiovascular risk factors.358 Many clinicians managing patients with an incident stroke despite taking anticoagulation will be tempted to switch their anticoagulant regimen. While there may be some advantage in switching from VKAs to DOACs for protection against future recurrent ischaemic or haemorrhagic stroke, this task force does not recommend routinely switching from one DOAC to another, or from a DOAC to a VKA, since this has no proven efficacy.252,356,359 There may be individual reasons for switching, including potential interactions with new drugs; however, there is no consistent data across countries that adherence or efficacy differs between once- and twice-daily approaches.360,361 Emerging, but observational evidence suggests that switching provides limited reduction in the risk of recurrent ischaemic stroke.252,356,359 The alternative strategy of adding antiplatelet therapy to OAC may lead to an increased risk of bleeding.356,359 Aside from thorough attention to underlying risk factors and comorbidities, the approach to management of patients with a stroke despite OAC remains a distinct challenge.

Recommendation Table 9

Recommendations for thromboembolism despite anticoagulation (see also Evidence Table 9)

Recommendation Table 9

Recommendations for thromboembolism despite anticoagulation (see also Evidence Table 9)

6.5. Percutaneous left atrial appendage occlusion

Percutaneous left atrial appendage occlusion (LAAO) is a device-based therapy that aims to prevent ischaemic stroke in patients with AF.362,363 In the VKA era, two RCTs compared warfarin with LAAO using the Watchman device. The 5-year pooled outcomes demonstrated a similar rate of the composite endpoint (cardiovascular or unexplained death, systemic embolism, and stroke) between the LAAO and warfarin arms. Those randomized to LAAO had significantly lower rates of haemorrhagic stroke and all-cause death, but also a 71% non-significant increase in ischaemic stroke and systemic embolism.364 With DOACs demonstrating similar rates of major bleeding to aspirin,242 warfarin in the control arms in these trials is no longer standard of care and hence the place of LAAO in current practice is unclear. The Amulet occluder is an alternative LAAO device which was non-inferior in an RCT to the Watchman device for safety events (procedure-related complications, death, or major bleeding) and thromboembolism.365 In the PRAGUE-17 trial, 402 AF patients were randomized to DOAC or LAAO (Watchman or Amulet), with non-inferiority reported for a broad composite primary endpoint of stroke, TIA, systemic embolism, cardiovascular death, major or non-major clinically relevant bleeding, and procedure/device-related complications.366,367 Larger trials368,369 are expected to provide more comprehensive data that can add to the current evidence base (see Supplementary data online, Additional Evidence Table S10).

Pending further RCTs (see Supplementary data online, Table S4), patients with a contraindication to all of the OAC options (the four DOACs and VKAs) have the most appropriate rationale for LAAO implantation, despite the paradox that the need for post-procedure antithrombotic treatment exposes the patient to a bleeding risk that may be equivalent to that of DOACs. Regulatory approvals based on RCT protocols suggest the need for 45 days of VKA plus aspirin after implantation, followed by 6 months of DAPT in patients with no major peri-device leaks, and then ongoing aspirin (see Supplementary data online, Figure S2).370–372 However, real-world practice is markedly different and also varied. Direct oral anticoagulant administration at full or reduced dose has been proposed as a treatment alternative to warfarin.373 Observational studies have also supported the use of antiplatelet therapy without associated increases in device-related thrombosis or stroke.374–376 In a propensity-matched comparison of patients receiving limited early OAC vs. antiplatelet treatment post-Watchman implantation, thromboembolic event rates and bleeding complications were similar.377 While waiting for solid RCT data (NCT03445949, NCT03568890),378 pertinent decisions on antithrombotic treatment are usually made on an individualized basis.379–381 Prevention of recurrent stroke, in addition to OAC, is another potential indication for LAAO. Only limited data are so far available from registries,382 with ongoing trials expected to provide more insight (NCT03642509, NCT05963698).

Left atrial appendage occlusion device implantation is associated with procedural risk including stroke, major bleeding, device-related thrombus, pericardial effusion, vascular complications, and death.362,383–385 Voluntary registries enrolling patients considered ineligible for OAC have reported low peri-procedural risk,372,376,386,387 although national registries report in-hospital major adverse event rates of 9.5% in centres performing 5–15 LAAO cases per year, and 5.6% performing 32–211 cases per year (P < .001).388 Registries with new-generation devices report a lower complication rate compared with RCT data.389,390 Device-related thrombi occur with an incidence of 1.7%–7.2% and are associated with a higher risk of ischaemic stroke.386,391–397 Their detection can be documented as late as 1 year post-implantation in one-fifth of patients, thus mandating a late ‘rule-out’ imaging approach.391 Likewise, follow-up screening for peri-device leaks is relevant, as small leaks (0–5 mm) are present in ∼25% and have been associated with higher thromboembolic and bleeding events during 1 year follow-up in a large observational registry of one particular device.398

Recommendation Table 10

Recommendations for percutaneous left atrial appendage occlusion (see also Evidence Table 10)

Recommendation Table 10

Recommendations for percutaneous left atrial appendage occlusion (see also Evidence Table 10)

6.6. Surgical left atrial appendage occlusion

Surgical occlusion or exclusion of the left atrial appendage (LAA) can contribute to stroke prevention in patients with AF undergoing cardiac surgery.399,400 The Left Atrial Appendage Occlusion Study (LAAOS III) randomized 4811 patients with AF to undergo or not undergo LAAO at the time of cardiac surgery for another indication. During a mean of 3.8 years follow-up, ischaemic stroke or systemic embolism occurred in 114 patients (4.8%) in the occlusion group and 168 (7.0%) in the control arm (HR, 0.67; 95% CI, 0.53–0.85; P = .001).401 The LAAOS III trial did not compare appendage occlusion with anticoagulation (77% of participants continued to receive OAC), and therefore, surgical LAA closure should be considered as an adjunct therapy to prevent thromboembolism in addition to anticoagulation in patients with AF.

There are no RCT data showing a beneficial effect on ischaemic stroke or systemic embolism in patients with AF undergoing LAAO during endoscopic or hybrid AF ablation. A meta-analysis of RCT and observational data showed no differences in stroke prevention or all-cause mortality when comparing LAA clipping during thoracoscopic AF ablation with percutaneous LAAO and catheter ablation.402 While the percutaneous LAAO/catheter ablation group showed a higher acute success rate, it was also associated with a higher risk of haemorrhage during the peri-operative period. In an observational study evaluating 222 AF patients undergoing LAA closure using a clipping device as a part of endoscopic or hybrid AF ablation, complete closure was achieved in 95% of patients.403 There were no intra-operative complications, and freedom from a combined endpoint of ischaemic stroke, haemorrhagic stroke, or TIA was 99.1% over 369 patient-years of follow-up. Trials evaluating the beneficial effect of surgical LAA closure in patients undergoing cardiac surgery but without a known history of AF are ongoing (NCT03724318, NCT02701062).404

There is a potential advantage for stand-alone epicardial over percutaneous LAA closure in patients with a contraindication for OAC, as there is no need for post-procedure anticoagulation after epicardial closure. Observational data show that stand-alone LAA closure using an epicardial clip is feasible and safe.405 A multidisciplinary team approach can facilitate the choice between epicardial or percutaneous LAA closure in such patients.406 The majority of safety data and experience in epicardial LAA closure originate from a single clipping device (AtriClip)403,407,408 (see Supplementary data online, Additional Evidence Table S11).

Recommendation Table 11

Recommendations for surgical left atrial appendage occlusion (see also Evidence Table 11)

Recommendation Table 11

Recommendations for surgical left atrial appendage occlusion (see also Evidence Table 11)

6.7. Bleeding risk

6.7.1. Assessment of bleeding risk

When initiating antithrombotic therapy, modifiable bleeding risk factors should be managed to improve safety (Figure 10).414–418 This includes strict control of hypertension, advice to reduce excess alcohol intake, avoidance of unnecessary antiplatelet or anti-inflammatory agents, and attention to OAC therapy (adherence, control of TTR if on VKAs, and review of interacting medications). Clinicians should consider the balance between stroke and bleeding risk—as factors for both are dynamic and overlapping, they should be re-assessed at each review depending on the individual patient.419–421 Bleeding risk factors are rarely a reason to withdraw or withhold OAC in eligible patients, as the risk of stroke without anticoagulation often outweighs the risk of major bleeding.422,423 Patients with non-modifiable risk factors should be reviewed more often, and where appropriate, a multidisciplinary team approach should be instituted to guide management.

Modifying the risk of bleeding associated with OAC.
Figure 10

Modifying the risk of bleeding associated with OAC.

DOAC, direct oral anticoagulant; GI, gastrointestinal; INR, international normalized ratio of prothrombin time; NSAID, non-steroidal anti-inflammatory drug; OAC, oral anticoagulant; TTR, time in therapeutic range; VKA, vitamin K antagonist. aAbsolute contraindications for OAC therapy are rare, and include primary intracranial tumours and intracerebral bleeds related to amyloid angiopathy. In most cases, contraindications may be relative or temporary. Left atrial appendage occlusion can be performed through a percutaneous or endoscopic approach.

Several bleeding risk scores have been developed to account for a wide range of clinical factors (see Supplementary data online, Table S5 and Additional Evidence Tables S12 and S13).424 Systematic reviews and validation studies in external cohorts have shown contrasting results and only modest predictive ability.244,425–434 This task force does not recommend a specific bleeding risk score given the uncertainty in accuracy and potential adverse implications of not providing appropriate OAC to those at thromboembolic risk. There are very few absolute contraindications to OAC (especially DOAC therapy). Whereas primary intracranial tumours435 or an intracerebral bleed related to cerebral amyloid angiopathy436 are examples where OAC should be avoided, many other contraindications are relative or temporary. For example, a DOAC can often be safely initiated or re-initiated after acute bleeding has stopped, as long as the source has been fully investigated and managed. Co-prescription of proton pump inhibitors is common in clinical practice for patients receiving OAC that are at high risk of gastrointestinal bleeding. However, the evidence base is limited and not specifically in patients with AF. Whereas observational studies have shown potential benefit from proton pump inhibitors,437 a large RCT in patients receiving low-dose anticoagulation and/or aspirin for stable cardiovascular disease found that pantoprazole had no significant impact on upper gastrointestinal bleeding events compared with placebo (HR, 0.88; 95% CI, 0.67–1.15).438 Hence, the use of gastric protection should be individualized for each patient according to the totality of their perceived bleeding risk.

Recommendation Table 12

Recommendations for assessment of bleeding risk (see also Evidence Table 12)

Recommendation Table 12

Recommendations for assessment of bleeding risk (see also Evidence Table 12)

6.7.2. Management of bleeding on anticoagulant therapy

General management of bleeding in patients receiving OAC is outlined in Figure 11. Cause-specific management is beyond the scope of these guidelines, and will depend on the individual circumstances of the patient and the healthcare environment.447 Assessment of patients with active bleeding should include confirmation of the bleeding site, bleeding severity, type/dose/timepoint of last anticoagulant intake, concomitant use of other antithrombotic agents, and other factors influencing bleeding risk (renal function, platelet count, and medications such as non-steroidal anti-inflammatories). INR testing and information on recent results are invaluable for patients taking VKAs. Specific coagulation tests for DOACs include diluted thrombin time, ecarin clotting time, ecarin chromogenic assay for dabigatran, and chromogenic anti-factor Xa assay for rivaroxaban, apixaban, and edoxaban.447–449 Diagnostic and treatment interventions to identify and manage the cause of bleeding (e.g. gastroscopy) should be performed promptly.

Management of oral anticoagulant-related bleeding in patients with AF.
Figure 11

Management of oral anticoagulant-related bleeding in patients with AF.

DOAC, direct oral anticoagulant; FFP, fresh frozen plasma; INR, international normalized ratio of prothrombin time; OAC, oral anticoagulant; PCC, prothrombin complex concentrate; VKA, vitamin K antagonist.

In cases of minor bleeding, temporary withdrawal of OAC while the cause is managed is usually sufficient, noting that the reduction in anticoagulant effect is dependent on the INR level for VKAs or the half-life of the particular DOAC.

For major bleeding events in patients taking VKAs, administration of fresh frozen plasma restores coagulation more rapidly than vitamin K, but prothrombin complex concentrates achieve even faster blood coagulation with fewer complications, and so are preferrable to achieve haemostasis.450 In DOAC-treated patients where the last DOAC dose was taken within 2–4 h, charcoal administration and/or gastric lavage may reduce further exposure. If the patient is taking dabigatran, idarucizumab can fully reverse its anticoagulant effect and help to achieve haemostasis within 2–4 h in uncontrolled bleeding.451 Dialysis can also be effective in reducing dabigatran concentration. Andexanet alfa rapidly reverses the activity of factor Xa inhibitors (apixaban, edoxaban, rivaroxaban) (see Supplementary data online, Additional evidence Table S14). An open-label RCT comparing andexanet alfa to usual care in patients presenting with acute ICH within 6 h of symptom onset was stopped early due to improved control of bleeding after 450 patients had been randomized.452 As DOAC-specific antidotes are not yet available in all institutions, prothrombin complex concentrates are often used in cases of serious bleeding on factor Xa inhibitors, with evidence limited to observational studies.453

Due to the complexities of managing bleeding in patients taking OAC, it is advisable that each institution develop specific policies involving a multidisciplinary team that includes cardiologists, haematologists, emergency physicians/intensive care specialists, surgeons, and others. It is also important to educate patients taking anticoagulants on the signs and symptoms of bleeding events and to alert their healthcare provider when such events occur.335

The decision to reinstate OAC will be determined by the severity, cause, and subsequent management of bleeding, preferably by a multidisciplinary team and with close monitoring. Failure to reinstitute OAC after a bleed significantly increases the risk of MI, stroke, and death.454 However, if the cause of severe or life-threatening bleeds cannot be treated or reversed, the risk of ongoing bleeding may outweigh the benefit of thromboembolic protection.335

Recommendation Table 13

Recommendations for management of bleeding in anticoagulated patients (see also Evidence Table 13)

Recommendation Table 13

Recommendations for management of bleeding in anticoagulated patients (see also Evidence Table 13)

7. [R] Reduce symptoms by rate and rhythm control

Most patients diagnosed with AF will need therapies and/or interventions to control heart rate, revert to sinus rhythm, or maintain sinus rhythm to limit symptoms or improve outcomes. While the concept of choosing between rate and rhythm control is often discussed, in reality most patients require a combination approach which should be consciously re-evaluated during follow-up. Within a patient-centred and shared-management approach, rhythm control should be a consideration in all suitable AF patients, with explicit discussion of benefits and risks.

7.1. Management of heart rate in patients with AF

Limiting tachycardia is an integral part of AF management and is often sufficient to improve AF-related symptoms. Rate control is indicated as initial therapy in the acute setting, in combination with rhythm control therapies, or as the sole treatment strategy to control heart rate and reduce symptoms. Limited evidence exists to inform the best type and intensity of rate control treatment.457 The approach to heart rate control presented in Figure 7 can be used for all types of AF, including paroxysmal, persistent, and permanent AF.

Recommendation Table 14

Recommendations for heart rate control in patients with AF (see also Evidence Table 14)

Recommendation Table 14

Recommendations for heart rate control in patients with AF (see also Evidence Table 14)

7.1.1. Indications and target heart rate

The optimal heart rate target in AF patients depends on the setting, symptom burden, presence of heart failure, and whether rate control is combined with a rhythm control strategy. In the RACE II (Rate Control Efficacy in Permanent Atrial Fibrillation) RCT of patients with permanent AF, lenient rate control (target heart rate <110 [beats per minute] b.p.m.) was non-inferior to a strict approach (<80 b.p.m. at rest; <110 b.p.m. during exercise; Holter for safety) for a composite of clinical events, NYHA class, or hospitalization.186,459 Similar results were found in a post-hoc combined analysis from the AFFIRM (Atrial Fibrillation Follow-up Investigation of Rhythm Management) and the RACE (Rate Control versus Electrical cardioversion) studies.474 Therefore, lenient rate control is an acceptable initial approach, unless there are ongoing symptoms or suspicion of tachycardia-induced cardiomyopathy, where stricter targets may be indicated.

7.1.2. Heart rate control in the acute setting

In acute settings, physicians should always evaluate and manage underlying causes for the initiation of AF prior to, or in parallel to, instituting acute rate and/or rhythm control. These include treating sepsis, addressing fluid overload, or managing cardiogenic shock. The choice of drug (Table 12) will depend on the patient's characteristics, presence of heart failure and LVEF, and haemodynamic profile (Figure 7). In general for acute rate control, beta-blockers (for all LVEF) and diltiazem/verapamil (for LVEF >40%) are preferred over digoxin because of their more rapid onset of action and dose-dependent effects.462,475,476 More selective beta-1 receptor blockers have a better efficacy and safety profile than unselective beta-blockers.477 Combination therapy with digoxin may be required in acute settings (combination of beta-blockers with diltiazem/verapamil should be avoided except in closely monitored situations).177,478 In selected patients who are haemodynamically unstable or with severely impaired LVEF, intravenous amiodarone, landiolol, or digoxin can be used.472,473,479

7.1.3. Long-term heart rate control

Pharmacological rate control can be achieved with beta-blockers, diltiazem, verapamil, digoxin, or combination therapy (Table 12) (see Supplementary data online, Additional Evidence Table S15).480

Table 12

Drugs for rate control in AF

AgentaIntravenous administrationUsual range for oral maintenance doseContraindicated
Beta-blockersb
Metoprolol tartrate2.5–5 mg bolus over 2 mins; up to 15 mg maximal cumulative dose25–100 mg twice dailyIn case of asthma, non-selective beta-blockers should be avoided. Contraindicated in acute HF and history of severe bronchospasm.
Metoprolol XL (succinate)N/A50–200 mg once daily
BisoprololN/A1.25–20 mg once daily
AtenololcN/A25–100 mg once daily
Esmolol500 µg/kg i.v. bolus over 1 min; followed by 50–300 µg/kg/minN/A
Landiolol100 µg/kg i.v. bolus over 1 min; followed by 10–40 µg/kg/minN/A
NebivololN/A2.5–10 mg once daily
CarvedilolN/A3.125–50 mg twice daily
Non-dihydropyridine calcium channel antagonists
Verapamil2.5–10 mg i.v. bolus over 5 min40 mg twice daily to 480 mg (extended release) once dailyContraindicated if LVEF ≤40%. Adapt doses in hepatic and renal impairment.
Diltiazem0.25 mg/kg i.v. bolus over 5 min, then 5–15 mg/h60 mg three times daily to 360 mg (extended release) once daily
Digitalis glycosides
Digoxin0.5 mg i.v. bolus (0.75–1.5 mg over 24 h in divided doses)0.0625–0.25 mg once dailyHigh plasma levels associated with adverse events.
Check renal function before starting digoxin and adapt dose in CKD patients.
Digitoxin0.4–0.6 mg0.05–0.1 mg once daily
Other
Amiodaroned300 mg i.v. diluted in 250 mL 5% dextrose over 30–60 min (preferably via central venous cannula), followed by 900–1200 mg i.v. over 24 h diluted in 500–1000 mL via a central venous cannula200 mg once daily after loading
Loading: 200 mg three times daily for 4 weeks, then 200 mg daily or less as appropriate (reduce other rate control drugs according to heart rate)
Contraindicated in iodine sensitivity.
Serious potential adverse effects (including pulmonary, ophthalmic, hepatic, and thyroid). Consider numerous drug interactions.
AgentaIntravenous administrationUsual range for oral maintenance doseContraindicated
Beta-blockersb
Metoprolol tartrate2.5–5 mg bolus over 2 mins; up to 15 mg maximal cumulative dose25–100 mg twice dailyIn case of asthma, non-selective beta-blockers should be avoided. Contraindicated in acute HF and history of severe bronchospasm.
Metoprolol XL (succinate)N/A50–200 mg once daily
BisoprololN/A1.25–20 mg once daily
AtenololcN/A25–100 mg once daily
Esmolol500 µg/kg i.v. bolus over 1 min; followed by 50–300 µg/kg/minN/A
Landiolol100 µg/kg i.v. bolus over 1 min; followed by 10–40 µg/kg/minN/A
NebivololN/A2.5–10 mg once daily
CarvedilolN/A3.125–50 mg twice daily
Non-dihydropyridine calcium channel antagonists
Verapamil2.5–10 mg i.v. bolus over 5 min40 mg twice daily to 480 mg (extended release) once dailyContraindicated if LVEF ≤40%. Adapt doses in hepatic and renal impairment.
Diltiazem0.25 mg/kg i.v. bolus over 5 min, then 5–15 mg/h60 mg three times daily to 360 mg (extended release) once daily
Digitalis glycosides
Digoxin0.5 mg i.v. bolus (0.75–1.5 mg over 24 h in divided doses)0.0625–0.25 mg once dailyHigh plasma levels associated with adverse events.
Check renal function before starting digoxin and adapt dose in CKD patients.
Digitoxin0.4–0.6 mg0.05–0.1 mg once daily
Other
Amiodaroned300 mg i.v. diluted in 250 mL 5% dextrose over 30–60 min (preferably via central venous cannula), followed by 900–1200 mg i.v. over 24 h diluted in 500–1000 mL via a central venous cannula200 mg once daily after loading
Loading: 200 mg three times daily for 4 weeks, then 200 mg daily or less as appropriate (reduce other rate control drugs according to heart rate)
Contraindicated in iodine sensitivity.
Serious potential adverse effects (including pulmonary, ophthalmic, hepatic, and thyroid). Consider numerous drug interactions.

AF, atrial fibrillation; CKD, chronic kidney disease; HF, heart failure; i.v., intravenous; min, minutes; N/A, not available or not widely available. Maximum doses have been defined based on the summary of product characteristic of each drug.

aAll rate control drugs are contraindicated in Wolff–Parkinson–White syndrome; also intravenous amiodarone.

bOther beta-blockers are available but not recommended as specific rate control therapy in AF and therefore not mentioned here (e.g. propranolol and labetalol).

cNo data on atenolol; should not be used in heart failure with reduced ejection fraction or in pregnancy.

dLoading regimen may vary; i.v. dosage should be considered when calculating total load.

© ESC 2024
Table 12

Drugs for rate control in AF

AgentaIntravenous administrationUsual range for oral maintenance doseContraindicated
Beta-blockersb
Metoprolol tartrate2.5–5 mg bolus over 2 mins; up to 15 mg maximal cumulative dose25–100 mg twice dailyIn case of asthma, non-selective beta-blockers should be avoided. Contraindicated in acute HF and history of severe bronchospasm.
Metoprolol XL (succinate)N/A50–200 mg once daily
BisoprololN/A1.25–20 mg once daily
AtenololcN/A25–100 mg once daily
Esmolol500 µg/kg i.v. bolus over 1 min; followed by 50–300 µg/kg/minN/A
Landiolol100 µg/kg i.v. bolus over 1 min; followed by 10–40 µg/kg/minN/A
NebivololN/A2.5–10 mg once daily
CarvedilolN/A3.125–50 mg twice daily
Non-dihydropyridine calcium channel antagonists
Verapamil2.5–10 mg i.v. bolus over 5 min40 mg twice daily to 480 mg (extended release) once dailyContraindicated if LVEF ≤40%. Adapt doses in hepatic and renal impairment.
Diltiazem0.25 mg/kg i.v. bolus over 5 min, then 5–15 mg/h60 mg three times daily to 360 mg (extended release) once daily
Digitalis glycosides
Digoxin0.5 mg i.v. bolus (0.75–1.5 mg over 24 h in divided doses)0.0625–0.25 mg once dailyHigh plasma levels associated with adverse events.
Check renal function before starting digoxin and adapt dose in CKD patients.
Digitoxin0.4–0.6 mg0.05–0.1 mg once daily
Other
Amiodaroned300 mg i.v. diluted in 250 mL 5% dextrose over 30–60 min (preferably via central venous cannula), followed by 900–1200 mg i.v. over 24 h diluted in 500–1000 mL via a central venous cannula200 mg once daily after loading
Loading: 200 mg three times daily for 4 weeks, then 200 mg daily or less as appropriate (reduce other rate control drugs according to heart rate)
Contraindicated in iodine sensitivity.
Serious potential adverse effects (including pulmonary, ophthalmic, hepatic, and thyroid). Consider numerous drug interactions.
AgentaIntravenous administrationUsual range for oral maintenance doseContraindicated
Beta-blockersb
Metoprolol tartrate2.5–5 mg bolus over 2 mins; up to 15 mg maximal cumulative dose25–100 mg twice dailyIn case of asthma, non-selective beta-blockers should be avoided. Contraindicated in acute HF and history of severe bronchospasm.
Metoprolol XL (succinate)N/A50–200 mg once daily
BisoprololN/A1.25–20 mg once daily
AtenololcN/A25–100 mg once daily
Esmolol500 µg/kg i.v. bolus over 1 min; followed by 50–300 µg/kg/minN/A
Landiolol100 µg/kg i.v. bolus over 1 min; followed by 10–40 µg/kg/minN/A
NebivololN/A2.5–10 mg once daily
CarvedilolN/A3.125–50 mg twice daily
Non-dihydropyridine calcium channel antagonists
Verapamil2.5–10 mg i.v. bolus over 5 min40 mg twice daily to 480 mg (extended release) once dailyContraindicated if LVEF ≤40%. Adapt doses in hepatic and renal impairment.
Diltiazem0.25 mg/kg i.v. bolus over 5 min, then 5–15 mg/h60 mg three times daily to 360 mg (extended release) once daily
Digitalis glycosides
Digoxin0.5 mg i.v. bolus (0.75–1.5 mg over 24 h in divided doses)0.0625–0.25 mg once dailyHigh plasma levels associated with adverse events.
Check renal function before starting digoxin and adapt dose in CKD patients.
Digitoxin0.4–0.6 mg0.05–0.1 mg once daily
Other
Amiodaroned300 mg i.v. diluted in 250 mL 5% dextrose over 30–60 min (preferably via central venous cannula), followed by 900–1200 mg i.v. over 24 h diluted in 500–1000 mL via a central venous cannula200 mg once daily after loading
Loading: 200 mg three times daily for 4 weeks, then 200 mg daily or less as appropriate (reduce other rate control drugs according to heart rate)
Contraindicated in iodine sensitivity.
Serious potential adverse effects (including pulmonary, ophthalmic, hepatic, and thyroid). Consider numerous drug interactions.

AF, atrial fibrillation; CKD, chronic kidney disease; HF, heart failure; i.v., intravenous; min, minutes; N/A, not available or not widely available. Maximum doses have been defined based on the summary of product characteristic of each drug.

aAll rate control drugs are contraindicated in Wolff–Parkinson–White syndrome; also intravenous amiodarone.

bOther beta-blockers are available but not recommended as specific rate control therapy in AF and therefore not mentioned here (e.g. propranolol and labetalol).

cNo data on atenolol; should not be used in heart failure with reduced ejection fraction or in pregnancy.

dLoading regimen may vary; i.v. dosage should be considered when calculating total load.

© ESC 2024

The choice of rate control drugs depends on symptoms, comorbidities, and the potential for side effects and interactions. Combination therapy of different rate-controlling drugs should be considered only when needed to achieve the target heart rate, and careful follow-up to avoid bradycardia is advised. Combining beta-blockers with verapamil or diltiazem should only be performed in secondary care with regular monitoring of heart rate by 24 h ECG to check for bradycardia.459 Some antiarrhythmic drugs (AADs) also have rate-limiting properties (e.g. amiodarone, sotalol), but they should generally be used only for rhythm control. Dronedarone should not be instituted for rate control since it increases rates of heart failure, stroke, and cardiovascular death in permanent AF.481

Beta-blockers, specifically beta-1 selective adrenoreceptor antagonists, are often first-line rate-controlling agents largely based on their acute effect on heart rate and the beneficial effects demonstrated in patients with chronic HFrEF. However, the prognostic benefit of beta-blockers seen in HFrEF patients with sinus rhythm may not be present in patients with AF.133,482

Verapamil and diltiazem are non-dihydropyridine calcium channel blockers. They provide rate control461 and have a different adverse effect profile, making verapamil or diltiazem useful for those experiencing side effects from beta-blockers.483 In a 60 patient crossover RCT, verapamil and diltiazem did not lead to the same reduction in exercise capacity as seen with beta-blockers, and had a beneficial impact on BNP.480

Digoxin and digitoxin are cardiac glycosides that inhibit the sodium–potassium adenosine triphosphatase and augment parasympathetic tone. In RCTs, there is no association between the use of digoxin and any increase in all-cause mortality.185,484 Lower doses of digoxin may be associated with better prognosis.185 Serum digoxin concentrations can be monitored to avoid toxicity,485 especially in patients at higher risk due to older age, renal dysfunction, or use of interacting medications. In RATE-AF (RAte control Therapy Evaluation in permanent Atrial Fibrillation), a trial in patients with symptomatic permanent AF, there was no difference between low-dose digoxin and bisoprolol for patient-reported quality of life outcomes at 6 months. However, those randomized to digoxin demonstrated fewer adverse effects, a greater improvement in mEHRA and NYHA scores, and a reduction in BNP.48 Two ongoing RCTs are addressing digoxin and digitoxin use in patients with HFrEF with and without AF (EudraCT-2013-005326-38, NCT03783429).486

Due to its broad extracardiac adverse effect profile, amiodarone is reserved as a last option when heart rate cannot be controlled even with maximal tolerated combination therapy, or in patients who do not qualify for atrioventricular node ablation and pacing. Many of the adverse effects from amiodarone have a direct relationship with cumulative dose, restricting the long-term value of amiodarone for rate control.487

7.1.4. Atrioventricular node ablation and pacemaker implantation

Ablation of the atrioventricular node and pacemaker implantation (‘ablate and pace’) can lower and regularize heart rate in patients with AF (see Supplementary data online, Additional Evidence Table S16). The procedure has a low complication rate and a low long-term mortality risk.468,488 The pacemaker should be implanted a few weeks before the atrioventricular node ablation, with the initial pacing rate after ablation set at 70–90 b.p.m.489,490 This strategy does not worsen LV function,491 and may even improve LVEF in selected patients.492,493 The evidence base has typically included older patients. For younger patients, ablate and pace should only be considered if heart rate remains uncontrolled despite consideration of other pharmacological and non-pharmacological treatment options. The choice of pacing therapy (right ventricular or biventricular pacing) depends on patient characteristics, presence of heart failure, and LVEF.187,494

In severely symptomatic patients with permanent AF and at least one hospitalization for heart failure, atrioventricular node ablation combined with CRT should be considered. In the APAF-CRT (Ablate and Pace for Atrial Fibrillation-cardiac resynchronization therapy) trial in a population with narrow QRS complexes, atrioventricular node ablation combined with CRT was superior to rate control drugs for the primary outcomes (all-cause mortality, and death or hospitalization for heart failure), and secondary outcomes (symptom burden and physical limitation).470,471 Conduction system pacing may become a potentially useful alternate pacing mode when implementing a pace and ablate strategy, once safety and efficacy have been confirmed in larger RCTs.495,496 In CRT recipients, the presence (or occurrence) of AF is one of the main reasons for suboptimal biventricular pacing.187 Improvement of biventricular pacing is indicated and can be reached by intensification of rate control drug regimens, atrioventricular node ablation, or rhythm control, depending on patient and AF characteristics.187

7.2. Rhythm control strategies in patients with AF

7.2.1. General principles and anticoagulation

Rhythm control refers to therapies dedicated to restoring and maintaining sinus rhythm. These treatments include cardioversion, AADs, percutaneous catheter ablation, endoscopic and hybrid ablation, and open surgical approaches (see Supplementary data online, Additional Evidence Table S17). Rhythm control is never a strategy on its own; instead, it should always be part of the AF-CARE approach.

In patients with acute or worsening haemodynamic instability thought to be caused by AF, rapid electrical cardioversion is recommended. For other patients, a wait-and-see approach should be considered as an alternative to immediate cardioversion (Figure 12). The Rate Control versus Electrical Cardioversion Trial 7–Acute Cardioversion versus Wait and See (RACE 7 ACWAS) trial in patients with recent-onset symptomatic AF without haemodynamic compromise showed a wait-and-see approach for spontaneous conversion until 48 h after the onset of AF symptoms was non-inferior as compared with immediate cardioversion at 4 weeks follow-up.10

Approaches for cardioversion in patients with AF.
Figure 12

Approaches for cardioversion in patients with AF.

AF, atrial fibrillation; CHA2DS2-VA, congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years; h, hour; LMWH, low molecular weight heparin; DOAC, direct oral anticoagulant; OAC, oral anticoagulant; TOE, transoesophageal echocardiography; UFH, unfractionated heparin; VKA, vitamin K antagonist. Flowchart for decision-making on cardioversion of AF depending on clinical presentation, AF onset, oral anticoagulation intake, and risk factors for stroke. aSee Section 6.

Since the publication of landmark trials more than 20 years ago, the main reason to consider longer-term rhythm control therapy has been the reduction in symptoms from AF.497–500 Older studies have shown that the institution of a rhythm control strategy using AADs does not reduce mortality and morbidity when compared with a rate control-only strategy,497–500 and may increase hospitalization.457 In contrast, multiple studies have shown that rhythm control strategies have a positive effect on quality of life once sinus rhythm is maintained.501,502 Therefore, in the case of uncertainty of the presence of symptoms associated with AF, an attempt to restore sinus rhythm is a rational first step. In patients with symptoms, patient factors that favour an attempt at rhythm control should be considered, including suspected tachycardiomyopathy, a brief AF history, non-dilated left atrium, or patient preference.

Rhythm control strategies have significantly evolved due to an increasing experience in the safe use of antiarrhythmic drugs,17 consistent use of OAC, improvements in ablation technology,503–509 and identification and management of risk factors and comorbidities.39,510,511 In the ATHENA trial (A Placebo-Controlled, Double-Blind, Parallel Arm Trial to Assess the Efficacy of Dronedarone 400 mg twice daily for the Prevention of Cardiovascular Hospitalization or Death from Any Cause in Patients with Atrial Fibrillation/Atrial Flutter), dronedarone significantly reduced the risk of hospitalization due to cardiovascular events or death as compared with placebo in patients with paroxysmal or persistent AF.512 The CASTLE-AF trial (Catheter Ablation versus Standard Conventional Treatment in Patients With Left Ventricle Dysfunction and AF) demonstrated that a rhythm control strategy with catheter ablation can improve mortality and morbidity in selected patients with HFrEF and an implanted cardiac device.4 In end-stage HFrEF, the CASTLE-HTx trial (Catheter Ablation for Atrial Fibrillation in Patients With End-Stage Heart Failure and Eligibility for Heart Transplantation) found, in a single centre, that catheter ablation combined with guideline-directed medical therapy significantly reduced the composite of death from any cause, implantation of left ventricular assist device, or urgent heart transplantation compared with medical treatment.513 At the same time, however, the CABANA trial (Catheter Ablation versus Anti-arrhythmic Drug Therapy for Atrial Fibrillation) could not demonstrate a significant difference in mortality and morbidity between catheter ablation and standard rhythm and/or rate control drugs in symptomatic AF patients older than 64 years, or younger than 65 years with risk factors for stroke.3 EAST-AFNET 4 (Early treatment of Atrial fibrillation for Stroke prevention Trial) reported that implementation of a rhythm control strategy within 1 year compared with usual care significantly reduced the risk of cardiovascular death, stroke, or hospitalization for heart failure or acute coronary syndrome in patients older than 75 years or with cardiovascular conditions.17 Of note, rhythm control was predominantly pursued with antiarrhythmic drugs (80% of patients in the intervention arm). Usual care consisted of rate control therapy; only when uncontrolled AF-related symptoms occurred was rhythm control considered. Patients in the EAST-AFNET 4 trial all had cardiovascular risk factors but were at an early stage of AF, with more than 50% being in sinus rhythm and 30% being asymptomatic at the start of the study.

Based on all of these studies, this task force concludes that implementation of a rhythm control strategy can be safely instituted and confers amelioration of AF-related symptoms. Beyond control of symptoms, sinus rhythm maintenance should also be pursued to reduce morbidity and mortality in selected groups of patients.4,17,502,513,514

Any rhythm control procedure has an inherent risk of thromboembolism. Patients undergoing cardioversion require at least 3 weeks of therapeutic anticoagulation (adherence to DOACs or INR >2 if VKA) prior to the electrical or pharmacological procedure. In acute settings or when early cardioversion is needed, transoesophageal echocardiography (TOE) can be performed to exclude cardiac thrombus prior to cardioversion. These approaches have been tested in multiple RCTs.319–321 In the case of thrombus detection, therapeutic anticoagulation should be instituted for a minimum of 4 weeks followed by repeat TOE to ensure thrombus resolution. When the definite duration of AF is less than 48 hours, cardioversion has typically been considered without the need for pre-procedure OAC or TOE for thrombus exclusion. However, the ‘definite’ onset of AF is often not known, and observational data suggest that stroke/thromboembolism risk is lowest within a much shorter time period.515–519 This task force reached consensus that safety should come first. Cardioversion is not recommended if AF duration is longer than 24 hours, unless the patient has already received at least 3 weeks of therapeutic anticoagulation or a TOE is performed to exclude intracardiac thrombus. Most patients should continue OAC for at least 4 weeks post-cardioversion. Only for those without thromboembolic risk factors and sinus rhythm restoration within 24 h of AF onset is post-cardioversion OAC optional. In the presence of any thromboembolic risk factors, long-term OAC should be instituted irrespective of the rhythm outcome.

Recommendation Table 15

Recommendations for general concepts in rhythm control (see also Evidence Table 15)

Recommendation Table 15

Recommendations for general concepts in rhythm control (see also Evidence Table 15)

7.2.2. Electrical cardioversion

Electrical cardioversion (ECV) can be safely applied in the elective and acute setting (see Supplementary data online, Additional Evidence Table S18) with sedation by intravenous midazolam, propofol, or etomidate.530 Structured and integrated care for patients with acute-onset AF at the emergency department is associated with better outcomes without compromising safety.531 Rates of major adverse clinical events after cardioversion are significantly lower with DOACs compared with warfarin.293

Blood pressure monitoring and oximetry should be used routinely. Intravenous atropine or isoproterenol, or temporary transcutaneous pacing, should be available in case of post-cardioversion bradycardia. Biphasic defibrillators are standard because of their superior efficacy compared with monophasic defibrillators.532–534 There is no single optimal position for electrodes, with a meta-analysis of 10 RCTs showing no difference in sinus rhythm restoration comparing anterior-posterior with antero-lateral electrode positioning.535 Applying active compression to the defibrillation pads is associated with lower defibrillation thresholds, lower total energy delivery, fewer shocks for successful ECV, and higher success rates.536 A randomized trial showed that maximum fixed-energy shocks were more effective than low-escalating energy for ECV.537

Immediate administration of vernakalant,538 or pre-treatment for 3–4 days with flecainide,539,540 ibutilide,541,542 propafenone,543 or amiodarone544–546 improves the rate of successful ECV and can facilitate long-term maintenance of sinus rhythm by preventing early recurrent AF.547 A meta-analysis demonstrated that pre-treatment with amiodarone (200–800 mg/day for 1–6 weeks pre-cardioversion) and post-treatment (200 mg/day) significantly improved the restoration and maintenance of sinus rhythm after ECV of AF.546

In some cases of persistent AF there is no clear relationship between the arrhythmia and symptoms. In these cases, restoring sinus rhythm by ECV might serve to confirm the impact of arrhythmia on symptoms and/or on heart failure symptoms and signs. Such an approach might be useful to identify truly asymptomatic individuals, to assess the impact of AF on LV function in patients with HFrEF, and to distinguish AF-related symptoms from heart failure symptoms.

Recommendation Table 16

Recommendations for electrical cardioversion of AF (see also Evidence Table 16)

Recommendation Table 16

Recommendations for electrical cardioversion of AF (see also Evidence Table 16)

7.2.3. Pharmacological cardioversion

Pharmacological cardioversion to sinus rhythm is an elective procedure in haemodynamically stable patients. It is less effective than electrical cardioversion for restoration of sinus rhythm,549 with timing of cardioversion being a significant determinant of success.550 There are limited contemporary data on the true efficacy of pharmacological cardioversion, which are likely biased by the spontaneous restoration of sinus rhythm in 76%–83% of patients with recent-onset AF (10%–18% within the first 3 h, 55%–66% within 24 h, and 69% within 48 h).10,119,445,551–555

The choice of a specific drug is based on the type and severity of concomitant heart disease (Table 13). A meta-analysis demonstrated that intravenous vernakalant and flecainide have the highest conversion rate within 4 h, possibly allowing discharge from the emergency department and reducing hospital admissions. Intravenous and oral formulations of Class IC antiarrhythmics (flecainide more so than propafenone) are superior regarding conversion rates within 12 h, while amiodarone efficacy is exhibited in a delayed fashion (within 24 h).556 Pharmacological cardioversion does not require fasting, sedation, or anaesthesia. Anticoagulation should be started or continued according to a formal (re-)assessment of thromboembolic risk.554,557–559

Table 13

Antiarrhythmic drugs for sinus rhythm restoration

DrugAdministration routeInitial dosingSubsequent dosing [long-term approach]Acute success rate and time to sinus rhythmContraindications and precautions
FlecainideOral200–300 mg[long-term 50−150 mg twice daily]50%–60% at 3 h and 75%–85% at 6–8 h (3–8 h)
  • Should not be used in patients with severe structural or coronary artery disease, Brugada syndrome, or severe renal failure (CrCl <35 mL/min/1.73 m2).

  • Prior documentation of safety and efficacy in an inpatient setting is recommended prior to pill-in-the-pocket use.

  • An AVN-blocking agent should be administered to avoid 1:1 conduction if transformation to AFL.

  • Drug infusion should be discontinued in case of QRS widening >25% or bundle branch block occurrence.

  • Caution is needed in patients with sinus node disease and AVN dysfunction.

  • Do NOT use for conversion of atrial flutter.

Intravenous1–2 mg/kg over 10 min52%–95% (Up to 6 h)
PropafenoneOral450–600 mg[long-term 150-300 mg three times daily]45%–55% at 3 h, 69%–78% at 8 h (3–8 h)
Intravenous1.5–2 mg/kg over 10 min43%–89% (Up to 6 h)
AmiodaroneIntravenous (/oral)300 mg intravenous over 30–60 min900-1200 mg intravenous over 24 hours (or 200 mg oral three times daily for 4 weeks).[long-term 200 mg oral daily]44% (8–12 h to several days)
  • May cause phlebitis (use a large peripheral vein, avoid i.v. administration >24 h and use preferably volumetric pump).

  • May cause hypotension, bradycardia/atrioventricular block, QT prolongation.

  • Only if no other option in patients with hyperthyroidism (risk of thyrotoxicosis).

  • Consider the broad range of drug interactions.

IbutilideIntravenous1 mg over 10 min (0.01 mg/kg if body weight <60 kg)1 mg over 10 min
(10–20 min after the initial dose)
31%–51% (30–90 min) in AF
60–75% in AFL (60 min)
  • Should be used in the setting of a cardiac care unit as it may cause QT prolongation and torsades de pointes.

  • ECG monitoring for at least 4 h after administration to detect any proarrhythmic effects.

  • Should not be used in patients with prolonged QT, severe LVH, or low LVEF.

VernakalantIntravenous3 mg/kg over 10 min (maximum 339 mg)2 mg/kg over 10 min
(10–15 min after the initial dose) (maximum 226 mg)
50% within 10 min
  • Should not be used in patients with arterial hypotension (SBP <100 mmHg), recent ACS (within 1 month), NYHA III or IV HF, QT prolongation or severe aortic stenosis.

  • May cause arterial hypotension, QT prolongation, QRS widening, or non-sustained ventricular tachycardia.

DrugAdministration routeInitial dosingSubsequent dosing [long-term approach]Acute success rate and time to sinus rhythmContraindications and precautions
FlecainideOral200–300 mg[long-term 50−150 mg twice daily]50%–60% at 3 h and 75%–85% at 6–8 h (3–8 h)
  • Should not be used in patients with severe structural or coronary artery disease, Brugada syndrome, or severe renal failure (CrCl <35 mL/min/1.73 m2).

  • Prior documentation of safety and efficacy in an inpatient setting is recommended prior to pill-in-the-pocket use.

  • An AVN-blocking agent should be administered to avoid 1:1 conduction if transformation to AFL.

  • Drug infusion should be discontinued in case of QRS widening >25% or bundle branch block occurrence.

  • Caution is needed in patients with sinus node disease and AVN dysfunction.

  • Do NOT use for conversion of atrial flutter.

Intravenous1–2 mg/kg over 10 min52%–95% (Up to 6 h)
PropafenoneOral450–600 mg[long-term 150-300 mg three times daily]45%–55% at 3 h, 69%–78% at 8 h (3–8 h)
Intravenous1.5–2 mg/kg over 10 min43%–89% (Up to 6 h)
AmiodaroneIntravenous (/oral)300 mg intravenous over 30–60 min900-1200 mg intravenous over 24 hours (or 200 mg oral three times daily for 4 weeks).[long-term 200 mg oral daily]44% (8–12 h to several days)
  • May cause phlebitis (use a large peripheral vein, avoid i.v. administration >24 h and use preferably volumetric pump).

  • May cause hypotension, bradycardia/atrioventricular block, QT prolongation.

  • Only if no other option in patients with hyperthyroidism (risk of thyrotoxicosis).

  • Consider the broad range of drug interactions.

IbutilideIntravenous1 mg over 10 min (0.01 mg/kg if body weight <60 kg)1 mg over 10 min
(10–20 min after the initial dose)
31%–51% (30–90 min) in AF
60–75% in AFL (60 min)
  • Should be used in the setting of a cardiac care unit as it may cause QT prolongation and torsades de pointes.

  • ECG monitoring for at least 4 h after administration to detect any proarrhythmic effects.

  • Should not be used in patients with prolonged QT, severe LVH, or low LVEF.

VernakalantIntravenous3 mg/kg over 10 min (maximum 339 mg)2 mg/kg over 10 min
(10–15 min after the initial dose) (maximum 226 mg)
50% within 10 min
  • Should not be used in patients with arterial hypotension (SBP <100 mmHg), recent ACS (within 1 month), NYHA III or IV HF, QT prolongation or severe aortic stenosis.

  • May cause arterial hypotension, QT prolongation, QRS widening, or non-sustained ventricular tachycardia.

ACS, acute coronary syndromes; AF, atrial fibrillation; AFL, atrial flutter; AVN, atrioventricular node; CrCl, creatinine clearance; ECG, electrocardiogram; HF, heart failure; LVEF, left ventricular ejection fraction; LVH, left ventricular hypertrophy; NYHA, New York Heart Association; QT, QT interval; SBP, systolic blood pressure. Long-term dosage for maintenance of sinus rhythm is indicated in [square brackets]. Long-term oral dosing for dronedarone is 400 mg twice daily, and for sotalol 80-160 mg twice daily.

© ESC 2024
Table 13

Antiarrhythmic drugs for sinus rhythm restoration

DrugAdministration routeInitial dosingSubsequent dosing [long-term approach]Acute success rate and time to sinus rhythmContraindications and precautions
FlecainideOral200–300 mg[long-term 50−150 mg twice daily]50%–60% at 3 h and 75%–85% at 6–8 h (3–8 h)
  • Should not be used in patients with severe structural or coronary artery disease, Brugada syndrome, or severe renal failure (CrCl <35 mL/min/1.73 m2).

  • Prior documentation of safety and efficacy in an inpatient setting is recommended prior to pill-in-the-pocket use.

  • An AVN-blocking agent should be administered to avoid 1:1 conduction if transformation to AFL.

  • Drug infusion should be discontinued in case of QRS widening >25% or bundle branch block occurrence.

  • Caution is needed in patients with sinus node disease and AVN dysfunction.

  • Do NOT use for conversion of atrial flutter.

Intravenous1–2 mg/kg over 10 min52%–95% (Up to 6 h)
PropafenoneOral450–600 mg[long-term 150-300 mg three times daily]45%–55% at 3 h, 69%–78% at 8 h (3–8 h)
Intravenous1.5–2 mg/kg over 10 min43%–89% (Up to 6 h)
AmiodaroneIntravenous (/oral)300 mg intravenous over 30–60 min900-1200 mg intravenous over 24 hours (or 200 mg oral three times daily for 4 weeks).[long-term 200 mg oral daily]44% (8–12 h to several days)
  • May cause phlebitis (use a large peripheral vein, avoid i.v. administration >24 h and use preferably volumetric pump).

  • May cause hypotension, bradycardia/atrioventricular block, QT prolongation.

  • Only if no other option in patients with hyperthyroidism (risk of thyrotoxicosis).

  • Consider the broad range of drug interactions.

IbutilideIntravenous1 mg over 10 min (0.01 mg/kg if body weight <60 kg)1 mg over 10 min
(10–20 min after the initial dose)
31%–51% (30–90 min) in AF
60–75% in AFL (60 min)
  • Should be used in the setting of a cardiac care unit as it may cause QT prolongation and torsades de pointes.

  • ECG monitoring for at least 4 h after administration to detect any proarrhythmic effects.

  • Should not be used in patients with prolonged QT, severe LVH, or low LVEF.

VernakalantIntravenous3 mg/kg over 10 min (maximum 339 mg)2 mg/kg over 10 min
(10–15 min after the initial dose) (maximum 226 mg)
50% within 10 min
  • Should not be used in patients with arterial hypotension (SBP <100 mmHg), recent ACS (within 1 month), NYHA III or IV HF, QT prolongation or severe aortic stenosis.

  • May cause arterial hypotension, QT prolongation, QRS widening, or non-sustained ventricular tachycardia.

DrugAdministration routeInitial dosingSubsequent dosing [long-term approach]Acute success rate and time to sinus rhythmContraindications and precautions
FlecainideOral200–300 mg[long-term 50−150 mg twice daily]50%–60% at 3 h and 75%–85% at 6–8 h (3–8 h)
  • Should not be used in patients with severe structural or coronary artery disease, Brugada syndrome, or severe renal failure (CrCl <35 mL/min/1.73 m2).

  • Prior documentation of safety and efficacy in an inpatient setting is recommended prior to pill-in-the-pocket use.

  • An AVN-blocking agent should be administered to avoid 1:1 conduction if transformation to AFL.

  • Drug infusion should be discontinued in case of QRS widening >25% or bundle branch block occurrence.

  • Caution is needed in patients with sinus node disease and AVN dysfunction.

  • Do NOT use for conversion of atrial flutter.

Intravenous1–2 mg/kg over 10 min52%–95% (Up to 6 h)
PropafenoneOral450–600 mg[long-term 150-300 mg three times daily]45%–55% at 3 h, 69%–78% at 8 h (3–8 h)
Intravenous1.5–2 mg/kg over 10 min43%–89% (Up to 6 h)
AmiodaroneIntravenous (/oral)300 mg intravenous over 30–60 min900-1200 mg intravenous over 24 hours (or 200 mg oral three times daily for 4 weeks).[long-term 200 mg oral daily]44% (8–12 h to several days)
  • May cause phlebitis (use a large peripheral vein, avoid i.v. administration >24 h and use preferably volumetric pump).

  • May cause hypotension, bradycardia/atrioventricular block, QT prolongation.

  • Only if no other option in patients with hyperthyroidism (risk of thyrotoxicosis).

  • Consider the broad range of drug interactions.

IbutilideIntravenous1 mg over 10 min (0.01 mg/kg if body weight <60 kg)1 mg over 10 min
(10–20 min after the initial dose)
31%–51% (30–90 min) in AF
60–75% in AFL (60 min)
  • Should be used in the setting of a cardiac care unit as it may cause QT prolongation and torsades de pointes.

  • ECG monitoring for at least 4 h after administration to detect any proarrhythmic effects.

  • Should not be used in patients with prolonged QT, severe LVH, or low LVEF.

VernakalantIntravenous3 mg/kg over 10 min (maximum 339 mg)2 mg/kg over 10 min
(10–15 min after the initial dose) (maximum 226 mg)
50% within 10 min
  • Should not be used in patients with arterial hypotension (SBP <100 mmHg), recent ACS (within 1 month), NYHA III or IV HF, QT prolongation or severe aortic stenosis.

  • May cause arterial hypotension, QT prolongation, QRS widening, or non-sustained ventricular tachycardia.

ACS, acute coronary syndromes; AF, atrial fibrillation; AFL, atrial flutter; AVN, atrioventricular node; CrCl, creatinine clearance; ECG, electrocardiogram; HF, heart failure; LVEF, left ventricular ejection fraction; LVH, left ventricular hypertrophy; NYHA, New York Heart Association; QT, QT interval; SBP, systolic blood pressure. Long-term dosage for maintenance of sinus rhythm is indicated in [square brackets]. Long-term oral dosing for dronedarone is 400 mg twice daily, and for sotalol 80-160 mg twice daily.

© ESC 2024

A single self-administered oral dose of flecainide or propafenone (pill-in-the-pocket) is effective in symptomatic patients with infrequent and recent-onset paroxysmal AF. Safe implementation of this strategy requires patient screening to exclude sinus node dysfunction, atrioventricular conduction defects, or Brugada syndrome, as well as prior in-hospital validation of its efficacy and safety.560 An atrioventricular node-blocking drug should be instituted in patients treated with Class IC AADs to avoid 1:1 conduction if the rhythm transforms to AFL.561

Recommendation Table 17

Recommendations for pharmacological cardioversion of AF (see also Evidence Table 17)

Recommendation Table 17

Recommendations for pharmacological cardioversion of AF (see also Evidence Table 17)

7.2.4. Antiarrhythmic drugs

The aims of long-term rhythm control are to maintain sinus rhythm, improve quality of life, slow the progression of AF, and potentially reduce morbidity related to AF episodes (see Supplementary data online, Additional Evidence  Table S19).17,445,574,575 Antiarrhythmic drugs do not eliminate recurrences of AF, but in patients with paroxysmal or persistent AF, a recurrence is not equivalent to treatment failure if episodes are less frequent, briefer, or less symptomatic. Antiarrhythmic drugs also have a role for long-term rhythm control in AF patients that are considered ineligible or unwilling to undergo catheter or surgical ablation.

Before starting AAD treatment, reversible triggers should be identified and underlying comorbidities treated to reduce the arrhythmogenic substrate, prevent progression of AF, and facilitate maintenance of sinus rhythm.39,128 The RACE 3 trial, including patients with early persistent AF and mild-to-moderate heart failure (predominantly HFpEF and HFmrEF), showed that targeted therapy of underlying conditions improved sinus rhythm maintenance at 1 year (75% vs. 63% as compared with standard care).39 The selection of an AAD for long-term rhythm control requires careful evaluation that takes into account AF type, patient parameters, and safety profile.445 It also includes shared decision-making, balancing the benefit/risk ratio of AADs in comparison with other strategies. Notably, recent evidence has shown that careful institution of AADs can be performed safely.17

The long-term effectiveness of AADs is limited. In a meta-analysis of 59 RCTs, AADs reduced AF recurrences by 20%–50% compared with no treatment, placebo, or drugs for rate control.576,577 When one AAD fails to reduce AF recurrences, a clinically acceptable response may be achieved with another drug, particularly if from a different class.578 Combinations of AADs are not recommended. The data available suggest that AADs do not produce an appreciable effect on mortality or other cardiovascular complications with the exception of increased mortality signals for sotalol574,579,580 and amiodarone.581 In contrast, use of AADs within a rhythm control strategy can be associated with reduction of morbidity and mortality in selected patients.582

All AADs may produce serious cardiac (proarrhythmia, negative inotropism, hypotension) and extracardiac adverse effects (organ toxicity, predominantly amiodarone). Drug safety, rather than efficacy, should determine the choice of drug. The risk of proarrhythmia increases in patients with structural heart disease. Suggested doses for long-term oral AAD are presented in Table 13.577,583,584

Recommendation Table 18

Recommendations for antiarrhythmic drugs for long-term maintenance of sinus rhythm (see also Evidence Table 18)

Recommendation Table 18

Recommendations for antiarrhythmic drugs for long-term maintenance of sinus rhythm (see also Evidence Table 18)

7.2.5. Catheter ablation

Catheter ablation prevents AF recurrences, reduces AF burden, and improves quality of life in symptomatic paroxysmal or persistent AF where the patient is intolerant or does not respond to AAD.503–509 Multiple RCTs have provided evidence in favour of catheter ablation as a first-line approach for rhythm control in patients with paroxysmal AF, with a similar risk of adverse events as compared with initial AAD treatment (see Supplementary data online, Additional Evidence  Table S20).15,16,591–594 In contrast, it is not clear whether first-line ablation is superior to drug therapy in persistent AF. Catheter ablation may also have a role in patients with symptoms due to prolonged pauses upon AF termination, where non-randomized data have shown improved symptoms, and avoidance of pacemaker implantation.595–598

Pulmonary vein isolation (PVI) remains the cornerstone of AF catheter ablation,503,508,593,599 but the optimal ablation strategy has not been clarified in the non-paroxysmal AF population.600 New technologies are emerging, such as pulsed field ablation, in which high-amplitude electrical pulses are used to ablate the myocardium by electroporation with high tissue specificity. In a single-blind RCT of 607 patients, pulsed field ablation was non-inferior for efficacy and safety endpoints compared with conventional radiofrequency or cryoballoon ablation.601 Regarding timing of ablation, a small RCT found that delaying catheter ablation in patients with paroxysmal or persistent AF by 12 months (while on optimized medical therapy) did not impact on arrhythmia-free survival compared with ablation within 1 month.602

As with any type of rhythm control, many patients in clinical practice will not be suitable for catheter ablation due to factors that reduce the likelihood of a positive response, such as left atrial dilatation. Definitive evidence that supports the prognostic benefit of catheter ablation is needed before this invasive treatment can be considered for truly asymptomatic patients. As previously noted, the CABANA trial did not confirm a benefit of catheter ablation compared with medical therapy, although high crossover rates and low event rates may have diluted the treatment effect.3 Therefore, only highly selected asymptomatic patients could be candidates for catheter ablation, and only after detailed discussion of associated risks and potential benefit of delaying AF progression.4,603 Randomized trials have shown that AF catheter ablation in patients with HFrEF significantly reduces arrhythmia recurrence and increases ejection fraction, with improvement in clinical outcomes and mortality also observed in selected patients.4,513,514,604–612 Several characteristics, including but not limited to AF type, left atrial dilatation, and the presence of atrial and/or ventricular fibrosis, could refine patient selection to maximize outcome benefit from AF catheter ablation in patients with HFrEF.604,608,613–617 The prognostic value of catheter ablation in patients with HFpEF is less well established than for HFrEF.617–626

Recent registries and trials report varying rates of peri-procedural serious adverse events associated with catheter ablation (2.9%–7.2%) with a very low 30 day mortality rate (<0.1%). Operator experience and procedural volume at the ablation centre are critical, since they are associated with complication rates and 30 day mortality.627–631

Intermittent rhythm monitoring has typically been used to detect AF relapses following catheter ablation. Recent technology developments such as smartwatch or smartphone photoplethysmography and wearable patches may have an emerging role in post-ablation monitoring.632,633 In addition, implantable loop recorders have been used to quantify AF burden before and after ablation as an additional endpoint beyond binary AF elimination.634 Management of arrhythmia recurrence post-ablation is an informed, shared decision-making process driven by available options for symptom control. In the post-AF ablation context, there is data supporting a role for AAD continuation or re-initiation, even for previously ineffective drugs.635 A short-term AAD treatment (2–3 months) following ablation reduces early recurrences of AF,554,635–639 but does not affect late recurrences636,637,640–642 or 1 year clinical outcomes.642 Repeat PVI should be offered in patients with AF recurrence if symptom improvement was demonstrated after the first ablation, with shared decision-making and clear goals of treatment.643–645

Recommendation Table 19

Recommendations for catheter ablation of AF (see also Evidence Table 19)

Recommendation Table 19

Recommendations for catheter ablation of AF (see also Evidence Table 19)

7.2.6. Anticoagulation in patients undergoing catheter ablation

The presence of left atrial thrombus is a contraindication to catheter-based AF ablation due to the risk of thrombus dislodgement leading to ischaemic stroke. Patients planned for catheter ablation of AF with an increased risk of thromboembolism should be on OAC for at least 3 full weeks prior to the procedure.554,647

There is a wide range in practice for visualization of intra-atrial thrombi prior to catheter ablation, including TOE, intracardiac echocardiography, or delayed phase cardiac computed tomography (CT).554,648 The prevalence of left atrial thrombus was 1.3% and 2.7% in two meta-analyses of observational studies in patients planned for catheter ablation of AF on adequate OAC.649,650 The prevalence of left atrial thrombus was higher in patients with elevated stroke risk scores, and in patients with non-paroxysmal compared with paroxysmal AF.650 In addition, several patient subgroups with AF have increased risk of ischaemic stroke and intracardiac thrombus even if treated with adequate anticoagulation, including those with cardiac amyloidosis, rheumatic heart disease, and hypertrophic cardiomyopathy (HCM). Cardiac imaging before catheter ablation should be considered in these high-risk patient groups regardless of preceding effective OAC. Observational studies suggest that patients with a low thromboembolic risk profile may be managed without visualization of the LAA,651–653 but no RCTs have been performed (see Supplementary data online, Additional Evidence Table S21).

For patients who have been anticoagulated prior to the ablation procedure it is recommended to avoid interruption of OAC (see Supplementary data online, Additional Evidence Table S22).654–656 Patients with interrupted OAC showed an increase in silent stroke detected by brain magnetic resonance imaging (MRI) as compared with those with uninterrupted OAC.657–659 In a true uninterrupted DOAC strategy for once-daily dosing, a pre-procedural shift to evening intake might be considered to mitigate bleeding risk. Randomized trials show comparable safety and efficacy with minimally interrupted OAC (withholding the morning DOAC dose on the day of the procedure) and a totally uninterrupted peri-ablation OAC strategy.655

Anticoagulation with heparin during AF ablation is common practice.554 Post-ablation DOACs should be continued as per the dosing regimen when haemostasis has been achieved.335,554,647 All patients should be kept on an OAC for at least 2 months after an AF ablation procedure irrespective of estimated thromboembolic risk (see Supplementary data online, Additional Evidence Table S23).647 Meta-analyses of observational studies have tried to assess the safety of stopping OAC treatment after catheter ablation for AF, but the results have been heterogenous.660–663 Until the completion of relevant RCTs (e.g. NCT02168829), it is recommended to continue OAC therapy post-AF ablation according to the patient's CHA2DS2-VA score and not the perceived success of the ablation procedure.554

Recommendation Table 20

Recommendations for anticoagulation in patients undergoing catheter ablation (see also Evidence Table 20)

Recommendation Table 20

Recommendations for anticoagulation in patients undergoing catheter ablation (see also Evidence Table 20)

7.2.7. Endoscopic and hybrid AF ablation

Minimally invasive surgical AF ablation can be performed via a thoracoscopic approach or a subxiphoid approach. The term endoscopic covers both strategies. Hybrid ablation approaches have been developed where endoscopic epicardial ablation on the beating heart is performed in combination with endocardial catheter ablation, either in a simultaneous or sequential procedure. The rationale for combining an endocardial with an epicardial approach is that a more effective transmural ablation strategy can be pursued.666,667

For paroxysmal AF, an endoscopic or hybrid ablation approach may be considered after a failed percutaneous catheter ablation strategy.668–670 Long-term follow-up of the FAST RCT (mean of 7.0 years), which included patients with paroxysmal and persistent AF, found arrhythmia recurrence was common but substantially lower with thoracoscopic ablation than catheter ablation: 34/61 patients (56%) compared with 55/63 patients (87%), with P < .001 for the comparison.669 For persistent AF, endoscopic or hybrid ablation approaches are suitable as a first procedure to maintain long-term sinus rhythm in selected patients.667–672 A meta-analysis of three RCTs confirmed a lower rate of atrial arrhythmia recurrence after thoracoscopic vs. catheter ablation (incidence rate ratio, 0.55; 95% CI, 0.38–0.78; with no heterogeneity between trials).669 An RCT with 12 month follow-up published after the meta-analysis in patients with long-standing persistent AF found no difference in arrhythmia freedom comparing thoracoscopic with catheter ablation.673 Although overall morbidity and mortality of both techniques is low, endoscopic and hybrid ablation have higher complication rates than catheter ablation, but similar long-term rates of the composite of mortality, MI, or stroke.667,669

More recent trials have assessed the efficacy and safety of the hybrid epicardial-plus-endocardial approach in persistent AF refractory to AAD therapy, including a single-centre RCT670 and two multicentre RCTs.671,674 Across these trials, hybrid ablation was consistently superior to catheter ablation alone for maintaining long-term sinus rhythm, without significant differences in major adverse events. Notably, these studies were typically performed in highly experienced centres (see Supplementary data online, Additional Evidence Table S24).

Similar to other rhythm control approaches, this task force recommends that OAC are continued in all patients who have a risk of thromboembolism, irrespective of rhythm outcome, and regardless of LAA exclusion performed as part of the surgical procedure.

Recommendation Table 21

Recommendations for endoscopic and hybrid AF ablation (see also Evidence Table 21)

Recommendation Table 21

Recommendations for endoscopic and hybrid AF ablation (see also Evidence Table 21)

7.2.8. AF ablation during cardiac surgery

Atrial fibrillation is a significant risk factor for early mortality, late mortality, and stroke in patients referred for cardiac surgery.675–677 The best validated method of surgical ablation is the Maze procedure, consisting of a pattern of transmural lesions including PVI, with subsequent modifications using bipolar radiofrequency and/or cryothermy ablation with LAA amputation (see Supplementary data online, Additional Evidence  Table S25).678–681 Education and training, close co-operation within a multidisciplinary team, and shared decision-making can improve the quality and outcomes of surgical ablation.682

A number of RCTs have shown that surgical AF ablation during cardiac surgery increases freedom from arrhythmia recurrence.683–688 Performing surgical AF ablation, mainly targeting those patients needing mitral valve surgery, is not associated with increased morbidity or mortality.678,683–685 Observational data, including large registries, have supported the potential value of surgical AF ablation,689–700 but further RCTs are needed to evaluate which patients should be selected, and whether this approach contributes to the prevention of stroke, thromboembolism, and death.

Data on pacemaker implantation rates after surgical AF ablation are variable and are likely influenced by centre experience and the patients treated (e.g. underlying sinus node disease). In a systematic review of 22 RCTs (1726 patients), permanent pacemaker implantation rates were higher with surgical AF ablation than without concomitant AF surgery (6.0% vs. 4.1%; RR, 1.69; 95% CI, 1.12–2.54).701 Observational registry data from contemporary cohorts (2011–2020) suggest an overall pacemaker rate post-operatively of 2.1% in patients selected for surgical AF ablation, with no discernible impact of surgical ablation on the need for a pacemaker, but higher rates in those needing multivalve surgery.702 With a safety-first approach in mind, imaging is advised during surgical AF ablation to exclude thrombus and help to plan the surgical approach (e.g. with TOE), regardless of effective pre-procedural anticoagulant use.

Recommendation Table 22

Recommendations for AF ablation during cardiac surgery (see also Evidence Table 22)

Recommendation Table 22

Recommendations for AF ablation during cardiac surgery (see also Evidence Table 22)

7.2.9. Atrial tachycardia after pulmonary vein isolation

After any ablation for AF, recurrent arrhythmias may manifest as AF, but also as atrial tachycardia (AT). Although AT may be perceived as a step in the right direction by the treating physician, this view is often not shared by the patient because AT can be equally or more symptomatic than the original AF. Conventionally, an early arrhythmia recurrence post-PVI (whether AT, AF, or flutter) is considered potentially transitory.708 Recent trials using continuous implantable loop recorders for peri-procedural monitoring have provided insight into the incidence and significance of early arrhythmia recurrences, and have confirmed a link between early and later recurrence.709 Discussion of management options for AT post-ablation should ideally involve a multidisciplinary team with experience in interventional management of complex arrhythmias, considering technical challenges, procedural efficacy, and safety, in the context of patient preferences.

8. [E] Evaluation and dynamic reassessment

The development and progression of AF results from continuous interactions between underlying mechanisms (electrical, cellular, neurohormonal, and haemodynamic), coupled with a broad range of clinical factors and associated comorbidities. Each individual factor has considerable variability over time, affecting its contribution to the AF-promoting substrate. The risk profile of each patient is also far from static, and requires a dynamic mode of care to ensure optimal AF management.710,711 Patients with AF require periodic reassessment of therapy based on this changing risk status if we are to improve the overall quality of care. Timely attention to modifiable factors and underpinning comorbidities has the potential to slow or reverse the progression of AF, increase quality of life, and prevent adverse outcomes such as heart failure, thromboembolism, and major bleeding.

The [E] in AF-CARE encompasses the range of activity needed by healthcare professionals and patients to: (i) thoroughly evaluate associated comorbidities and risk factors that can guide treatment; and (ii) provide the dynamic assessment needed to ensure that treatment plans remain suited to that particular patient. This task force recommends an adaptive strategy that not only reacts to changes notified by a patient, but also proactively seeks out issues where altering management could impact on patient wellbeing. Avoidance of unnecessary and costly follow-up is also inherent in this framework, with educated and empowered patients contributing to identifying the need for access to specialist care or an escalation of management. The patient-centred, shared decision philosophy is embedded to improve efficiency in models of care and to address the needs of patients with AF.

Medical history and the results of any tests should be regularly re-evaluated to address the dynamic nature of comorbidities and risk factors.712 This may have impact on therapeutic decisions; e.g. resumption of full-dose DOAC therapy after improvement in the patient's renal function. The timing of review of the AF-CARE pathway is patient specific and should respond to changes in clinical status. In most cases, this task force advises re-evaluation 6 months after initial presentation, and then at least annually by a healthcare professional in primary or secondary care (see Figure 3).

8.1. Implementation of dynamic care

A multidisciplinary-based approach is advocated to improve implementation of dynamic AF-CARE (see Figure 2); although potentially resource intensive, this is preferred to more simplistic or opportunistic methods. For example, in a pragmatic trial of 47 333 AF patients identified through health insurance claims, there was no difference in OAC initiation at 1 year in those randomized to a single mailout of patient and clinician education, compared with those in the usual care group.713 For co-ordination of care there is a core role for cardiologists, general practitioners, specialized nurses, and pharmacists.714 If needed, and depending on local resources, others may also be involved (cardiac surgeons, physiotherapists, neurologists, psychologists, and other allied health professionals). It is strongly advocated that one core team member co-ordinates care, and that additional team members become involved according to the needs of the individual patient throughout their AF trajectory.

Several organizational models of integrated care for AF have been evaluated, but which components are most useful remains unclear. Some models include a multidisciplinary team,715,716 while others are nurse-led79,122,124,717 or cardiologist-led.79,122,124,717 Several published models used computerized decision support systems or electronic health applications.79,122,715,718 Evaluation within RCTs has demonstrated mixed results due to the variety of methods tested and differences in regional care. Several studies report significant improvements with respect to adherence to anticoagulation, cardiovascular mortality, and hospitalization relative to standard of care.121–123 However, the RACE 4 (IntegRAted Chronic Care Program at Specialized AF Clinic Versus Usual CarE in Patients with Atrial Fibrillation) trial, which included 1375 patients, failed to demonstrate superiority of nurse-led over usual care.79 New studies of the components and optimal models for delivery for integrated care approaches in routine practice are ongoing (ACTRN12616001109493, NCT03924739).

8.2. Improving treatment adherence

Advances in the care of patients with AF can only be effective if appropriate tools are available to support the implementation of the treatment regimen.719 A number of factors are related to the optimal implementation of care at the level of: (i) the individual patient (culture, cognitive impairment, and psychological status); (ii) the treatment (complexity, side effects, polypharmacy, impact on daily life, and cost); (iii) the healthcare system (access to treatment and multidisciplinary approach); and (iv) the healthcare professional (knowledge, awareness of guidelines, expertise, and communication skills). A collaborative approach to patient care, based upon shared decision-making and goals tailored to individual patient needs, is crucial in promoting ongoing patient adherence to the agreed treatment regimen.720 Even when treatment seems feasible for the individual, patients often lack access to reliable and up-to-date information about risks and benefits of various treatment options, and consequently are not empowered to engage in their own management. A sense of ownership that promotes the achievement of joint goals can be encouraged through the use of educational programmes, websites (such as https://afibmatters.org), app-based tools, and individually tailored treatment protocols which take into account gender, ethnic, socioeconomic, environmental, and work factors. In addition, practical tools (e.g. schedules, apps, brochures, reminders, pillboxes) can help to implement treatment in daily life.721,722 Regular review by members of the multidisciplinary team enables the evolution of a flexible and responsive management regimen that the patient will find easier to follow.

8.3. Cardiac imaging

A TTE is a valuable asset across all four AF-CARE domains when there are changes in the clinical status of an individual patient (Figure 13).723–725 The key findings to consider from an echocardiogram are any structural heart disease (e.g. valvular disease or left ventricular hypertrophy), impairment of left ventricular function (systolic and/or diastolic to classify heart failure subtype), left atrial dilatation, and right heart dysfunction.59,67,726 To counter irregularity when in AF, obtaining measurements in cardiac cycles that follow two similar RR intervals can improve the value of parameters compared with sequential averaging of cardiac cycles.723,727 Contrast TTE or alternative imaging modalities may be required where image quality is poor, and quantification of left ventricular systolic function is needed for decisions on rate or rhythm control. Other cardiac imaging techniques, such as cardiac magnetic resonance (CMR), CT, TOE, and nuclear imaging can be valuable when: (i) TTE quality is suboptimal for diagnostic purposes; (ii) additional information is needed on structure, substrate, or function; and (iii) to support decisions on interventional procedures (see Supplementary data online, Figure S1).59,724,725,728 As with TTE, other types of cardiac imaging can be challenging in the context of AF irregularity or with rapid heart rate, requiring technique-specific modifications when acquiring ECG-gated sequences.729–731

Relevance of echocardiography in the AF-CARE pathway.
Figure 13

Relevance of echocardiography in the AF-CARE pathway.

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; CHA2DS2-VA, congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/transient ischaemic attack/arterial thromboembolism (2 points), vascular disease, age 65–74 years; LAA, left atrial appendage; LV, left ventricle.

8.4. Patient-reported outcome measures

Patients with AF have a lower quality of life compared with the general population.732 Improvement in quality of life and functional status should play a key role in assessing and reassessing treatment decisions (see Supplementary data online, Additional Evidence  Table S26).36 Patient-reported outcome measures are valuable to measure quality of life, functional status, symptoms, and treatment burden for patients with AF over time.55,733–735 Patient-reported outcome measures are playing an increasing role in clinical trials to assess the success of treatment; however, they remain under-utilized.736,737 They can be divided into generic or disease-specific tools, with the latter helping to provide insight into AF-related impacts.738 However, multimorbidity can still confound the sensitivity of all PROMs, impacting on association with other established metrics of treatment performance such as mEHRA symptom class and natriuretic peptides.48 Intervention studies have demonstrated an association between improvement in PROM scores and reduction in AF burden and symptoms.48,738

Atrial fibrillation-specific questionnaires include the AF 6 (AF6),739 Atrial Fibrillation Effect on QualiTy-of-Life (AFEQT),740 the Atrial Fibrillation Quality of Life Questionnaire (AFQLQ),741 the Atrial Fibrillation Quality of Life (AF-QoL),742 and the Quality of Life in Atrial Fibrillation (QLAF).743 The measurement properties of most of these tools lack sufficient validation.49 The International Consortium for Health Outcomes Measurement (ICHOM) working group recommends the use of the AFEQT PROM or a symptom questionnaire called the Atrial Fibrillation Severity Scale (AFSS) for measuring exercise tolerance and the impact of symptoms in AF.744 Through wider use of patient experience measures, there is an opportunity at the institutional level to improve the quality of care delivered to patients with AF.49–55

Recommendation Table 23

Recommendations to improve patient experience (see also Evidence Table 23)

Recommendation Table 23

Recommendations to improve patient experience (see also Evidence Table 23)

9. The AF-CARE pathway in specific clinical settings

The following sections detail specific clinical settings where approaches to AF-CARE may vary. Unless specially discussed, measures for [C] comorbidity and risk factor management, [A] avoidance of stroke and thromboembolism, [R] rate and rhythm control, and [E] evaluation and dynamic reassessment should follow the standard pathways introduced in Section 4.

9.1. AF-CARE in unstable patients

Unstable patients with AF include those with haemodynamic instability caused by the arrhythmia or acute cardiac conditions, and severely ill patients who develop AF (sepsis, trauma, surgery, and particularly cancer-related surgery). Conditions such as sepsis, adrenergic overstimulation, and electrolyte disturbances contribute to onset and recurrence of AF in these patients. Spontaneous restoration of sinus rhythm has been reported in up to 83% during the first 48 h after appropriate treatment of the underlying cause.551,745

Emergency electrical cardioversion is still considered the first-choice treatment if sinus rhythm is thought to be beneficial, despite the limitation of having a high rate of immediate relapse.746 Amiodarone is a second-line option because of its delayed activity; however, it may be an appropriate alternative in the acute setting.747,748 In a multicentre cohort study carried out in the United Kingdom and the United States of America, amiodarone and beta-blockers were similarly effective for rate control in intensive care patients, and superior to digoxin and calcium channel blockers.749 The ultra-short acting and highly selective beta-blocker landiolol can safely control rapid AF in patients with low ejection fraction and acutely decompensated heart failure, with a limited impact on myocardial contractility or blood pressure.477,750,751

9.2. AF-CARE in acute and chronic coronary syndromes

The incidence of AF in acute coronary syndromes (ACS) ranges from 2% to 23%.752 The risk of new-onset AF is increased by 60%–77% in patients suffering an MI,753 and AF may be associated with an increased risk of ST-segment elevation myocardial infarction (STEMI) or non-STEMI ACS.754 Overall, 10%–15% of AF patients undergo percutaneous intervention (PCI) for CAD.755 In addition, AF is a common precipitant for type 2 MI.756 Observational studies show that patients with both ACS and AF are less likely to receive appropriate antithrombotic therapy757 and more likely to experience adverse outcomes.758 Peri-procedural management of patients with ACS or chronic coronary syndromes (CCS) are detailed in the 2023 ESC Guidelines for the management of acute coronary syndromes and 2024 ESC Guidelines for the management of chronic coronary syndromes.759,760

The combination of AF with ACS is the area where use of multiple antithrombotic drugs is most frequently indicated, consisting of antiplatelet agents plus OAC (Figure 14) (see Supplementary data online, Additional Evidence Table S27). There is a general trend to decrease the duration of DAPT to reduce bleeding; however, this may increase ischaemic events and stent thrombosis.761,762 In ACS there is a high risk of predominantly platelet-driven atherothrombosis and thus of coronary ischaemic events. Acute coronary syndromes treated by PCI require DAPT for improved short- and long-term prognosis. Therefore, a peri-procedural triple antithrombotic regimen including an OAC, aspirin, and a P2Y12 inhibitor should be the default strategy for most patients. Major thrombotic events vs. major bleeding risk need to be balanced when prescribing antiplatelet therapy and OAC after the acute phase and/or after PCI. The combination of OAC (preferably a DOAC) and a P2Y12 inhibitor results in less major bleeding than triple therapy that includes aspirin. Clopidogrel is the preferred P2Y12 inhibitor, as the evidence for ticagrelor and prasugrel is less clear with higher bleeding risk.763–769 Ongoing trials will add to our knowledge about safely combining DOACs with antiplatelet agents (NCT04981041, NCT04436978). When using VKAs with antiplatelet agents, there is consensus opinion to use an INR range of 2.0–2.5 to mitigate excess bleeding risk.

Antithrombotic therapy in patients with AF and acute or chronic coronary syndromes.
Figure 14

Antithrombotic therapy in patients with AF and acute or chronic coronary syndromes.

ACS, acute coronary syndromes; CCS, chronic coronary syndrome; DOAC, direct oral anticoagulant; INR, international normalized ratio of prothrombin time; OAC, oral anticoagulant; P2Y12i, P2Y12-receptor inhibitor antiplatelet agents (clopidogrel, prasugrel, ticagrelor); PCI, percutaneous intervention; TTR, time in therapeutic range; VKA, vitamin K antagonist. The flowchart applies to those patients with an indication for oral anticoagulant therapy. aThe full standard dose of DOACs should be used unless the patient fulfils dose-reduction criteria (Table 11). When rivaroxaban or dabigatran are used as the DOAC and concerns about bleeding risk prevail over stent thrombosis or ischaemic stroke, the reduced dose should be considered (15 mg and 110 mg respectively; Class IIa). bIn patients with diabetes mellitus undergoing coronary stent implantation, prolonging triple antithrombotic therapy for up to 3 months may be of value if thrombotic risk outweighs the bleeding risk.

Short–term triple therapy (≤1 week) is recommended for all patients without diabetes after ACS or PCI. In pooled analyses of RCTs, omitting aspirin in patients with ACS undergoing PCI has the potential for higher rates of ischaemic/stent thrombosis, without impact on incident stroke.761,762,770–772 None of the trials were powered for ischaemic events. All patients in AUGUSTUS (an open–label, 2 × 2 factorial, randomized controlled clinical trial to evaluate the safety of apixaban vs. vitamin k antagonist and aspirin vs. aspirin placebo in patients with AF and ACS or PCI) received aspirin plus a P2Y12 inhibitor for a median time of 6 days.773 At the end of the trial, apixaban and a P2Y12 inhibitor without aspirin was the optimal treatment regimen for most patients with AF and ACS and/or PCI, irrespective of the patient's baseline bleeding and stroke risk.774,775

Prolonged triple therapy up to 1 month after ACS/PCI should be considered in patients at high ischaemic risk, e.g. STEMI, prior stent thrombosis, complex coronary procedures, and prolonged cardiac instability, even though these patients were not adequately represented in the RCTs so far available.776 In AF patients with ACS or CCS and diabetes mellitus undergoing coronary stent implantation, prolonging triple therapy with low-dose aspirin, clopidogrel, and an OAC up to 3 months may be of benefit if thrombotic risk outweighs bleeding risk in the individual patient.207

The evidence for ACS treated without revascularization is limited. Six to 12 months of a single antiplatelet agent in addition to a long-term DOAC is usually sufficient and can minimize bleeding risk.760,764,774 Although there are no head-to-head comparisons between aspirin and clopidogrel, studies have typically used clopidogrel. In patients with stable CCS for more than 12 months, sole therapy with a DOAC is sufficient and no additional antiplatelet therapy is required.353 In patients at potential risk of gastrointestinal bleeding, use of proton pump inhibitors is reasonable during combined antithrombotic therapy, although evidence in AF patients is limited.437,777–779 Multimorbid patients with ACS or CCS need careful assessment of ischaemic risk and management of modifiable bleeding risk factors, with a comprehensive work-up to individually adapt antithrombotic therapy.

Recommendation Table 24

Recommendations for patients with acute coronary syndromes or undergoing percutaneous intervention (see also Evidence Table 24)

Recommendation Table 24

Recommendations for patients with acute coronary syndromes or undergoing percutaneous intervention (see also Evidence Table 24)

9.3. AF-CARE in vascular disease

Peripheral arterial disease (PAD) is common in patients with AF, ranging from 6.7% to 14% of patients.783,784 Manifest PAD is associated with incident AF.785 PAD predicts a higher mortality in patients with AF and is an independent predictor of stroke in those not on OAC.783,786 Patients with lower extremity artery disease and AF also have a higher overall mortality and risk of major cardiac events.784,787,788 A public health database of >40 000 patients hospitalized for PAD or critical limb ischaemia showed AF to be an independent predictor for mortality (HR, 1.46; 95% CI, 1.39–1.52) and ischaemic stroke (HR, 1.63; 95% CI, 1.44–1.85) as compared with propensity-matched controls.784 Similarly, in patients undergoing carotid endarterectomy or stenting, the presence of AF is associated with higher mortality (OR, 1.59; 95% CI, 1.11–2.26).789

Anticoagulation alone is usually sufficient in the chronic disease phase, with DOACs being the preferred agents despite one RCT subanalysis showing a higher risk of bleeding as compared with warfarin.790 In the case of recent endovascular revascularization, a period of combination with single antiplatelet therapy should be considered, weighing bleeding and thrombotic risks and keeping the period of combination antithrombotic therapy as brief as possible (ranging between 1 month for peripheral791 and 90 days for neuro-interventional procedures).792

9.4. AF-CARE in acute stroke or intracranial haemorrhage

9.4.1. Management of acute ischaemic stroke

Management of acute stroke in patients with AF is beyond the scope of these guidelines. In AF patients presenting with acute ischaemic stroke while taking OAC, acute therapy depends on the treatment regimen and intensity of OAC. Management should be co-ordinated by a specialist neurologist team according to relevant guidelines.793

9.4.2. Introduction or re-introduction of anticoagulation after ischaemic stroke

The optimal time for administering OAC in patients with acute cardioembolic stroke and AF remains unclear. Randomized control trials have been unable to provide any evidence to support the administration of anticoagulants or heparin in patients with acute ischaemic stroke within 48 h from stroke onset. This suggests that low-dose aspirin should be administered to all patients during this timeframe.794

Two trials have examined the use of DOAC therapy early after stroke, with no difference in clinical outcomes compared with delayed DOAC prescription. The ELAN (Early versus Late initiation of direct oral Anticoagulants in post-ischaemic stroke patients with atrial fibrillatioN) trial randomized 2013 patients with acute ischaemic stroke and AF to open-label early use of DOACs (<48 h after minor/moderate stroke; day 6–7 after major stroke) vs. later DOAC prescription (day 3–4 after minor stroke; day 6–7 after moderate stroke; day 12–14 after major stroke). There was no significant difference in the composite thromboembolic, bleeding, and vascular death outcome at 30 days (risk difference early vs. late, −1.18%; 95% CI, −2.84 to 0.47).795 The TIMING (Timing of Oral Anticoagulant Therapy in Acute Ischemic Stroke With Atrial Fibrillation) trial, a registry-based, non-inferiority, open-label, blinded endpoint trial randomized 888 patients within 72 h of ischaemic stroke onset to early (≤4 days) or delayed (5–10 days) DOAC initiation. Early DOAC use was non-inferior to the delayed strategy for the composite of thromboembolism, bleeding and all-cause mortality at 90 days (risk difference, −1.79%; 95% CI, −5.31% to 1.74%).796 Two ongoing trials will provide further guidance on the most appropriate timing of DOAC therapy after ischaemic stroke (NCT03759938, NCT03021928).

9.4.3. Introduction or re-introduction of anticoagulation after haemorrhagic stroke

There is insufficient evidence currently to recommend whether OAC should be started or re-started after ICH to protect against the high risk of ischaemic stroke in these patients (see Supplementary data online, Additional Evidence Table S28). Data from two pilot trials are available. The APACHE-AF (Apixaban After Anticoagulation-associated Intracerebral Haemorrhage in Patients With Atrial Fibrillation) trial was a prospective, randomized, open-label trial with masked endpoint assessment; 101 patients who survived 7–90 days after anticoagulation-associated ICH were randomized to apixaban or no OAC. During a median of 1.9 years follow-up (222 person-years), there was no difference in non-fatal stroke or vascular death, with an annual event rate of 12.6% with apixaban and 11.9% with no OAC (adjusted HR, 1.05; 95% CI, 0.48–2.31; P = .90).797 SoSTART (Start or STop Anticoagulants Randomised Trial) was an open-label RCT in 203 patients with AF after symptomatic spontaneous ICH. Starting OAC was not non-inferior to avoiding long-term (≥1 year) OAC, with ICH recurrence in 8/101 (8%) vs. 4/102 (4%) patients (adjusted HR, 2.42; 95% CI, 0.72–8.09). Mortality occurred in 22/101 (22%) patients in the OAC group vs. 11/102 (11%) patients where OAC were avoided.798

Until additional trials report on the clinical challenge of post-ICH anticoagulation (NCT03950076, NCT03996772), an individualized multidisciplinary approach is advised led by an expert neurology team.

9.5. AF-CARE for trigger-induced AF

Trigger-induced AF is defined as new AF in the immediate association of a precipitating and potentially reversible factor. Also known as ‘secondary’ AF, this task force prefer the term trigger-induced as there are almost always underlying factors in individual patients that can benefit from full consideration of the AF-CARE pathway. The most common precipitant unmasking a tendency to AF is acute sepsis, where AF prevalence is between 9% and 20% and has been associated with a worse prognosis.11–14 The degree of inflammation correlates with the incidence of AF,799 which may partly explain the wide variability across studies in prevalence, as well as recurrence of AF. Longer-term data suggest that AF triggered by sepsis recurs after discharge in between a third to a half of patients.12,800–807 In addition to other acute triggers which may be causal (such as alcohol808,809 and illicit drug use810), numerous conditions are also associated with chronic inflammation leading to subacute stimuli for AF (Table 14). The specific trigger of an operative procedure is discussed in Section 9.6.

Table 14

Non-cardiac conditions associated with trigger-induced AF

Acute conditions
Infections (bacterial and viral)
Pericarditis, myocarditis
Emergency conditions (burn injury, severe trauma, shock)
Binge alcohol consumption
Drug use, including methamphetamines, cocaine, opiates, and cannabis
Acute interventions, procedures, and surgery
Endocrine disorders (thyroid, adrenal, pituitary, others)
Acute conditions
Infections (bacterial and viral)
Pericarditis, myocarditis
Emergency conditions (burn injury, severe trauma, shock)
Binge alcohol consumption
Drug use, including methamphetamines, cocaine, opiates, and cannabis
Acute interventions, procedures, and surgery
Endocrine disorders (thyroid, adrenal, pituitary, others)
Chronic conditions with inflammation and enhanced AF substrate
Immune-mediated diseases (rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, coeliac disease, psoriasis, others)
Obesity
Chronic obstructive airways disease
Obstructive sleep apnoea
Cancer
Fatty liver disease
Stress
Endocrine disorders (see Section 9.10)
Chronic conditions with inflammation and enhanced AF substrate
Immune-mediated diseases (rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, coeliac disease, psoriasis, others)
Obesity
Chronic obstructive airways disease
Obstructive sleep apnoea
Cancer
Fatty liver disease
Stress
Endocrine disorders (see Section 9.10)
Table 14

Non-cardiac conditions associated with trigger-induced AF

Acute conditions
Infections (bacterial and viral)
Pericarditis, myocarditis
Emergency conditions (burn injury, severe trauma, shock)
Binge alcohol consumption
Drug use, including methamphetamines, cocaine, opiates, and cannabis
Acute interventions, procedures, and surgery
Endocrine disorders (thyroid, adrenal, pituitary, others)
Acute conditions
Infections (bacterial and viral)
Pericarditis, myocarditis
Emergency conditions (burn injury, severe trauma, shock)
Binge alcohol consumption
Drug use, including methamphetamines, cocaine, opiates, and cannabis
Acute interventions, procedures, and surgery
Endocrine disorders (thyroid, adrenal, pituitary, others)
Chronic conditions with inflammation and enhanced AF substrate
Immune-mediated diseases (rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, coeliac disease, psoriasis, others)
Obesity
Chronic obstructive airways disease
Obstructive sleep apnoea
Cancer
Fatty liver disease
Stress
Endocrine disorders (see Section 9.10)
Chronic conditions with inflammation and enhanced AF substrate
Immune-mediated diseases (rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, coeliac disease, psoriasis, others)
Obesity
Chronic obstructive airways disease
Obstructive sleep apnoea
Cancer
Fatty liver disease
Stress
Endocrine disorders (see Section 9.10)

After meeting the diagnostic criteria for AF (see Section 3.2), the management of trigger-induced AF is recommended to follow the AF-CARE principles, with critical consideration of underlying risk factors and comorbidities. Based on retrospective and observational data, patients with AF and trigger-induced AF seem to carry the same thromboembolic risk as patients with primary AF.811,812 In the acute phase of sepsis, patients show an unclear risk–benefit profile with anticoagulation therapy.813,814 Prospective studies on anticoagulation in patients with triggered AF episodes are lacking.802,812,815 Acknowledging that there are no RCTs specifically available in this population to assess trigger-induced AF, long-term OAC therapy should be considered in suitable patients with trigger-induced AF who are at elevated risk of thromboembolism, starting OAC after the acute trigger has been corrected and considering the anticipated net clinical benefit and informed patient preferences. As with any decision on OAC, not all patients will be suitable for OAC, depending on relative and absolute contraindications and the risk of major bleeding. The approach to rate and rhythm control will depend on subsequent recurrence of AF or any associated symptoms, and re-evaluation should be individualized to take account of the often high AF recurrence rate.

Recommendation Table 25

Recommendations for trigger-induced AF (see also Evidence Table 25)

Recommendation Table 25

Recommendations for trigger-induced AF (see also Evidence Table 25)

9.6. AF-CARE in post-operative patients

Peri-operative AF describes the onset of the arrhythmia during an ongoing intervention. Post-operative AF (POAF), defined as new-onset AF in the immediate post-operative period, is a common complication with clinical impact that occurs in 30%–50% of patients undergoing cardiac surgery,816–818 and in 5%–30% of patients undergoing non-cardiac surgery. Intra- and post-operative changes and specific AF triggers (including peri-operative complications) and pre-existing AF-related risk factors and comorbidities increase the susceptibility to POAF.819 Although POAF episodes may be self-terminating, POAF is associated with 4–5 times increase in recurrent AF during the next 5 years,820,821 and is a risk factor for stroke, MI, heart failure, and death.822–827 Other adverse events associated with POAF include haemodynamic instability, prolonged hospital stay, infections, renal complications, bleeding, increased in-hospital death, and greater healthcare cost.828–830

While multiple strategies to prevent POAF with pre-treatment or acute drug treatment have been described, there is a lack of evidence from large RCTs. Pre-operative use of propranolol or carvedilol plus N-acetyl cysteine in cardiac and non-cardiac surgery is associated with a reduced incidence of POAF,831–834 but not major adverse events.835 An umbrella review of 89 RCTs from 23 meta-analyses (19 211 patients, but not necessarily in AF) showed no benefit from beta-blockers in cardiac surgery for mortality, MI, or stroke. In non-cardiac surgery, beta-blockers were associated with reduced rates of MI after surgery (RR range, 0.08–0.92), but higher mortality (RR range, 1.03–1.31), and increased risk of stroke (RR range, 1.33–7.72).836 Prevention of peri-operative AF can also be achieved with amiodarone. In a meta-analyses, amiodarone (oral or intravenous [i.v.]) and beta-blockers were equally effective in reducing post-operative AF,837 but their combination was better than beta-blockers alone.838 Lower cumulative doses of amiodarone (<3000 mg during the loading phase) could be effective, with fewer adverse events.837,839,840 Withdrawal of beta-blockers should be avoided due to increased risk of POAF.841 Other treatment strategies (steroids, magnesium, sotalol, (bi)atrial pacing, and botulium injection into the epicardial fat pad) lack scientific evidence for the prevention of peri-operative AF.842,843 Peri-operative posterior pericardiotomy, due to the reduction of post-operative pericardial effusion, showed a significant decrease in POAF in patients undergoing cardiac surgery (OR, 0.44; 95% CI, 0.27–0.70; P = .0005).844–846 In 3209 patients undergoing non-cardiac thoracic surgery, colchicine did not lead to any significant reduction in AF compared with placebo (HR, 0.85; 95% CI, 0.65–1.10; P = .22).847

The evidence for prevention of ischaemic stroke in POAF by OAC is limited.822,827 Oral anticoagulant therapy is associated with a high bleeding risk soon after cardiac surgery or major non-cardiac interventions.827 Conversely, meta-analyses of observational cohort studies suggest a possible protective impact of OAC in POAF for all-cause mortality848 and a lower risk of thromboembolic events following cardiac surgery, accompanied by higher rates of bleeding.849 This task force recommends to treat post-operative AF according to the AF-CARE pathway as discussed for trigger-induced AF (with the [R] pathway the same as for first-diagnosed AF). Ongoing RCTs in cardiac surgery (NCT04045665) and non-cardiac surgery (NCT03968393) will inform optimal long-term OAC use among patients with POAF. While awaiting the results of these trials, this task force recommends that after acute bleeding risk has settled, long-term OAC should be considered in patients with POAF according to their thromboembolic risk factors.

Recommendation Table 26

Recommendations for management of post-operative AF (see also Evidence Table 26)

Recommendation Table 26

Recommendations for management of post-operative AF (see also Evidence Table 26)

9.7. AF-CARE in embolic stroke of unknown source

The term ‘embolic stroke of undetermined source’ (ESUS) was introduced to identify non-lacunar strokes whose mechanism is likely to be embolic, but the source remains unidentified.856 Of note, these patients have a recurrent risk of stroke of 4%–5% per year.856 The main embolic sources associated with ESUS are concealed AF, atrial cardiomyopathy, left ventricular disease, atherosclerotic plaques, patent foramen ovale (PFO), valvular diseases, and cancer. Atrial cardiomyopathy and left ventricular disease are the most prevalent causes.856 AF is reported to be the underlying mechanism in 30% of ESUS patients.857–859 The detection of AF among ESUS patients increases the longer cardiac monitoring is provided (see Supplementary data online, Additional Evidence Table S29).857,860–864 This also holds for the duration of implantable cardiac monitoring, with probability of AF detection ranging from 2% with 1 week to over 20% by 3 years.865 In patients with ESUS, factors associated with an increased detection of AF are increasing age,866,867 left atrial enlargement,866 cortical location of stroke,868 large or small vessel disease,863 an increased number of atrial premature beats per 24 h,868 rhythm irregularity,859 and risk stratification scores (such as CHA2DS2-VASc,869 Brown ESUS-AF,870 HAVOC,871 and C2HEST872). This task force recommends prolonged monitoring depending on the presence of the above-mentioned risk markers.865,873,874

Currently available evidence, including two completed RCTs and one stopped for futility, do not support the use of DOACs compared with aspirin in patients with acute ESUS without documented AF.875–877 Ongoing trials will provide further guidance (NCT05134454, NCT05293080, NCT04371055).

Recommendation Table 27

Recommendations for patients with embolic stroke of unknown source (see also Evidence Table 27)

Recommendation Table 27

Recommendations for patients with embolic stroke of unknown source (see also Evidence Table 27)

9.8. AF-CARE during pregnancy

Atrial fibrillation is one of the most common arrhythmias during pregnancy, with prevalence increasing due to higher maternal age and changes in lifestyle, and because more women with congenital heart disease survive to childbearing age.878–881 Rapid atrioventricular conduction of AF may have serious haemodynamic consequences for mother and foetus. AF during pregnancy is associated with an increased risk of death.882 A multidisciplinary approach is essential to prevent maternal and foetal complications, bringing together gynaecologists, neonatologists, anaesthesiologists, and cardiologists experienced in maternal medicine.883

Pregnancy is associated with a hypercoagulable state and increased thromboembolic risk.884 The same rules for risk assessment of thromboembolism should be used as in non-pregnant women, as detailed in the 2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy.885 The preferred agents for anticoagulation of AF during pregnancy are unfractionated or low molecular weight heparins (LMWHs), which do not cross the placenta. Vitamin K antagonists should be avoided in the first trimester (risk of miscarriage, teratogenicity) and from week 36 onwards (risk of foetal intracranial bleeding if early unexpected delivery). Direct oral anticoagulants are not recommended during pregnancy due to concerns about safety.886 However, an accidental exposure during pregnancy should not lead to a recommendation for termination of the pregnancy.887 Vaginal delivery should be advised for most women, but is contraindicated during VKA treatment because of the risk of foetal intracranial bleeding.885

Intravenous selective beta-1 receptor blockers are recommended as first choice for acute heart rate control of AF.888 This does not include atenolol, which can lead to intrauterine growth retardation.889 If beta-blockers fail, digoxin and verapamil can be considered for rate control (verapamil should be avoided in the first trimester). Rhythm control is the preferred strategy during pregnancy. Electrical cardioversion is recommended if there is haemodynamic instability, considerable risk to mother or foetus, or with concomitant HCM. Electrical cardioversion can be performed safely without compromising foetal blood flow, and the consequent risk for foetal arrhythmias or pre-term labour is low. The foetal heart rate should be closely monitored throughout and after cardioversion, which should generally be preceded by anticoagulation.885 In haemodynamically stable women without structural heart disease, intravenous ibutilide or flecainide may be considered for termination of AF, but experience is limited.890 Catheter ablation is normally avoided during pregnancy,883 but is technically feasible without radiation in refractory symptomatic cases with a minimal/zero fluoroscopy approach.883

Counselling is important in women of childbearing potential prior to pregnancy, highlighting the potential risks of anticoagulation and rate or rhythm control drugs (including teratogenic risk, where relevant). Contraception and timely switch to safe drugs should be proactively discussed.

Recommendation Table 28

Recommendations for patients with AF during pregnancy (see also Evidence Table 28)

Recommendation Table 28

Recommendations for patients with AF during pregnancy (see also Evidence Table 28)

9.9. AF-CARE in congenital heart disease

Survival of patients with congenital heart disease has increased over time, but robust data on the management of AF are missing and available evidence is derived mainly from observational studies. Oral anticoagulants are recommended for all patients with AF and intracardiac repair, cyanotic congenital heart disease, Fontan palliation, or systemic right ventricle irrespective of the individuals' thromboembolic risk factors.897 Patients with AF and other congenital heart diseases should follow the general risk stratification for OAC use in AF (i.e. depending on the thromboembolic risk or CHA2DS2-VA score). Direct oral anticoagulants are contraindicated in patients with mechanical heart valves,331 but appear safe in patients with congenital heart disease,898,899 or those with a valvular bioprosthesis.900,901

Rate control drugs such as selective beta-1 receptor blockers, verapamil, diltiazem, and digoxin can be used with caution, with monitoring for bradycardia and hypotension. Rhythm control strategies such as amiodarone may be effective, but warrant monitoring for bradycardia. When cardioversion is planned, both 3 weeks of OAC and TOE should be considered because thrombi are common in patients with congenital heart disease and atrial arrhythmias.902,903 Ablation approaches can be successful in patients with congenital heart disease, but AF recurrence rates may be high (see Supplementary data online, Additional Evidence Table S30).

In patients with atrial septal defect, closure may be performed before the fourth decade of life to decrease the risk of AF or AFL.904 Patients with stroke who underwent closure of their PFO may have an increased risk of AF,905 but in patients with PFO and AF, PFO closure is not recommended for stroke prevention. AF surgery or catheter ablation can be considered at the time of closure of the atrial septal defect within a multidisciplinary team.906–908 AF catheter ablation of late atrial arrhythmias is likely to be effective after surgical atrial septal closure.909

Recommendation Table 29

Recommendations for patients with AF and congenital heart disease (see also Evidence Table 29)

Recommendation Table 29

Recommendations for patients with AF and congenital heart disease (see also Evidence Table 29)

9.10. AF-CARE in endocrine disorders

Endocrine dysfunction is closely related to AF, both as the direct action of endocrine hormones and as a consequence of treatments for endocrine disease. Optimal management of endocrine disorders is therefore part of the AF-CARE pathway.910,911

Clinical and subclinical hyperthyroidism, as well as subclinical hypothyroidism, are associated with an increased risk of AF.912,913 Patients presenting with new-onset or recurrent AF should be tested for thyroid-stimulating hormone (TSH) levels. The risk of AF is enhanced in vulnerable patients, including the elderly and those with structural atrial diseases,914,915 as well as cancer patients on immune checkpoint inhibitors.916,917 In hyperthyroidism, and even in the euthyroid range, the risk of AF increases according to the reduction in TSH and elevated levels of thyroxine.918,919 Moreover, the risk of stroke is higher in patients with hyperthyroidism, which can be mitigated by treating the thyroid disorder.920,921 Amiodarone induces thyroid dysfunction in 15%–20% of treated patients, leading to both hypo- and hyperthyroidism,922,923 which warrants referral to an endocrinologist (see Supplementary data online for further details).

Hypercalcaemia may also induce arrhythmias, but the role of primary hyperparathyroidism in incident AF is poorly studied. Surgical parathyroidectomy has been found to reduce both supraventricular and ventricular premature beats.924–926 Primary aldosteronism is related to an increased risk of AF through direct actions and vascular effects,927,928 with a three-fold higher rate of incident AF compared with patients with essential hypertension.929 Increases in genetically predicted plasma cortisol are associated with greater risk of AF, and patients with adrenal incidentalomas with subclinical cortisol secretion have a higher prevalence of AF.930,931 Acromegaly may predispose to an increased substrate for AF, with incident AF rates significantly higher than controls in long-term follow-up, even after adjusting for AF risk factors.932

The association between type 2 diabetes and AF is discussed in Sections 5.3 (AF recurrence) and Section 10.5 (incident AF). In addition to insulin-resistance mechanisms typical of type 2 diabetes, the loss of insulin signalling has recently been associated with electrical changes that can lead to AF. Type 1 diabetes is associated with an increased risk of several cardiovascular diseases including AF.933–937

9.11. AF-CARE in inherited cardiomyopathies and primary arrhythmia syndromes

A higher incidence and prevalence of AF have been described in patients with inherited cardiomyopathies and primary arrhythmia syndromes.271,938–970 AF can be the presenting or only clinically overt feature.969,971–975 AF in these patients is associated with adverse clinical outcomes,947,954,959,963,965,976–978 and has important implications on management (see Supplementary data online, Additional Evidence Table S31). When AF presents at a young age, there should be a careful interrogation about family history and a search for underlying disease.979

Rhythm control approaches may be challenging in patients with inherited cardiomyopathies and primary arrhythmia syndromes. For example, many drugs have a higher risk of adverse events or may be contraindicated (e.g. amiodarone and sotalol in congenital long QT syndrome, and Class IC AADs in Brugada syndrome) (see Supplementary Data online, Table S6). Owing to long-term adverse effects, chronic use of amiodarone is problematic in these typically young individuals. In patients with an implantable cardioverter defibrillator, AF is a common cause of inappropriate shocks.959,966,980,981 Programming a single high-rate ventricular fibrillation zone ≥210–220 b.p.m. with long detection time is safe,950,953,982 and is suggested in patients without documented slow monomorphic ventricular tachycardia. Implantation of an atrial lead may be considered in the case of significant bradycardia with beta-blocker treatment.

Patients with Wolff–Parkinson–White syndrome and AF are at risk of fast ventricular rates from rapid conduction of atrial electrical activity to the ventricles via the accessory pathway, potentially leading to ventricular fibrillation and sudden death.983,984 Immediate electrical cardioversion is needed for haemodynamically compromised patients with pre-excited AF, and atrioventricular node-modulating drugs should be avoided.985,986 Pharmacological cardioversion can be attempted using ibutilide987 or flecainide, while propafenone should be used with caution due to effects on the atrioventricular node.988,989 Amiodarone should be avoided in pre-excited AF due to its delayed action. Further details on inherited cardiomyopathies can be found in the 2023 ESC Guidelines for the management of cardiomyopathies.990

9.12. AF-CARE in cancer

All types of cancer show an increased risk of AF, with prevalence varying from 2% to 28%.991–995 The occurrence of AF may often be related to a pre-existing atrial substrate with vulnerability to AF. AF may be an indicator of an occult cancer, but also can appear in the context of concomitant surgery, chemotherapy, or radiotherapy.916,994,996 Risk of AF is dependent on, among other factors, the cancer type and stage,997 and is greater in older patients with pre-existing cardiovascular disease.991,993,994 Some procedures are associated with higher incidence of AF, including lung surgery (from 6% to 32%) and non-thoracic surgery such as a colectomy (4%–5%).994

Atrial fibrillation in the context of cancer is associated with a two-fold higher risk of systemic thromboembolism and stroke, and six-fold increased risk of heart failure.991,994 On the other hand, the coexistence of cancer increases the risk of all-cause mortality and major bleeding in patients with AF.998 Bleeding in those receiving OAC can also unmask the presence of cancer.999

Stroke risk scores may underestimate thromboembolic risk in cancer patients.1000 The association between cancer, AF, and ischaemic stroke also differs between cancer types. In some types of cancer, the risk of bleeding seems to exceed the risk of thromboembolism.998 Risk stratification is therefore complex in this population, and should be performed on an individual basis considering cancer type, stage, prognosis, bleeding risk, and other risk factors. These aspects can change within a short period of time, requiring dynamic assessment and management.

As with non-cancer patients, DOACs in those with cancer have similar efficacy and better safety compared with VKAs.1001–1010 Low molecular weight heparin is a short-term anticoagulation option, mostly during some cancer treatments, recent active bleeding, or thrombocytopaenia.1011 Decision-making on AF management, including on rhythm control, is best performed within a cardio-oncology multidisciplinary team.916,1012 Attention is required on interactions with cancer treatments, in particular QT-interval prolongation with AADs.

9.13. AF-CARE in older, multimorbid, or frail patients

Atrial fibrillation increases with age, and older patients more frequently have multimorbidity and frailty which are associated with worse clinical outcomes.1013–1016 Multimorbidity is the coexistence of two or more medically diagnosed diseases in the same individual. Frailty is defined as a person more vulnerable and less able to respond to a stressor or acute event, increasing the risk of adverse outcomes.1016,1017 The prevalence of frailty in AF varies due to different methods of assessment from 4.4% to 75.4%, and AF prevalence in the frail population ranges from 48.2% to 75.4%.1018 Frailty status is a strong independent risk factor for new-onset AF among older adults with hypertension.1019

Atrial fibrillation in frail patients is associated with less use of OAC and lower rates of management with a rhythm control strategy.1015,1018,1020 Oral anticoagulation initiation in older, frail multimorbid AF patients has improved since the introduction of DOACs, but is still lower in AF patients at older age (OR, 0.98 per year; 95% CI, 0.98–0.98), with dementia (OR, 0.57; 95% CI, 0.55–0.58), or frailty (OR, 0.74; 95% CI, 0.72–0.76).1021 The value of observational data which show potential benefit from OAC (in particular, DOACs) is limited due to prescription biases.1022–1027 Frail patients aged ≥75 years with polypharmacy and stable on a VKA may remain on the VKA rather than switching to a DOAC (Section 6.2).309

9.14. AF-CARE in atrial flutter

Due to the association between AFL and thromboembolic outcomes, and the frequent development of AF in patients with AFL, the management of comorbidities and risk factors in AFL should mirror that for AF (see Section 5). Similarly, the approach to prevent thromboembolism in AFL includes peri-procedural and long-term OAC (see Section 6). Rate control can be difficult to achieve in AFL, despite combination therapy. Rhythm control is often the first-line approach,983 with small randomized trials showing that cavo-tricuspid isthmus (CTI) ablation is superior to AADs.1028,1029 Recurrence of AFL is uncommon after achieving and confirming bidirectional block in typical CTI-dependent AFL. However, the majority of patients (50%–70%) have manifested AF during long-term follow-up in observational studies after AFL ablation.1030,1031 Hence the necessity for long-term dynamic re-evaluation in all patients with AFL in keeping with the AF-CARE approach. More detail on the management of AFL and other atrial arrhythmias is described in the 2019 ESC Guidelines for the management of patients with supraventricular tachycardia.983

Recommendation Table 30

Recommendations for prevention of thromboembolism in atrial flutter (see also Evidence Table 30)

Recommendation Table 30

Recommendations for prevention of thromboembolism in atrial flutter (see also Evidence Table 30)

10. Screening and prevention of AF

10.1. Epidemiology of AF

Atrial fibrillation is the most common sustained arrhythmia worldwide, with an estimated global prevalence in 2019 of 59.7 million persons with AF.1033 Incident cases of AF are doubling every few decades.1034 Future increases are anticipated, in particular in middle-income countries.1034 In community-based individuals, the prevalence of AF in a United States of America cohort was up to 5.9%.1035 The age-standardized prevalence and incidence rates have remained constant over time.1033,1036 The increase in overall prevalence is largely attributable to population growth, ageing, and survival from other cardiac conditions. In parallel, increases in risk factor burden, better awareness, and improved detection of AF have been observed.1037 The lifetime risk of AF has been estimated to be as high as 1 in 3 for older individuals,1038 with age-standardized incidence rates higher for men than women. Populations of European ancestry are typically found to have higher AF prevalence, individuals of African ancestry have worse outcomes, and other groups may have less access to interventions.1039–1041 Socioeconomic and other factors likely play a role in racial and ethnic differences in AF, but studies are also limited due to differences in how groups access healthcare. Greater deprivation in socioeconomic and living status is associated with higher AF incidence.1042

10.2. Screening tools for AF

In recent years, an abundance of novel devices that can monitor heart rhythm have come to the market, including fitness bands and smartwatches. Although the evidence for clinical effectiveness of digital devices is limited, they may be useful in detecting AF, and their clinical, economic, legal, and policy implications merit further investigation.1043,1044 Devices for AF detection can broadly be divided into those that provide an ECG, and those with non-ECG approaches such as photoplethysmography (Figure 15 and Table 15).

Non-invasive diagnostic methods for AF screening.
Figure 15

Non-invasive diagnostic methods for AF screening.

AF, atrial fibrillation; BP, blood pressure; ECG, electrocardiogram; PPG, photoplethysmography.

Table 15

Tools for AF screening

Tools for AF screening
  • (i) Pulse palpation1045

  • (ii) Use of artificial intelligence algorithms to identify patients at risk1046

  • (iii) ECG-based devices

    • (a) Conventional ECG devices

      • (1) Classic 12-lead ECG 1047

      • (2) Holter monitoring (from 24 h to a week or more)1048

      • (3) Mobile cardiac telemetry (during hospitalization)1049

      • (4) Handheld devices1050–1052

      • (5) Wearable patches (up to 14 days)1053–1067

      • (6) Biotextiles (up to 30 days)1068–1072

      • (7) Smart devices (30 s)1073–1091

    • (b) Implantable loop recorders (3–5 years)1092–1099

  • (iv) Non-ECG-based devices

    • (a) Photoplethysmography and automatic algorithms: contact (fingertip, smart device, band) and contactless (video)1100–1106

    • (b) Oscillometry (blood pressure monitors that derive heart rhythm regularity algorithmically)1107–1110

    • (c) Mechanocardiography (accelerometers and gyroscopes to sense the mechanical activity of the heart)1111

    • (d) Contactless video plethysmography (through video monitoring)1112–1115

    • (e) Smart speakers (through the identification of abnormal heart rate patterns)1116

Tools for AF screening
  • (i) Pulse palpation1045

  • (ii) Use of artificial intelligence algorithms to identify patients at risk1046

  • (iii) ECG-based devices

    • (a) Conventional ECG devices

      • (1) Classic 12-lead ECG 1047

      • (2) Holter monitoring (from 24 h to a week or more)1048

      • (3) Mobile cardiac telemetry (during hospitalization)1049

      • (4) Handheld devices1050–1052

      • (5) Wearable patches (up to 14 days)1053–1067

      • (6) Biotextiles (up to 30 days)1068–1072

      • (7) Smart devices (30 s)1073–1091

    • (b) Implantable loop recorders (3–5 years)1092–1099

  • (iv) Non-ECG-based devices

    • (a) Photoplethysmography and automatic algorithms: contact (fingertip, smart device, band) and contactless (video)1100–1106

    • (b) Oscillometry (blood pressure monitors that derive heart rhythm regularity algorithmically)1107–1110

    • (c) Mechanocardiography (accelerometers and gyroscopes to sense the mechanical activity of the heart)1111

    • (d) Contactless video plethysmography (through video monitoring)1112–1115

    • (e) Smart speakers (through the identification of abnormal heart rate patterns)1116

ECG, electrocardiogram.

© ESC 2024
Table 15

Tools for AF screening

Tools for AF screening
  • (i) Pulse palpation1045

  • (ii) Use of artificial intelligence algorithms to identify patients at risk1046

  • (iii) ECG-based devices

    • (a) Conventional ECG devices

      • (1) Classic 12-lead ECG 1047

      • (2) Holter monitoring (from 24 h to a week or more)1048

      • (3) Mobile cardiac telemetry (during hospitalization)1049

      • (4) Handheld devices1050–1052

      • (5) Wearable patches (up to 14 days)1053–1067

      • (6) Biotextiles (up to 30 days)1068–1072

      • (7) Smart devices (30 s)1073–1091

    • (b) Implantable loop recorders (3–5 years)1092–1099

  • (iv) Non-ECG-based devices

    • (a) Photoplethysmography and automatic algorithms: contact (fingertip, smart device, band) and contactless (video)1100–1106

    • (b) Oscillometry (blood pressure monitors that derive heart rhythm regularity algorithmically)1107–1110

    • (c) Mechanocardiography (accelerometers and gyroscopes to sense the mechanical activity of the heart)1111

    • (d) Contactless video plethysmography (through video monitoring)1112–1115

    • (e) Smart speakers (through the identification of abnormal heart rate patterns)1116

Tools for AF screening
  • (i) Pulse palpation1045

  • (ii) Use of artificial intelligence algorithms to identify patients at risk1046

  • (iii) ECG-based devices

    • (a) Conventional ECG devices

      • (1) Classic 12-lead ECG 1047

      • (2) Holter monitoring (from 24 h to a week or more)1048

      • (3) Mobile cardiac telemetry (during hospitalization)1049

      • (4) Handheld devices1050–1052

      • (5) Wearable patches (up to 14 days)1053–1067

      • (6) Biotextiles (up to 30 days)1068–1072

      • (7) Smart devices (30 s)1073–1091

    • (b) Implantable loop recorders (3–5 years)1092–1099

  • (iv) Non-ECG-based devices

    • (a) Photoplethysmography and automatic algorithms: contact (fingertip, smart device, band) and contactless (video)1100–1106

    • (b) Oscillometry (blood pressure monitors that derive heart rhythm regularity algorithmically)1107–1110

    • (c) Mechanocardiography (accelerometers and gyroscopes to sense the mechanical activity of the heart)1111

    • (d) Contactless video plethysmography (through video monitoring)1112–1115

    • (e) Smart speakers (through the identification of abnormal heart rate patterns)1116

ECG, electrocardiogram.

© ESC 2024

Most consumer-based devices use photoplethysmography, and several large studies have been performed typically in low-risk individuals.633,1076,1117,1118 In an RCT of 5551 participants invited by their health insurer, smartphone-based photoplethysmography increased the odds of OAC-treated new AF by 2.12 (95% CI, 1.19–3.76; P = .01) compared with usual care.605 RCTs powered for assessment of clinical outcomes are still lacking for consumer-based AF screening. Further head-to-head comparisons between novel digital devices and those commonly used in healthcare settings are needed to establish their comparative effectiveness in the clinical setting and account for different populations and settings.1119 In a systematic review of smartphone-based photoplethysmography compared with a reference ECG, unrealistically high sensitivity and specificity were noted, likely due to small, low-quality studies with a high degree of patient selection bias.1120 Hence, when AF is suggested by a photoplethysmography device or any other screening tool, a single-lead or continuous ECG tracing of >30 s or 12-lead ECG showing AF analysed by a physician with expertise in ECG rhythm interpretation is recommended to establish a definitive diagnosis of AF.1091,1121–1125

The combination of big data and artificial intelligence (AI) is having an increasing impact on the field of electrophysiology. Algorithms have been created to improve automated AF diagnosis and several algorithms to aid diagnostics are being investigated.1046 However, the clinical performance and broad applicability of these solutions are not yet known. The use of AI may enable future treatment changes to be assessed with dynamic and continuous patient-directed monitoring using wearable devices.1126 There are still challenges in the field that need clarification, such as data acquisition, model performance, external validity, clinical implementation, algorithm interpretation, and confidence, as well as the ethical aspects.1127

10.3. Screening strategies for AF

Screening can be performed systematically, with an invitation issued to a patient, or opportunistically, at the time of an ad hoc meeting with a healthcare professional. Regardless of the mode of invitation, screening should be part of a structured programme1128 and is not the same as identification of AF during a routine healthcare visit or secondary to arrhythmia symptoms.

Screening can be done at a single timepoint (snapshot of the heart rhythm), e.g. using pulse palpation or a 12-lead ECG. Screening can also be of an extended duration, i.e. prolonged, using either intermittent or continuous monitoring of heart rhythm. Most studies using an opportunistic strategy have screened for AF at a single timepoint with short duration (such as a single timepoint ECG), compared with systematic screening studies that have mainly used prolonged (repeated or continuous) rhythm assessment.1129 The optimal screening method will vary depending on the population being studied (Figure 16) (see Supplementary data online, Additional Evidence Table S32). More sensitive methods will detect more AF but may lead to an increased risk of false positives and an increased detection of low burden AF, whereas more specific methods result in less false positives, at the risk of missing AF.

Approaches to screening for AF.
Figure 16

Approaches to screening for AF.

AF, atrial fibrillation; AF-CARE, atrial fibrillation—[C] Comorbidity and risk factor management, [A] Avoid stroke and thromboembolism, [R] Reduce symptoms by rate and rhythm control, [E] Evaluation and dynamic reassessment; CHA2DS2-VA, congestive heart failure, hypertension, age ≥75 years (2 points), diabetes mellitus, prior stroke/TIA/arterial thromboembolism (2 points), vascular disease, age 65–74 years; ECG, electrocardiogram. See Figure 15 for non-invasive ECG methods.

Invasive monitoring of heart rhythm in high-risk populations extended for several years has been shown to result in device-detected AF prevalence of around 30%, albeit most of whom have a low burden of AF.5,857,1130,1131 Pacemaker studies have shown that patients with a low burden of device-detected subclinical AF have a lower risk of ischaemic stroke.5,24,1131,1132 This has been confirmed in RCTs assessing DOAC use in patients with device-detected subclinical AF (see Section 6.1.1).5,281,282 The burden needed for device-detected subclinical AF to translate into stroke risk is not known, and further studies are clearly needed.1133,1134 Benefit and cost-effectiveness of screening are discussed in the Supplementary data online.

Recommendation Table 31

Recommendations for screening for AF (see also Evidence Table 31)

Recommendation Table 31

Recommendations for screening for AF (see also Evidence Table 31)

10.3.1. Single timepoint screening ‘snapshot’

Several cluster RCTs in primary care settings have explored whether screening performed as a snapshot of the heart rhythm at one timepoint can detect more AF compared with usual care in individuals aged ≥65 years.1138–1140 No increased detection of AF was seen in groups randomized to single timepoint screening.1138–1140 These findings were confirmed in a meta-analysis of RCTs showing that screening as a one-time event did not increase detection of AF compared with usual care.1135 Notably, these studies were performed in healthcare settings where the detection of AF in the population might be high, hence the results might not be generalizable to healthcare settings with a lower spontaneous AF detection. There are no RCTs addressing clinical outcomes in patients with AF detected by single timepoint screening.1123,1135

10.3.2. Prolonged screening

Studies using prolonged screening have shown an increased detection of AF leading to initiation of OAC.1129,1135,1141 Two RCTs have investigated the effect on clinical outcomes in prolonged screening for AF.5,6 In the STROKESTOP trial (Systematic ECG Screening for Atrial Fibrillation Among 75 Year Old Subjects in the Region of Stockholm and Halland, Sweden), 75- and 76-year-olds were randomized to be invited to prolonged screening for AF using single-lead ECGs twice daily for 2 weeks, or to standard of care. After a median of 6.9 years there was a small reduction in the primary combined endpoint of all-cause mortality, stroke, systematic embolism, and severe bleeding in favour of prolonged screening (HR, 0.96; 95% CI, 0.92–1.00; P = .045).6 In the LOOP (Atrial Fibrillation Detected by Continuous ECG Monitoring) trial, individuals at increased risk of stroke were randomized to receive an implantable loop recorder that monitored heart rhythm for an average of 3.3 years, or to a control group receiving standard of care. Although there was a higher detection of AF (31.8%) and subsequent initiation of OAC in the loop recorder group compared with standard of care (12.2%), this was not accompanied by a difference in the primary outcome of stroke or systemic embolism.5 In a meta-analysis of recent RCTs on the outcome of stroke, a small but significant benefit was seen in favour of prolonged screening (RR, 0.91; 95% CI, 0.84–0.99).1136 This was not repeated in a second meta-analysis including older RCTs, where no risk reduction was seen with regard to mortality or stroke.1135 Notably, both these meta-analyses are likely underpowered to assess clinical outcomes.

10.4. Factors associated with incident AF

The most common risk predictors for incident (new-onset) AF are shown in Table 16. While the factors listed are robustly associated with incident AF in observational studies, it is not known whether the relationships are causal. Studies using Mendelian randomization (genetic proxies for risk factors to estimate causal effects) robustly implicate systolic BP and higher BMI as causal risk factors for incident AF.1142

Table 16

Factors associated with incident AF

Demographic factorsAge1149–1151
Male sex1149–1152
European ancestry1149,1150
Lower socioeconomic status1150
Lifestyle behavioursSmoking/tobacco use1149–1151
Alcohol intake1149,1150
Physical inactivity1149,1150
Vigorous exercise1153–1156
Competitive or athlete-level endurance sports1151,1157
Caffeine1158–1160
Comorbidities and risk factorsHypertension1149–1151
Heart failure178,1149–1151,1161
Valvular disease1149,1151,1162–1164
Coronary artery disease1149,1151,1161,1165
Peripheral arterial disease785
Congenital heart disease1149,1166
Heart rate, heart rate variability1167,1168
Total cholesterol1149,1150
Low-density lipoprotein cholesterol1150
High-density lipoprotein cholesterol1150
Triglycerides1150
Impaired glucose tolerance,1169–1172diabetes mellitus1149–1151,1169
Renal dysfunction/CKD1149–1151,1173,1174
Obesity1149–1151,1175,1176
Body mass index, weight1149–1151
Height1150
Sleep apnoea1149,1151,1177,1178
Chronic obstructive pulmonary disease1179
Subclinical atherosclerosisCoronary artery calcification1149,1151,1180
Carotid IMT and carotid plaque1149,1151,1181,1182
ECG abnormalitiesPR interval prolongation1149,1151,1183
Sick sinus syndrome1149,1184,1185
Wolff–Parkinson–White1149,1186
Genetic factorsFamily history of AF1149,1151,1187–1190
AF-susceptible loci identified by GWAS1149,1151,1191,1192
Short QT syndrome1149
Genetic cardiomyopathies990,1193
BiomarkersC-reactive protein1150,1151
Fibrinogen1150
Growth differentiation factor-151194
Natriuretic peptides (atrial and B-type)1195–1200
Cardiac troponins1199
Inflammatory biomarkers 1149,1151
OthersThyroid dysfunction912,1149–1151
Autoimmune diseases1150
Air pollution1149,1201
Sepsis1149,1202
Psychological factors 1203,1204
Demographic factorsAge1149–1151
Male sex1149–1152
European ancestry1149,1150
Lower socioeconomic status1150
Lifestyle behavioursSmoking/tobacco use1149–1151
Alcohol intake1149,1150
Physical inactivity1149,1150
Vigorous exercise1153–1156
Competitive or athlete-level endurance sports1151,1157
Caffeine1158–1160
Comorbidities and risk factorsHypertension1149–1151
Heart failure178,1149–1151,1161
Valvular disease1149,1151,1162–1164
Coronary artery disease1149,1151,1161,1165
Peripheral arterial disease785
Congenital heart disease1149,1166
Heart rate, heart rate variability1167,1168
Total cholesterol1149,1150
Low-density lipoprotein cholesterol1150
High-density lipoprotein cholesterol1150
Triglycerides1150
Impaired glucose tolerance,1169–1172diabetes mellitus1149–1151,1169
Renal dysfunction/CKD1149–1151,1173,1174
Obesity1149–1151,1175,1176
Body mass index, weight1149–1151
Height1150
Sleep apnoea1149,1151,1177,1178
Chronic obstructive pulmonary disease1179
Subclinical atherosclerosisCoronary artery calcification1149,1151,1180
Carotid IMT and carotid plaque1149,1151,1181,1182
ECG abnormalitiesPR interval prolongation1149,1151,1183
Sick sinus syndrome1149,1184,1185
Wolff–Parkinson–White1149,1186
Genetic factorsFamily history of AF1149,1151,1187–1190
AF-susceptible loci identified by GWAS1149,1151,1191,1192
Short QT syndrome1149
Genetic cardiomyopathies990,1193
BiomarkersC-reactive protein1150,1151
Fibrinogen1150
Growth differentiation factor-151194
Natriuretic peptides (atrial and B-type)1195–1200
Cardiac troponins1199
Inflammatory biomarkers 1149,1151
OthersThyroid dysfunction912,1149–1151
Autoimmune diseases1150
Air pollution1149,1201
Sepsis1149,1202
Psychological factors 1203,1204

AF, atrial fibrillation; CKD, chronic kidney disease; GWAS, genome-wide association studies; HF, heart failure; IMT, intima-media thickness.

© ESC 2024
Table 16

Factors associated with incident AF

Demographic factorsAge1149–1151
Male sex1149–1152
European ancestry1149,1150
Lower socioeconomic status1150
Lifestyle behavioursSmoking/tobacco use1149–1151
Alcohol intake1149,1150
Physical inactivity1149,1150
Vigorous exercise1153–1156
Competitive or athlete-level endurance sports1151,1157
Caffeine1158–1160
Comorbidities and risk factorsHypertension1149–1151
Heart failure178,1149–1151,1161
Valvular disease1149,1151,1162–1164
Coronary artery disease1149,1151,1161,1165
Peripheral arterial disease785
Congenital heart disease1149,1166
Heart rate, heart rate variability1167,1168
Total cholesterol1149,1150
Low-density lipoprotein cholesterol1150
High-density lipoprotein cholesterol1150
Triglycerides1150
Impaired glucose tolerance,1169–1172diabetes mellitus1149–1151,1169
Renal dysfunction/CKD1149–1151,1173,1174
Obesity1149–1151,1175,1176
Body mass index, weight1149–1151
Height1150
Sleep apnoea1149,1151,1177,1178
Chronic obstructive pulmonary disease1179
Subclinical atherosclerosisCoronary artery calcification1149,1151,1180
Carotid IMT and carotid plaque1149,1151,1181,1182
ECG abnormalitiesPR interval prolongation1149,1151,1183
Sick sinus syndrome1149,1184,1185
Wolff–Parkinson–White1149,1186
Genetic factorsFamily history of AF1149,1151,1187–1190
AF-susceptible loci identified by GWAS1149,1151,1191,1192
Short QT syndrome1149
Genetic cardiomyopathies990,1193
BiomarkersC-reactive protein1150,1151
Fibrinogen1150
Growth differentiation factor-151194
Natriuretic peptides (atrial and B-type)1195–1200
Cardiac troponins1199
Inflammatory biomarkers 1149,1151
OthersThyroid dysfunction912,1149–1151
Autoimmune diseases1150
Air pollution1149,1201
Sepsis1149,1202
Psychological factors 1203,1204
Demographic factorsAge1149–1151
Male sex1149–1152
European ancestry1149,1150
Lower socioeconomic status1150
Lifestyle behavioursSmoking/tobacco use1149–1151
Alcohol intake1149,1150
Physical inactivity1149,1150
Vigorous exercise1153–1156
Competitive or athlete-level endurance sports1151,1157
Caffeine1158–1160
Comorbidities and risk factorsHypertension1149–1151
Heart failure178,1149–1151,1161
Valvular disease1149,1151,1162–1164
Coronary artery disease1149,1151,1161,1165
Peripheral arterial disease785
Congenital heart disease1149,1166
Heart rate, heart rate variability1167,1168
Total cholesterol1149,1150
Low-density lipoprotein cholesterol1150
High-density lipoprotein cholesterol1150
Triglycerides1150
Impaired glucose tolerance,1169–1172diabetes mellitus1149–1151,1169
Renal dysfunction/CKD1149–1151,1173,1174
Obesity1149–1151,1175,1176
Body mass index, weight1149–1151
Height1150
Sleep apnoea1149,1151,1177,1178
Chronic obstructive pulmonary disease1179
Subclinical atherosclerosisCoronary artery calcification1149,1151,1180
Carotid IMT and carotid plaque1149,1151,1181,1182
ECG abnormalitiesPR interval prolongation1149,1151,1183
Sick sinus syndrome1149,1184,1185
Wolff–Parkinson–White1149,1186
Genetic factorsFamily history of AF1149,1151,1187–1190
AF-susceptible loci identified by GWAS1149,1151,1191,1192
Short QT syndrome1149
Genetic cardiomyopathies990,1193
BiomarkersC-reactive protein1150,1151
Fibrinogen1150
Growth differentiation factor-151194
Natriuretic peptides (atrial and B-type)1195–1200
Cardiac troponins1199
Inflammatory biomarkers 1149,1151
OthersThyroid dysfunction912,1149–1151
Autoimmune diseases1150
Air pollution1149,1201
Sepsis1149,1202
Psychological factors 1203,1204

AF, atrial fibrillation; CKD, chronic kidney disease; GWAS, genome-wide association studies; HF, heart failure; IMT, intima-media thickness.

© ESC 2024

A high degree of interaction occurs between all factors related to AF development (see Supplementary data online, Additional Evidence Table S33).1038,1039,1143–1145 For ease of clinical application, risk prediction tools have combined various factors, and have recently employed machine learning algorithms for prediction.1146,1147 Classical risk scores are also available with variable predictive ability and model performance (see Supplementary data online, Table S7).1148 Improved outcomes when using these risk scores have yet to be demonstrated. Although knowledge is rapidly increasing about the genetic basis for AF in some patients, the value of genetic screening is limited at the present time (see Supplementary data online).

10.5. Primary prevention of AF

Preventing the onset of AF before clinical manifestation has clear potential to improve the lives of the general population and reduce the considerable health and social care costs associated with development of AF. Whereas the [C] in AF-CARE is focused on the effective management of risk factors and comorbidities to limit AF recurrence and progression, there is also evidence they can be targeted to prevent AF. Available data are presented below for hypertension, heart failure, type 2 diabetes mellitus, obesity, sleep apnoea syndrome, physical activity, and alcohol, although many other risk markers can also be targeted. Further information on each factor's attributable risk for AF is provided in the Supplementary data online (see Supplementary data online, Evidence Table 32 and additional Evidence Tables S34–S39).

Recommendation Table 32

Recommendations for primary prevention of AF (see also Evidence Table 32)

Recommendation Table 32

Recommendations for primary prevention of AF (see also Evidence Table 32)

10.5.1. Hypertension

Management of hypertension has been associated with a reduction in incident AF.1205–1207,1232 In the LIFE (Losartan Intervention for End point reduction in hypertension) trial, a 10 mmHg reduction in systolic BP was associated with a 17% reduction in incident AF.1207 Secondary analysis of RCTs and observational studies suggest that ACE inhibitors or ARBs may be superior to beta-blockers, calcium channel blockers, or diuretics for the prevention of incident AF.1233–1236

10.5.2. Heart failure

Long-standing established pharmacological treatments for HFrEF have been associated with a reduction in incident AF. The use of ACE inhibitors or ARBs in patients with known HFrEF was associated with a 44% reduction in incidence of AF.1208 Similarly, beta-blockers in HFrEF led to a 33% reduction in the odds of incident AF.133 Mineralocorticoid receptor antagonists have also been shown to reduce the risk of new-onset AF by 42% in patients with HFrEF.1209 Although there have been variable effects of SGLT2 inhibitors on incident AF, several meta-analyses have demonstrated that there is an 18%–37% reduction in incident AF.136,1210,1211,1237 However, treatment of HFrEF with sacubitril/valsartan has not yet been shown to confer any adjunctive benefit in reducing new-onset AF when compared with ACE inhibitors/ARBs alone.1238 There is some evidence to suggest that effective CRT in eligible patients with HFrEF reduces the risk of incident AF.1239 To date, no treatments in HFpEF have been shown to reduce incident AF.

10.5.3. Type 2 diabetes mellitus

The integrated care of type 2 diabetes, based on lifestyle and pharmacological treatments for comorbidities such as obesity, hypertension, and dyslipidaemia, are useful steps in preventing atrial remodelling and subsequent AF. Intensive glucose-lowering therapy targeting an HbA1c level of <6.0% (<42 mmol/mol) failed to show a protective effect on incident AF.1240 More than glycaemic control per se, the class of glucose-lowering agent may influence the risk of AF.1240 Insulin promotes adipogenesis and cardiac fibrosis, and sulfonylureas have been consistently associated with an increased risk of AF.193 Observational studies have associated metformin with lower rates of incident AF.1224,1225,1241–1243 Various recent studies and meta-analyses point to the positive role of SGLT2 inhibitors to reduce the risk of incident AF in diabetic and non-diabetic patients.136,1226,1244–1246 Pooled data from 22 trials including 52 951 patients with type 2 diabetes and heart failure showed that SGLT2 inhibitors compared with placebo can significantly reduce the incidence of AF by 18% in studies on diabetes, and up to 37% in heart failure with or without type 2 diabetes.1210,1211

10.5.4. Obesity

Management of weight is important in the prevention of AF. In a large population-based cohort study, normal weight was associated with a reduced risk of incident AF compared with those who were obese (4.7% increase in the risk of incident AF for each 1 kg/m2 increase of BMI).208 In the Women's Health Study, participants who became obese had a 41% increased risk of incident AF compared with those who maintained their BMI <30 kg/m2.1212 Similarly, observational studies in populations using bariatric surgery for weight loss in morbidly obese individuals (BMI ≥40 kg/m2) have observed a lower risk of incident AF.1227–1231

10.5.5. Sleep apnoea syndrome

Although it would seem rational to optimize sleep habits, to date there is no conclusive evidence to support this for the primary prevention of AF. The SAVE (Sleep Apnea cardioVascular Endpoints) trial failed to demonstrate a difference in clinical outcomes in those randomized to CPAP therapy or placebo.230 There was no difference in incident AF, albeit the analysis of AF was not based on systematic screening but rather on clinically documented AF.

10.5.6. Physical activity

Several studies have demonstrated beneficial effects of moderate physical activity on cardiovascular health.1247 Moderate aerobic exercise may also reduce the risk of new-onset AF.1214–1219 It should be noted that the incidence of AF appears to be increased among athletes, with a meta-analysis of observational studies showing a 2.5-fold increased risk of AF compared with non-athlete controls.1248

10.5.7. Alcohol intake

The premise that reducing alcohol intake can prevent AF is based on observational studies linking alcohol to an excess risk of incident AF in a dose-dependent manner (see Supplementary data online).1220–1222 In addition, a population cohort study of those with high alcohol consumption (>60 g/day for men and >40 g/day for women) found that abstinence from alcohol was associated with a lower incidence of AF compared with patients who continued heavy drinking.1223

11. Key messages

  1. General management: optimal treatment according to the AF-CARE pathway, which includes: [C] Comorbidity and risk factor management; [A] Avoid stroke and thromboembolism; [R] Reduce symptoms by rate and rhythm control; and [E] Evaluation and dynamic reassessment.

  2. Shared care: patient-centred AF management with joint decision-making and a multidisciplinary team.

  3. Equal care: avoid health inequalities based on gender, ethnicity, disability, and socioeconomic factors.

  4. Education: for patients, family members, caregivers, and healthcare professionals to aid shared decision-making.

  5. Diagnosis: clinical AF requires confirmation on an ECG device to initiate risk stratification and AF management.

  6. Initial evaluation: medical history, assessment of symptoms and their impact, blood tests, echocardiography/other imaging, patient-reported outcome measures, and risk factors for thromboembolism and bleeding.

  7. Comorbidities and risk factors: thorough evaluation and management critical to all aspects of care for patients with AF to avoid recurrence and progression of AF, improve success of AF treatments, and prevent AF-related adverse outcomes.

  8. Focus on conditions associated with AF: including hypertension, heart failure, diabetes mellitus, obesity, obstructive sleep apnoea, physical inactivity, and high alcohol intake.

  9. Assessing the risk of thromboembolism: use locally validated risk tools or the CHA2DS2-VA score and assessment of other risk factors, with reassessment at periodic intervals to assist in decisions on anticoagulant prescription.

  10. Oral anticoagulants: recommended for all eligible patients, except those at low risk of incident stroke or thromboembolism (CHA2DS2-VA = 1 anticoagulation should be considered; CHA2DS2-VA ≥2 anticoagulation recommended).

  11. Choice of anticoagulant: DOACs (apixaban, dabigatran, edoxaban, and rivaroxaban) are preferred over VKAs (warfarin and others), except in patients with mechanical heart valves and mitral stenosis.

  12. Dose/range of anticoagulant: use full standard doses for DOACs unless the patient meets specific dose-reduction criteria; for VKAs, keep INR generally 2.0–3.0, and in range for >70% of the time.

  13. Switching anticoagulants: switch from a VKA to DOAC if risk of intracranial haemorrhage or poor control of INR levels.

  14. Bleeding risk: modifiable bleeding risk factors should be managed to improve safety; bleeding risk scores should not be used to decide on starting or withdrawing anticoagulants.

  15. Antiplatelet therapy: avoid combining anticoagulants and antiplatelet agents, unless the patient has an acute vascular event or needs interim treatment for procedures.

  16. Rate control therapy: use beta-blockers (any ejection fraction), digoxin (any ejection fraction), or diltiazem/verapamil (LVEF >40%) as initial therapy in the acute setting, an adjunct to rhythm control therapies, or as a sole treatment strategy to control heart rate and symptoms.

  17. Rhythm control: consider in all suitable AF patients, explicitly discussing with patients all potential benefits and risks of cardioversion, antiarrhythmic drugs, and catheter or surgical ablation to reduce symptoms and morbidity.

  18. Safety first: keep safety and anticoagulation in mind when considering rhythm control; e.g. delay cardioversion and provide at least 3 weeks of anticoagulation beforehand if AF duration >24 h, and consider toxicity and drug interactions for antiarrhythmic therapy.

  19. Cardioversion: use electrical cardioversion in cases of haemodynamic instability; otherwise choose electrical or pharmacological cardioversion based on patient characteristics and preferences.

  20. Indication for long-term rhythm control: the primary indication should be reduction in AF-related symptoms and improvement in quality of life; for selected patient groups, sinus rhythm maintenance can be pursued to reduce morbidity and mortality.

  21. Success or failure of rhythm control: continue anticoagulation according to the patient's individual risk of thromboembolism, irrespective of whether they are in AF or sinus rhythm.

  22. Catheter ablation: consider as second-line option if antiarrhythmic drugs fail to control AF, or first-line option in patients with paroxysmal AF.

  23. Endoscopic or hybrid ablation: consider if catheter ablation fails, or an alternative to catheter ablation in persistent AF despite antiarrhythmic drugs.

  24. Atrial fibrillation ablation during cardiac surgery: perform in centres with experienced teams, especially for patients undergoing mitral valve surgery.

  25. Dynamic evaluation: periodically reassess therapy and give attention to new modifiable risk factors that could slow/reverse the progression of AF, increase quality of life, and prevent adverse outcomes.

12. Gaps in evidence

The following bullet list gives the most important gaps in evidence where new clinical trials could substantially aid the patient pathway:

Definition and clinical impact of AF

  • Paroxysmal AF is not one entity, and patterns of AF progression and regression are highly variable. It is uncertain what the relevance is for treatment strategies and management decisions.

  • Thirty seconds as definition for clinical AF needs validation and evaluation whether it is related to AF-related outcomes.

  • Definition, clinical features, diagnosis, and implementation for treatment choices of atrial cardiomyopathy in patients with AF is unsettled.

  • Diversity in AF presentation, underlying pathophysiological mechanisms, and associated comorbidities is incompletely understood with regard to differences in sex, gender, race/ethnicity, socioeconomic state, education, and differences between low-, moderate-, and high-income countries.

  • Personalized risk prediction for AF incidence, AF progression, and associated outcomes remains challenging.

  • Insights into psychosocial and environmental factors and risk of AF and adverse outcomes in AF are understudied.

Patient-centred, multidisciplinary AF management

  • The benefit of additional education directed to patients, to family members, and to healthcare professionals in order to optimize shared decision-making still needs to be proved.

  • Access to patient-centred management according to the AF-CARE principles to ensure equality in healthcare provision and improve outcomes warrants evidence.

  • The place of remote monitoring and telemedicine for identification and follow-up of patients with AF, or its subgroups is non-established, though widely applied.

[C] Comorbidity and risk factor management

  • Methods to achieve consistent and reproducible weight loss in patients with AF requires substantial improvement. Despite some evidence demonstrating the benefits of weight loss, widespread adoption has been limited by the need for reproducible strategies.

  • The importance of sleep apnoea syndrome and its treatment on AF-related outcomes remains to be elucidated.

[A] Avoid stroke and thromboembolism

  • Data are lacking on how to treat patients with low risk of stroke (with a CHA2DS2-VA score of 0 or 1), as these patients were excluded from large RCTs.

  • Not enough evidence is available for OAC in elderly patients, frail polypharmacy patients, those with cognitive impairment/dementia, recent bleeding, previous ICH, severe end-stage renal failure, liver impairment, cancer, or severe obesity.

  • In elderly patients, routinely switching VKAs to DOACs is associated with increased bleeding risk; however, the reasons why this happens are unclear.

  • The selection of which patients with asymptomatic device-detected subclinical AF benefit from OAC therapy needs to be defined.

  • There is a lack of evidence whether and when to (re)start anticoagulation after intracranial haemorrhage.

  • There is lack of evidence about optimal anticoagulation in patients with ischaemic stroke or left atrial thrombus while being treated with OAC.

  • There is uncertainty about the place of LAA closure and how to manage antithrombotic post-procedural management when LAAO is performed.

  • Balance of thromboembolism and bleeding is unclear in patients with AF and incidental cerebral artery aneurysms identified on brain MRI.

[R] Reduce symptoms by rate and rhythm control

  • In some patients, AF can be benign in terms of symptoms and outcomes. In which patients rhythm control is not needed warrants investigation.

  • Application of antiarrhythmic drugs has been hampered by poor effectiveness and side effects; however, new antiarrhythmic drugs are needed to increase the therapeutic arsenal for AF patients.

  • The amount of AF reduction obtained by rhythm control to improve outcomes is unknown.

  • Large catheter ablation studies showed no improved outcome of patients with AF. Some small studies in specific subpopulations have observed an improved outcome. This warrants further investigation to provide each patient with AF with personalized treatment goals.

  • Uncertainty exists on the time of duration of AF and risk of stroke when performing a cardioversion.

  • The value of diagnostic cardioversion for persistent AF in steering management of AF is unknown.

  • Decisions on continuation of OAC are completely based on stroke risk scores and irrespective of having (episodes) of AF; whether this holds for patients undergoing successful catheter ablation is uncertain.

  • Large variability in ablation strategies and techniques exist for patients with persistent AF, or after first failed catheter ablation for paroxysmal AF. The optimal catheter ablation strategy and techniques, however, are unknown.

  • Sham-controlled intervention studies are lacking to determine the effects on AF symptoms, quality of life, and PROMS, accounting for the placebo effect that is associated with interventions.

The AF-CARE pathway in specific clinical settings

  • The optimal duration of triple therapy in patients with AF at high risk of recurrent coronary events after acute coronary syndrome is unclear.

  • The role of the coronary vessel involved and whether this should impact on the duration of combined OAC and antiplatelet treatment needs further study.

  • The role of antiplatelet therapy in patients with AF and peripheral artery disease on OAC is uncertain.

  • The use of DOACs in patients with congenital heart disease, particularly in patients with complex corrected congenital defects, is poorly studied.

  • Improved risk stratification for stroke in patients with AF and cancer, or with post-operative or trigger-induced AF is needed to inform on OAC treatment decisions.

Screening and prevention of AF

  • There are a lack of adequately powered randomized controlled studies on ischaemic stroke rate in patients screened for AF, both in the primary prevention setting and in secondary prevention (post-stroke), and its cost-effectiveness.

  • Population selection that might benefit the most from screening, the optimal duration of screening, and the burden of AF that might increase the risk for patients with screening-detected AF are uncertain.

  • Evaluation of strategies to support longer-term use of technologies for AF detection are awaited.

  • The role of photoplethysmography technology for AF screening in an effort to assess AF burden and reduce stroke is still unclear.

  • How new consumer devices and wearable technology can be used for diagnostic and monitoring purposes in routine clinical practice needs to be clarified.

13. ‘What to do’ and ‘What not to do’ messages from the guidelines

Table 17 lists all Class I and Class III recommendations from the text alongside their level of evidence.

Table 17

‘What to do’ and ‘what not to do’

Table 17

‘What to do’ and ‘what not to do’

14. Evidence tables

Evidence tables are available at European Heart Journal online.

15. Data availability statement

No new data were generated or analysed in support of this research.

16. Author information

Author/Task Force Member Affiliations: Michiel Rienstra, Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Karina V. Bunting, Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom, Cardiology Department, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom; Ruben Casado-Arroyo, Department of Cardiology, H.U.B.-Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium; Valeria Caso, Stroke Unit, Santa della Misericordia Hospital, Perugia, Italy; Harry J.G.M. Crijns, Cardiology Maastricht University Medical Centre, Maastricht, Netherlands, Cardiology Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands; Tom J. R. De Potter, Department of Cardiology, OLV Hospital, Aalst, Belgium; Jeremy Dwight (United Kingdom), ESC Patient Forum, Sophia Antipolis, France; Luigina Guasti, Department of Medicine and Surgery, University of Insubria, Varese, Italy, Division of Geriatrics and Clinical Gerontology, ASST-Settelaghi, Varese, Italy; Thorsten Hanke, Clinic For Cardiac Surgery, Asklepios Klinikum, Harburg, Hamburg, Germany; Tiny Jaarsma, Department of Cardiology, Linkoping University, Linkoping, Sweden, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands; Maddalena Lettino, Department for Cardiac, Thoracic and Vascular Diseases, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; Maja-Lisa Løchen, Deparment of Clincal Medicine UiT, The Arctic University of Norway, Tromsø, Norway, Department of Cardiology, University Hospital of North Norway, Tromsø, Norway; R. Thomas Lumbers, Institute of Health Informatics, University College London, London, United Kingdom, Saint Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom, University College Hospital, University College London Hospitals NHS Trust, London, United Kingdom; Bart Maesen, Department of Cardiothoracic Surgery, Maastricht University Medical Centre+, Maastricht, Netherlands, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands; Inge Mølgaard (Denmark), ESC Patient Forum, Sophia Antipolis, France; Giuseppe M.C. Rosano, Department of Human Sciences and Promotion of Quality of Life, Chair of Pharmacology, San Raffaele University of Rome, Rome, Italy, Cardiology, San Raffaele Cassino Hospital, Cassino, Italy, Cardiovascular Academic Group, St George's University Medical School, London, United Kingdom; Prashanthan Sanders, Centre for Heart Rhythm Disorders, University of Adelaide, Adelaide, Australia, Department of Cardiology, Royal Adelaide Hospital, Adelaide, Australia; Renate B. Schnabel, Cardiology University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany, German Center for Cardiovascular Research (DZHK) Partner site Hamburg/Kiel/Lübeck, Germany; Piotr Suwalski, Department of Cardiac Surgery and Transplantology, National Medical Institute of the Ministry of Interior and Administration, Centre of Postgraduate Medical Education, Warsaw, Poland; Emma Svennberg, Department of Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden, Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden; Juan Tamargo, Pharmacology and Toxicology School of Medicine, Universidad Complutense, Madrid, Spain; Otilia Tica, Department of Cardiology, Emergency County Clinical Hospital of Bihor, Oradea, Romania, Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Vassil Traykov, Department of Invasive Electrophysiology, Acibadem City Clinic Tokuda University Hospital, Sofia, Bulgaria; and Stylianos Tzeis, Cardiology Department, Mitera Hospital, Athens, Greece.

17. Appendix

ESC Scientific Document Group

Includes Document Reviewers and ESC National Cardiac Societies.

Document Reviewers: Nikolaos Dagres (CPG Review Co-ordinator) (Germany), Bianca Rocca (CPG Review Co-ordinator) (Italy), Syed Ahsan (United Kingdom), Pietro Ameri (Italy), Elena Arbelo (Spain), Axel Bauer (Austria), Michael A. Borger (Germany), Sergio Buccheri (Sweden), Barbara Casadei (United Kingdom), Ovidiu Chioncel (Romania), Dobromir Dobrev (Germany), Laurent Fauchier (France), Bruna Gigante (Sweden), Michael Glikson (Israel), Ziad Hijazi (Sweden), Gerhard Hindricks (Germany), Daniela Husser (Germany), Borja Ibanez (Spain), Stefan James (Sweden), Stefan Kaab (Germany), Paulus Kirchhof (Germany), Lars Køber (Denmark), Konstantinos C. Koskinas (Switzerland), Thomas Kumler (Denmark), Gregory Y.H. Lip (United Kingdom), John Mandrola (United States of America), Nikolaus Marx (Germany), John William Mcevoy (Ireland), Borislava Mihaylova (United Kingdom), Richard Mindham (United Kingdom), Denisa Muraru (Italy), Lis Neubeck (United Kingdom), Jens Cosedis Nielsen (Denmark), Jonas Oldgren (Sweden), Maurizio Paciaroni (Italy), Agnes A. Pasquet (Belgium), Eva Prescott (Denmark), Filip Rega (Belgium), Francisco Javier Rossello (Spain), Marcin Rucinski (Poland), Sacha P. Salzberg (Switzerland), Sam Schulman (Canada), Philipp Sommer (Germany), Jesper Hastrup Svendsen (Denmark), Jurrien M. ten Berg (Netherlands), Hugo Ten Cate (Netherlands), Ilonca Vaartjes (Netherlands), Christiaan Jm. Vrints (Belgium), Adam Witkowski (Poland), and Katja Zeppenfeld (Netherlands).

ESC National Cardiac Societies actively involved in the review process of the 2024 ESC Guidelines for the management of atrial fibrillation:

Albania: Albanian Society of Cardiology, Leonard Simoni; Algeria: Algerian Society of Cardiology, Brahim Kichou; Armenia: Armenian Cardiologists Association, Hamayak S. Sisakian; Austria: Austrian Society of Cardiology, Daniel Scherr; Belgium: Belgian Society of Cardiology, Frank Cools; Bosnia and Herzegovina: Association of Cardiologists of Bosnia and Herzegovina, Elnur Smajić; Bulgaria: Bulgarian Society of Cardiology, Tchavdar Shalganov; Croatia: Croatian Cardiac Society, Sime Manola; Cyprus: Cyprus Society of Cardiology, Panayiotis Avraamides; Czechia: Czech Society of Cardiology, Milos Taborsky; Denmark: Danish Society of Cardiology, Axel Brandes; Egypt: Egyptian Society of Cardiology, Ahmed M. El-Damaty; Estonia: Estonian Society of Cardiology, Priit Kampus; Finland: Finnish Cardiac Society, Pekka Raatikainen; France: French Society of Cardiology, Rodrigue Garcia; Georgia: Georgian Society of Cardiology, Kakhaber Etsadashvili; Germany: German Cardiac Society, Lars Eckardt; Greece: Hellenic Society of Cardiology, Eleftherios Kallergis; Hungary: Hungarian Society of Cardiology, László Gellér; Iceland: Icelandic Society of Cardiology, Kristján Guðmundsson; Ireland: Irish Cardiac Society, Jonathan Lyne; Israel: Israel Heart Society, Ibrahim Marai; Italy: Italian Federation of Cardiology, Furio Colivicchi; Kazakhstan: Association of Cardiologists of Kazakhstan, Ayan Suleimenovich Abdrakhmanov; Kosovo (Republic of): Kosovo Society of Cardiology, Ibadete Bytyci; Kyrgyzstan: Kyrgyz Society of Cardiology, Alina Kerimkulova; Latvia: Latvian Society of Cardiology, Kaspars Kupics; Lebanon: Lebanese Society of Cardiology, Marwan Refaat; Libya: Libyan Cardiac Society, Osama Abdulmajed Bheleel; Lithuania: Lithuanian Society of Cardiology, Jūratė Barysienė; Luxembourg: Luxembourg Society of Cardiology, Patrick Leitz; Malta: Maltese Cardiac Society, Mark A. Sammut; Moldova (Republic of): Moldavian Society of Cardiology, Aurel Grosu; Montenegro: Montenegro Society of Cardiology, Nikola Pavlovic; Morocco: Moroccan Society of Cardiology, Abdelhamid Moustaghfir; Netherlands: Netherlands Society of Cardiology, Sing-Chien Yap; North Macedonia: National Society of Cardiology of North Macedonia, Jane Taleski; Norway: Norwegian Society of Cardiology, Trine Fink; Poland: Polish Cardiac Society, Jaroslaw Kazmierczak; Portugal: Portuguese Society of Cardiology, Victor M. Sanfins; Romania: Romanian Society of Cardiology, Dragos Cozma; San Marino: San Marino Society of Cardiology, Marco Zavatta; Serbia: Cardiology Society of Serbia, Dragan V. Kovačević; Slovakia: Slovak Society of Cardiology, Peter Hlivak; Slovenia: Slovenian Society of Cardiology, Igor Zupan; Spain: Spanish Society of Cardiology, David Calvo; Sweden: Swedish Society of Cardiology, Anna Björkenheim; Switzerland: Swiss Society of Cardiology, Michael Kühne; Tunisia: Tunisian Society of Cardiology and Cardiovascular Surgery, Sana Ouali; Turkey: Turkish Society of Cardiology, Sabri Demircan; Ukraine: Ukrainian Association of Cardiology, Oleg S. Sychov; United Kingdom of Great Britain and Northern Ireland: British Cardiovascular Society, Andre Ng; and Uzbekistan: Association of Cardiologists of Uzbekistan, Husniddin Kuchkarov.

ESC Clinical Practice Guidelines (CPG) Committee: Eva Prescott (Chairperson) (Denmark), Stefan James (Co-Chairperson) (Sweden), Elena Arbelo (Spain), Colin Baigent (United Kingdom), Michael A. Borger (Germany), Sergio Buccheri (Sweden), Borja Ibanez (Spain), Lars Køber (Denmark), Konstantinos C. Koskinas (Switzerland), John William McEvoy (Ireland), Borislava Mihaylova (United Kingdom), Richard Mindham (United Kingdom), Lis Neubeck (United Kingdom), Jens Cosedis Nielsen (Denmark), Agnes A. Pasquet (Belgium), Amina Rakisheva (Kazakhstan), Bianca Rocca (Italy), Xavier Rossello (Spain), Ilonca Vaartjes (Netherlands), Christiaan Vrints (Belgium), Adam Witkowski (Poland), and Katja Zeppenfeld (Netherlands). Andrea Sarkozy* (Belgium) *Contributor either stepped down or was engaged in only a part of the review process.

18. References

1

Alam
 
M
,
Bandeali
 
SJ
,
Shahzad
 
SA
,
Lakkis
 
N
.
Real-life global survey evaluating patients with atrial fibrillation (REALISE-AF): results of an international observational registry
.
Expert Rev Cardiovasc Ther
 
2012
;
10
:
283
91
.

2

De With
 
RR
,
Erküner
 
Ö
,
Rienstra
 
M
,
Nguyen
 
BO
,
Körver
 
FWJ
,
Linz
 
D
, et al.  
Temporal patterns and short-term progression of paroxysmal atrial fibrillation: data from RACE V
.
Europace
 
2020
;
22
:
1162
72
.

3

Packer
 
DL
,
Mark
 
DB
,
Robb
 
RA
,
Monahan
 
KH
,
Bahnson
 
TD
,
Poole
 
JE
, et al.  
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
74
.

4

Marrouche
 
NF
,
Brachmann
 
J
,
Andresen
 
D
,
Siebels
 
J
,
Boersma
 
L
,
Jordaens
 
L
, et al.  
Catheter ablation for atrial fibrillation with heart failure
.
N Engl J Med
 
2018
;
378
:
417
27
.

5

Svendsen
 
JH
,
Diederichsen
 
SZ
,
Højberg
 
S
,
Krieger
 
DW
,
Graff
 
C
,
Kronborg
 
C
, et al.  
Implantable loop recorder detection of atrial fibrillation to prevent stroke (The LOOP Study): a randomised controlled trial
.
Lancet
 
2021
;
398
:
1507
16
.

6

Svennberg
 
E
,
Friberg
 
L
,
Frykman
 
V
,
Al-Khalili
 
F
,
Engdahl
 
J
,
Rosenqvist
 
M
.
Clinical outcomes in systematic screening for atrial fibrillation (STROKESTOP): a multicentre, parallel group, unmasked, randomised controlled trial
.
Lancet
 
2021
;
398
:
1498
506
.

7

Healey
 
JS
,
Connolly
 
SJ
,
Gold
 
MR
,
Israel
 
CW
,
Van Gelder
 
IC
,
Capucci
 
A
, et al.  
Subclinical atrial fibrillation and the risk of stroke
.
N Engl J Med
 
2012
;
366
:
120
9
.

8

McIntyre
 
WF
,
Healey
 
JS
,
Bhatnagar
 
AK
,
Wang
 
P
,
Gordon
 
JA
,
Baranchuk
 
A
, et al.  
Vernakalant for cardioversion of recent-onset atrial fibrillation: a systematic review and meta-analysis
.
Europace
 
2019
;
21
:
1159
66
.

9

Bager
 
JE
,
Martín
 
A
,
Carbajosa Dalmau
 
J
,
Simon
 
A
,
Merino
 
JL
,
Ritz
 
B
, et al.  
Vernakalant for cardioversion of recent-onset atrial fibrillation in the emergency department: the SPECTRUM study
.
Cardiology
 
2022
;
147
:
566
77
.

10

Pluymaekers
 
N
,
Dudink
 
E
,
Luermans
 
J
,
Meeder
 
JG
,
Lenderink
 
T
,
Widdershoven
 
J
, et al.  
Early or delayed cardioversion in recent-onset atrial fibrillation
.
N Engl J Med
 
2019
;
380
:
1499
508
.

11

Lubitz
 
SA
,
Yin
 
X
,
Rienstra
 
M
,
Schnabel
 
RB
,
Walkey
 
AJ
,
Magnani
 
JW
, et al.  
Long-term outcomes of secondary atrial fibrillation in the community: the Framingham Heart Study
.
Circulation
 
2015
;
131
:
1648
55
.

12

Wang
 
EY
,
Hulme
 
OL
,
Khurshid
 
S
,
Weng
 
LC
,
Choi
 
SH
,
Walkey
 
AJ
, et al.  
Initial precipitants and recurrence of atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e007716
.

13

Corica
 
B
,
Romiti
 
GF
,
Basili
 
S
,
Proietti
 
M
.
Prevalence of new-onset atrial fibrillation and associated outcomes in patients with sepsis: a systematic review and meta-analysis
.
J Pers Med
 
2022
;
12
:
547
.

14

Bedford
 
JP
,
Ferrando-Vivas
 
P
,
Redfern
 
O
,
Rajappan
 
K
,
Harrison
 
DA
,
Watkinson
 
PJ
, et al.  
New-onset atrial fibrillation in intensive care: epidemiology and outcomes
.
Eur Heart J Acute Cardiovasc Care
 
2022
;
11
:
620
8
.

15

Wazni
 
OM
,
Marrouche
 
NF
,
Martin
 
DO
,
Verma
 
A
,
Bhargava
 
M
,
Saliba
 
W
, et al.  
Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of symptomatic atrial fibrillation: a randomized trial
.
JAMA
 
2005
;
293
:
2634
40
.

16

Andrade
 
JG
,
Wells
 
GA
,
Deyell
 
MW
,
Bennett
 
M
,
Essebag
 
V
,
Champagne
 
J
, et al.  
Cryoablation or drug therapy for initial treatment of atrial fibrillation
.
N Engl J Med
 
2021
;
384
:
305
15
.

17

Kirchhof
 
P
,
Camm
 
AJ
,
Goette
 
A
,
Brandes
 
A
,
Eckardt
 
L
,
Elvan
 
A
, et al.  
Early rhythm-control therapy in patients with atrial fibrillation
.
N Engl J Med
 
2020
;
383
:
1305
16
.

18

Coats
 
AJS
,
Heymans
 
S
,
Farmakis
 
D
,
Anker
 
SD
,
Backs
 
J
,
Bauersachs
 
J
, et al.  
Atrial disease and heart failure: the common soil hypothesis proposed by the heart failure association of the European Society of Cardiology
.
Eur Heart J
 
2022
:
43
:
863
7
.

19

Schnabel
 
RB
,
Marinelli
 
EA
,
Arbelo
 
E
,
Boriani
 
G
,
Boveda
 
S
,
Buckley
 
CM
, et al.  
Early diagnosis and better rhythm management to improve outcomes in patients with atrial fibrillation: the 8th AFNET/EHRA consensus conference
.
Europace
 
2023
;
25
:
6
27
.

20

Goette
 
A
,
Kalman
 
JM
,
Aguinaga
 
L
,
Akar
 
J
,
Cabrera
 
JA
,
Chen
 
SA
, et al.  
EHRA/HRS/APHRS/SOLAECE expert consensus on atrial cardiomyopathies: definition, characterization, and clinical implication
.
Europace
 
2016
;
18
:
1455
90
.

21

Sagris
 
D
,
Georgiopoulos
 
G
,
Pateras
 
K
,
Perlepe
 
K
,
Korompoki
 
E
,
Milionis
 
H
, et al.  
Atrial high-rate episode duration thresholds and thromboembolic risk: a systematic review and meta-analysis
.
J Am Heart Assoc
 
2021
;
10
:
e022487
.

22

Kaufman
 
ES
,
Israel
 
CW
,
Nair
 
GM
,
Armaganijan
 
L
,
Divakaramenon
 
S
,
Mairesse
 
GH
, et al.  
Positive predictive value of device-detected atrial high-rate episodes at different rates and durations: an analysis from ASSERT
.
Heart Rhythm
 
2012
;
9
:
1241
6
.

23

Miyazawa
 
K
,
Pastori
 
D
,
Martin
 
DT
,
Choucair
 
WK
,
Halperin
 
JL
,
Lip
 
GYH
.
Characteristics of patients with atrial high rate episodes detected by implanted defibrillator and resynchronization devices
.
Europace
 
2022
;
24
:
375
83
.

24

Vitolo
 
M
,
Imberti
 
JF
,
Maisano
 
A
,
Albini
 
A
,
Bonini
 
N
,
Valenti
 
AC
, et al.  
Device-detected atrial high rate episodes and the risk of stroke/thromboembolism and atrial fibrillation incidence: a systematic review and meta-analysis
.
Eur J Intern Med
 
2021
;
92
:
100
6
.

25

Connolly
 
SJ
,
Ezekowitz
 
MD
,
Yusuf
 
S
,
Eikelboom
 
J
,
Oldgren
 
J
,
Parekh
 
A
, et al.  
Dabigatran versus warfarin in patients with atrial fibrillation
.
N Engl J Med
 
2009
;
361
:
1139
51
.

26

Giugliano
 
RP
,
Ruff
 
CT
,
Braunwald
 
E
,
Murphy
 
SA
,
Wiviott
 
SD
,
Halperin
 
JL
, et al.  
Edoxaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
 
2013
;
369
:
2093
104
.

27

Granger
 
CB
,
Alexander
 
JH
,
McMurray
 
JJ
,
Lopes
 
RD
,
Hylek
 
EM
,
Hanna
 
M
, et al.  
Apixaban versus warfarin in patients with atrial fibrillation
.
N Engl J Med
 
2011
;
365
:
981
92
.

28

Patel
 
MR
,
Mahaffey
 
KW
,
Garg
 
J
,
Pan
 
G
,
Singer
 
DE
,
Hacke
 
W
, et al.  
Rivaroxaban versus warfarin in nonvalvular atrial fibrillation
.
N Engl J Med
 
2011
;
365
:
883
91
.

29

Stroke prevention in atrial fibrillation study. Final results
.
Circulation
 
1991
;
84
:
527
39
.

30

Mannina
 
C
,
Jin
 
Z
,
Matsumoto
 
K
,
Ito
 
K
,
Biviano
 
A
,
Elkind
 
MSV
, et al.  
Frequency of cardiac arrhythmias in older adults: findings from the subclinical atrial fibrillation and risk of ischemic stroke (SAFARIS) study
.
Int J Cardiol
 
2021
;
337
:
64
70
.

31

Schnabel
 
RB
,
Pecen
 
L
,
Ojeda
 
FM
,
Lucerna
 
M
,
Rzayeva
 
N
,
Blankenberg
 
S
, et al.  
Gender differences in clinical presentation and 1-year outcomes in atrial fibrillation
.
Heart
 
2017
;
103
:
1024
30
.

32

Simantirakis
 
EN
,
Papakonstantinou
 
PE
,
Chlouverakis
 
GI
,
Kanoupakis
 
EM
,
Mavrakis
 
HE
,
Kallergis
 
EM
, et al.  
Asymptomatic versus symptomatic episodes in patients with paroxysmal atrial fibrillation via long-term monitoring with implantable loop recorders
.
Int J Cardiol
 
2017
;
231
:
125
30
.

33

Verma
 
A
,
Champagne
 
J
,
Sapp
 
J
,
Essebag
 
V
,
Novak
 
P
,
Skanes
 
A
, et al.  
Discerning the incidence of symptomatic and asymptomatic episodes of atrial fibrillation before and after catheter ablation (DISCERN AF): a prospective, multicenter study
.
JAMA Intern Med
 
2013
;
173
:
149
56
.

34

Sgreccia
 
D
,
Manicardi
 
M
,
Malavasi
 
VL
,
Vitolo
 
M
,
Valenti
 
AC
,
Proietti
 
M
, et al.  
Comparing outcomes in asymptomatic and symptomatic atrial fibrillation: a systematic review and meta-analysis of 81,462 patients
.
J Clin Med
 
2021
;
10
:
3979
.

35

Holmes
 
DN
,
Piccini
 
JP
,
Allen
 
LA
,
Fonarow
 
GC
,
Gersh
 
BJ
,
Kowey
 
PR
, et al.  
Defining clinically important difference in the atrial fibrillation effect on quality-of-life score
.
Circ Cardiovasc Qual Outcomes
 
2019
;
12
:
e005358
.

36

Jones
 
J
,
Stanbury
 
M
,
Haynes
 
S
,
Bunting
 
KV
,
Lobban
 
T
,
Camm
 
AJ
, et al.  
Importance and assessment of quality of life in symptomatic permanent atrial fibrillation: patient focus groups from the RATE-AF trial
.
Cardiology
 
2020
;
145
:
666
75
.

37

Abu
 
HO
,
Wang
 
W
,
Otabil
 
EM
,
Saczynski
 
JS
,
Mehawej
 
J
,
Mishra
 
A
, et al.  
Perception of atrial fibrillation symptoms: impact on quality of life and treatment in older adults
.
J Am Geriatr Soc
 
2022
;
70
:
2805
17
.

38

Rienstra
 
M
,
Vermond
 
RA
,
Crijns
 
HJ
,
Tijssen
 
JG
,
Van Gelder
 
IC
;
RACE Investigators
.
Asymptomatic persistent atrial fibrillation and outcome: results of the RACE study
.
Heart Rhythm
 
2014
;
11
:
939
45
.

39

Rienstra
 
M
,
Hobbelt
 
AH
,
Alings
 
M
,
Tijssen
 
JGP
,
Smit
 
MD
,
Brugemann
 
J
, et al.  
Targeted therapy of underlying conditions improves sinus rhythm maintenance in patients with persistent atrial fibrillation: results of the RACE 3 trial
.
Eur Heart J
 
2018
;
39
:
2987
96
.

40

Mulder
 
BA
,
Van Veldhuisen
 
DJ
,
Crijns
 
HJ
,
Tijssen
 
JG
,
Hillege
 
HL
,
Alings
 
M
, et al.  
Digoxin in patients with permanent atrial fibrillation: data from the RACE II study
.
Heart Rhythm
 
2014
;
11
:
1543
50
.

41

Kloosterman
 
M
,
Crijns
 
H
,
Mulder
 
BA
,
Groenveld
 
HF
,
Van Veldhuisen
 
DJ
,
Rienstra
 
M
, et al.  
Sex-related differences in risk factors, outcome, and quality of life in patients with permanent atrial fibrillation: results from the RACE II study
.
Europace
 
2020
;
22
:
1619
27
.

42

Park
 
YJ
,
Park
 
JW
,
Yu
 
HT
,
Kim
 
TH
,
Uhm
 
JS
,
Joung
 
B
, et al.  
Sex difference in atrial fibrillation recurrence after catheter ablation and antiarrhythmic drugs
.
Heart
 
2023
;
109
:
519
26
.

43

Kupper
 
N
,
van den Broek
 
KC
,
Widdershoven
 
J
,
Denollet
 
J
.
Subjectively reported symptoms in patients with persistent atrial fibrillation and emotional distress
.
Front Psychol
 
2013
;
4
:
192
.

44

Schnabel
 
RB
,
Michal
 
M
,
Wilde
 
S
,
Wiltink
 
J
,
Wild
 
PS
,
Sinning
 
CR
, et al.  
Depression in atrial fibrillation in the general population
.
PLoS One
 
2013
;
8
:
e79109
.

45

Gleason
 
KT
,
Dennison Himmelfarb
 
CR
,
Ford
 
DE
,
Lehmann
 
H
,
Samuel
 
L
,
Han
 
HR
, et al.  
Association of sex, age and education level with patient reported outcomes in atrial fibrillation
.
BMC Cardiovasc Disord
 
2019
;
19
:
85
.

46

Schnabel
 
RB
,
Pecen
 
L
,
Rzayeva
 
N
,
Lucerna
 
M
,
Purmah
 
Y
,
Ojeda
 
FM
, et al.  
Symptom burden of atrial fibrillation and its relation to interventions and outcome in Europe
.
J Am Heart Assoc
 
2018
;
7
:
e007559
.

47

Wynn
 
GJ
,
Todd
 
DM
,
Webber
 
M
,
Bonnett
 
L
,
McShane
 
J
,
Kirchhof
 
P
, et al.  
The European Heart Rhythm Association symptom classification for atrial fibrillation: validation and improvement through a simple modification
.
Europace
 
2014
;
16
:
965
72
.

48

Kotecha
 
D
,
Bunting
 
KV
,
Gill
 
SK
,
Mehta
 
S
,
Stanbury
 
M
,
Jones
 
JC
, et al.  
Effect of digoxin vs bisoprolol for heart rate control in atrial fibrillation on patient-reported quality of life: the RATE-AF randomized clinical trial
.
JAMA
 
2020
;
324
:
2497
508
.

49

Kotecha
 
D
,
Ahmed
 
A
,
Calvert
 
M
,
Lencioni
 
M
,
Terwee
 
CB
,
Lane
 
DA
.
Patient-reported outcomes for quality of life assessment in atrial fibrillation: a systematic review of measurement properties
.
PLoS One
 
2016
;
11
:
e0165790
.

50

Mantovan
 
R
,
Macle
 
L
,
De Martino
 
G
,
Chen
 
J
,
Morillo
 
CA
,
Novak
 
P
, et al.  
Relationship of quality of life with procedural success of atrial fibrillation (AF) ablation and postablation AF burden: substudy of the STAR AF randomized trial
.
Can J Cardiol
 
2013
;
29
:
1211
7
.

51

Samuel
 
M
,
Khairy
 
P
,
Champagne
 
J
,
Deyell
 
MW
,
Macle
 
L
,
Leong-Sit
 
P
, et al.  
Association of atrial fibrillation burden with health-related quality of life after atrial fibrillation ablation: substudy of the cryoballoon vs contact-force atrial fibrillation ablation (CIRCA-DOSE) randomized clinical trial
.
JAMA Cardiol
 
2021
;
6
:
1324
8
.

52

Sandhu
 
RK
,
Smigorowsky
 
M
,
Lockwood
 
E
,
Savu
 
A
,
Kaul
 
P
,
McAlister
 
FA
.
Impact of electrical cardioversion on quality of life for the treatment of atrial fibrillation
.
Can J Cardiol
 
2017
;
33
:
450
5
.

53

Terricabras
 
M
,
Mantovan
 
R
,
Jiang
 
CY
,
Betts
 
TR
,
Chen
 
J
,
Deisenhofer
 
I
, et al.  
Association between quality of life and procedural outcome after catheter ablation for atrial fibrillation: a secondary analysis of a randomized clinical trial
.
JAMA Netw Open
 
2020
;
3
:
e2025473
.

54

Zenger
 
B
,
Zhang
 
M
,
Lyons
 
A
,
Bunch
 
TJ
,
Fang
 
JC
,
Freedman
 
RA
, et al.  
Patient-reported outcomes and subsequent management in atrial fibrillation clinical practice: results from the Utah mEVAL AF program
.
J Cardiovasc Electrophysiol
 
2020
;
31
:
3187
95
.

55

Arbelo
 
E
,
Aktaa
 
S
,
Bollmann
 
A
,
D’Avila
 
A
,
Drossart
 
I
,
Dwight
 
J
, et al.  
Quality indicators for the care and outcomes of adults with atrial fibrillation
.
Europace
 
2021
;
23
:
494
5
.

56

Kvist
 
LM
,
Vinter
 
N
,
Urbonaviciene
 
G
,
Lindholt
 
JS
,
Diederichsen
 
ACP
,
Frost
 
L
.
Diagnostic accuracies of screening for atrial fibrillation by cardiac nurses versus radiographers
.
Open Heart
 
2019
;
6
:
e000942
.

57

Hijazi
 
Z
,
Oldgren
 
J
,
Siegbahn
 
A
,
Granger
 
CB
,
Wallentin
 
L
.
Biomarkers in atrial fibrillation: a clinical review
.
Eur Heart J
 
2013
;
34
:
1475
80
.

58

Berg
 
DD
,
Ruff
 
CT
,
Morrow
 
DA
.
Biomarkers for risk assessment in atrial fibrillation
.
Clin Chem
 
2021
;
67
:
87
95
.

59

Tops
 
LF
,
Schalij
 
MJ
,
Bax
 
JJ
.
Imaging and atrial fibrillation: the role of multimodality imaging in patient evaluation and management of atrial fibrillation
.
Eur Heart J
 
2010
;
31
:
542
51
.

60

Obeng-Gyimah
 
E
,
Nazarian
 
S
.
Advancements in imaging for atrial fibrillation ablation: is there a potential to improve procedural outcomes?
 
J Innov Card Rhythm Manag
 
2020
;
11
:
4172
8
.

61

Romero
 
J
,
Husain
 
SA
,
Kelesidis
 
I
,
Sanz
 
J
,
Medina
 
HM
,
Garcia
 
MJ
.
Detection of left atrial appendage thrombus by cardiac computed tomography in patients with atrial fibrillation: a meta-analysis
.
Circ Cardiovasc Imaging
 
2013
;
6
:
185
94
.

62

Bisbal
 
F
,
Benito
 
E
,
Teis
 
A
,
Alarcón
 
F
,
Sarrias
 
A
,
Caixal
 
G
, et al.  
Magnetic resonance imaging-guided fibrosis ablation for the treatment of atrial fibrillation: the ALICIA trial
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e008707
.

63

Khurram
 
IM
,
Habibi
 
M
,
Gucuk Ipek
 
E
,
Chrispin
 
J
,
Yang
 
E
,
Fukumoto
 
K
, et al.  
Left atrial LGE and arrhythmia recurrence following pulmonary vein isolation for paroxysmal and persistent AF
.
JACC Cardiovasc Imaging
 
2016
;
9
:
142
8
.

64

Marrouche
 
NF
,
Wilber
 
D
,
Hindricks
 
G
,
Jais
 
P
,
Akoum
 
N
,
Marchlinski
 
F
, et al.  
Association of atrial tissue fibrosis identified by delayed enhancement MRI and atrial fibrillation catheter ablation: the DECAAF study
.
JAMA
 
2014
;
311
:
498
506
.

65

Roney
 
CH
,
Sillett
 
C
,
Whitaker
 
J
,
Lemus
 
JAS
,
Sim
 
I
,
Kotadia
 
I
, et al.  
Applications of multimodality imaging for left atrial catheter ablation
.
Eur Heart J Cardiovasc Imaging
 
2021
;
23
:
31
41
.

66

Potter
 
A
,
Augustine
 
DX
,
Ingram
 
TE
.
Referring for echocardiography: when not to test
.
Br J Gen Pract
 
2021
;
71
:
333
4
.

67

Troughton
 
RW
,
Asher
 
CR
,
Klein
 
AL
.
The role of echocardiography in atrial fibrillation and cardioversion
.
Heart
 
2003
;
89
:
1447
54
.

68

Odutayo
 
A
,
Wong
 
CX
,
Hsiao
 
AJ
,
Hopewell
 
S
,
Altman
 
DG
,
Emdin
 
CA
.
Atrial fibrillation and risks of cardiovascular disease, renal disease, and death: systematic review and meta-analysis
.
BMJ
 
2016
;
354
:
i4482
.

69

Ruddox
 
V
,
Sandven
 
I
,
Munkhaugen
 
J
,
Skattebu
 
J
,
Edvardsen
 
T
,
Otterstad
 
JE
.
Atrial fibrillation and the risk for myocardial infarction, all-cause mortality and heart failure: a systematic review and meta-analysis
.
Eur J Prev Cardiol
 
2017
;
24
:
1555
66
.

70

Bassand
 
JP
,
Accetta
 
G
,
Al Mahmeed
 
W
,
Corbalan
 
R
,
Eikelboom
 
J
,
Fitzmaurice
 
DA
, et al.  
Risk factors for death, stroke, and bleeding in 28,628 patients from the GARFIELD-AF registry: rationale for comprehensive management of atrial fibrillation
.
PLoS One
 
2018
;
13
:
e0191592
.

71

Bassand
 
JP
,
Accetta
 
G
,
Camm
 
AJ
,
Cools
 
F
,
Fitzmaurice
 
DA
,
Fox
 
KA
, et al.  
Two-year outcomes of patients with newly diagnosed atrial fibrillation: results from GARFIELD-AF
.
Eur Heart J
 
2016
;
37
:
2882
9
.

72

Hornestam
 
B
,
Adiels
 
M
,
Wai Giang
 
K
,
Hansson
 
PO
,
Björck
 
L
,
Rosengren
 
A
.
Atrial fibrillation and risk of venous thromboembolism: a Swedish nationwide registry study
.
Europace
 
2021
;
23
:
1913
21
.

73

Lutsey
 
PL
,
Norby
 
FL
,
Alonso
 
A
,
Cushman
 
M
,
Chen
 
LY
,
Michos
 
ED
, et al.  
Atrial fibrillation and venous thromboembolism: evidence of bidirectionality in the atherosclerosis risk in communities study
.
J Thromb Haemost
 
2018
;
16
:
670
9
.

74

Koh
 
YH
,
Lew
 
LZW
,
Franke
 
KB
,
Elliott
 
AD
,
Lau
 
DH
,
Thiyagarajah
 
A
, et al.  
Predictive role of atrial fibrillation in cognitive decline: a systematic review and meta-analysis of 2.8 million individuals
.
Europace
 
2022
;
24
:
1229
39
.

75

Papanastasiou
 
CA
,
Theochari
 
CA
,
Zareifopoulos
 
N
,
Arfaras-Melainis
 
A
,
Giannakoulas
 
G
,
Karamitsos
 
TD
, et al.  
Atrial fibrillation is associated with cognitive impairment, all-cause dementia, vascular dementia, and Alzheimer’s disease: a systematic review and meta-analysis
.
J Gen Intern Med
 
2021
;
36
:
3122
35
.

76

Giannone
 
ME
,
Filippini
 
T
,
Whelton
 
PK
,
Chiari
 
A
,
Vitolo
 
M
,
Boriani
 
G
, et al.  
Atrial fibrillation and the risk of early-onset dementia: a systematic review and meta-analysis
.
J Am Heart Assoc
 
2022
;
11
:
e025653
.

77

Zuin
 
M
,
Roncon
 
L
,
Passaro
 
A
,
Bosi
 
C
,
Cervellati
 
C
,
Zuliani
 
G
.
Risk of dementia in patients with atrial fibrillation: short versus long follow-up. A systematic review and meta-analysis
.
Int J Geriatr Psychiatry
 
2021
;
36
:
1488
500
.

78

Mobley
 
AR
,
Subramanian
 
A
,
Champsi
 
A
,
Wang
 
X
,
Myles
 
P
,
McGreavy
 
P
, et al. Thromboembolic events and vascular dementia in patients with atrial fibrillation and low apparent stroke risk. Nat Med 2024. .

79

Wijtvliet
 
E
,
Tieleman
 
RG
,
van Gelder
 
IC
,
Pluymaekers
 
N
,
Rienstra
 
M
,
Folkeringa
 
RJ
, et al.  
Nurse-led vs. usual-care for atrial fibrillation
.
Eur Heart J
 
2020
;
41
:
634
41
.

80

Wong
 
CX
,
Brooks
 
AG
,
Lau
 
DH
,
Leong
 
DP
,
Sun
 
MT
,
Sullivan
 
T
, et al.  
Factors associated with the epidemic of hospitalizations due to atrial fibrillation
.
Am J Cardiol
 
2012
;
110
:
1496
9
.

81

Dai
 
H
,
Zhang
 
Q
,
Much
 
AA
,
Maor
 
E
,
Segev
 
A
,
Beinart
 
R
, et al.  
Global, regional, and national prevalence, incidence, mortality, and risk factors for atrial fibrillation, 1990–2017: results from the Global Burden of Disease Study 2017
.
Eur Heart J Qual Care Clin Outcomes
 
2021
;
7
:
574
82
.

82

Țica
 
O
,
Țica
 
O
,
Bunting
 
KV
,
deBono
 
J
,
Gkoutos
 
GV
,
Popescu
 
MI
, et al.  
Post-mortem examination of high mortality in patients with heart failure and atrial fibrillation
.
BMC Med
 
2022
;
20
:
331
.

83

Bassand
 
JP
,
Virdone
 
S
,
Badoz
 
M
,
Verheugt
 
FWA
,
Camm
 
AJ
,
Cools
 
F
, et al.  
Bleeding and related mortality with NOACs and VKAs in newly diagnosed atrial fibrillation: results from the GARFIELD-AF registry
.
Blood Adv
 
2021
;
5
:
1081
91
.

84

Pokorney
 
SD
,
Piccini
 
JP
,
Stevens
 
SR
,
Patel
 
MR
,
Pieper
 
KS
,
Halperin
 
JL
, et al.  
Cause of death and predictors of all-cause mortality in anticoagulated patients with nonvalvular atrial fibrillation: data from ROCKET AF
.
J Am Heart Assoc
 
2016
;
5
:
e002197
.

85

Granada
 
J
,
Uribe
 
W
,
Chyou
 
PH
,
Maassen
 
K
,
Vierkant
 
R
,
Smith
 
PN
, et al.  
Incidence and predictors of atrial flutter in the general population
.
J Am Coll Cardiol
 
2000
;
36
:
2242
6
.

86

Vadmann
 
H
,
Nielsen
 
PB
,
Hjortshoj
 
SP
,
Riahi
 
S
,
Rasmussen
 
LH
,
Lip
 
GY
, et al.  
Atrial flutter and thromboembolic risk: a systematic review
.
Heart
 
2015
;
101
:
1446
55
.

87

Lelorier
 
P
,
Humphries
 
KH
,
Krahn
 
A
,
Connolly
 
SJ
,
Talajic
 
M
,
Green
 
M
, et al.  
Prognostic differences between atrial fibrillation and atrial flutter
.
Am J Cardiol
 
2004
;
93
:
647
9
.

88

Biblo
 
LA
,
Yuan
 
Z
,
Quan
 
KJ
,
Mackall
 
JA
,
Rimm
 
AA
.
Risk of stroke in patients with atrial flutter
.
Am J Cardiol
 
2001
;
87
:
346
9
, A9.

89

Corrado
 
G
,
Sgalambro
 
A
,
Mantero
 
A
,
Gentile
 
F
,
Gasparini
 
M
,
Bufalino
 
R
, et al.  
Thromboembolic risk in atrial flutter. The FLASIEC (FLutter Atriale Società Italiana di Ecografia Cardiovascolare) multicentre study
.
Eur Heart J
 
2001
;
22
:
1042
51
.

90

Lin
 
YS
,
Chen
 
TH
,
Chi
 
CC
,
Lin
 
MS
,
Tung
 
TH
,
Liu
 
CH
, et al.  
Different implications of heart failure, ischemic stroke, and mortality between nonvalvular atrial fibrillation and atrial flutter–a view from a national cohort study
.
J Am Heart Assoc
 
2017
;
6
:
e006406
.

91

Giehm-Reese
 
M
,
Johansen
 
MN
,
Kronborg
 
MB
,
Jensen
 
HK
,
Gerdes
 
C
,
Kristensen
 
J
, et al.  
Discontinuation of oral anticoagulation and risk of stroke and death after ablation for typical atrial flutter: a nation-wide Danish cohort study
.
Int J Cardiol
 
2021
;
333
:
110
6
.

92

Gallagher
 
C
,
Rowett
 
D
,
Nyfort-Hansen
 
K
,
Simmons
 
S
,
Brooks
 
AG
,
Moss
 
JR
, et al.  
Patient-centered educational resources for atrial fibrillation
.
JACC Clin Electrophysiol
 
2019
;
5
:
1101
14
.

93

Chung
 
MK
,
Fagerlin
 
A
,
Wang
 
PJ
,
Ajayi
 
TB
,
Allen
 
LA
,
Baykaner
 
T
, et al.  
Shared decision making in cardiac electrophysiology procedures and arrhythmia management
.
Circ Arrhythm Electrophysiol
 
2021
;
14
:
e007958
.

94

Wang
 
PJ
,
Lu
 
Y
,
Mahaffey
 
KW
,
Lin
 
A
,
Morin
 
DP
,
Sears
 
SF
, et al.  
A randomized clinical trial to evaluate an atrial fibrillation stroke prevention shared decision-making pathway
.
J Am Heart Assoc
 
2022
;
12
:
e028562
.

95

Seaburg
 
L
,
Hess
 
EP
,
Coylewright
 
M
,
Ting
 
HH
,
McLeod
 
CJ
,
Montori
 
VM
.
Shared decision making in atrial fibrillation: where we are and where we should be going
.
Circulation
 
2014
;
129
:
704
10
.

96

Zhang
 
J
,
Lenarczyk
 
R
,
Marin
 
F
,
Malaczynska-Rajpold
 
K
,
Kosiuk
 
J
,
Doehner
 
W
, et al.  
The interpretation of CHA2DS2-VASc score components in clinical practice: a joint survey by the European Heart Rhythm Association (EHRA) scientific initiatives committee, the EHRA young electrophysiologists, the Association of Cardiovascular Nursing and Allied Professionals, and the European Society of Cardiology council on stroke
.
Europace
 
2021
;
23
:
314
22
.

97

Omoush
 
A
,
Aloush
 
S
,
Albashtawy
 
M
,
Rayan
 
A
,
Alkhawaldeh
 
A
,
Eshah
 
N
, et al.  
Nurses knowledge of anticoagulation therapy for atrial fibrillation patients: effectiveness of an educational course
.
Nurs Forum
 
2022
;
57
:
825
32
.

98

Heidbuchel
 
H
,
Dagres
 
N
,
Antz
 
M
,
Kuck
 
KH
,
Lazure
 
P
,
Murray
 
S
, et al.  
Major knowledge gaps and system barriers to guideline implementation among European physicians treating patients with atrial fibrillation: a European Society of Cardiology international educational needs assessment
.
Europace
 
2018
;
20
:
1919
28
.

99

Bunting
 
KV
,
Van Gelder
 
IC
,
Kotecha
 
D
.
STEEER-AF: a cluster-randomized education trial from the ESC
.
Eur Heart J
 
2020
;
41
:
1952
4
.

100

Tanner
 
FC
,
Brooks
 
N
,
Fox
 
KF
,
Gonçalves
 
L
,
Kearney
 
P
,
Michalis
 
L
, et al.  
ESC core curriculum for the cardiologist
.
Eur Heart J
 
2020
;
41
:
3605
92
.

101

Astin
 
F
,
Carroll
 
D
,
De Geest
 
S
,
Fernandez-Oliver
 
AL
,
Holt
 
J
,
Hinterbuchner
 
L
, et al.  
A core curriculum for the continuing professional development of nurses working in cardiovascular settings: developed by the education committee of the Council on Cardiovascular Nursing and Allied Professions (CCNAP) on behalf of the European Society of Cardiology
.
Eur J Cardiovasc Nurs
 
2015
;
14
:
S1
17
.

102

Sterlinski
 
M
,
Bunting
 
KV
,
Boriani
 
G
,
Boveda
 
S
,
Guasch
 
E
,
Mont
 
L
, et al. STEEER-AF Trial Team. Design and deployment of the STEEER-AF trial to evaluate and improve guideline adherence: a cluster-randomised trial by the European Society of Cardiology and European Heart Rhythm Association. Europace 2024:euae178.

103

Vinereanu
 
D
,
Lopes
 
RD
,
Bahit
 
MC
,
Xavier
 
D
,
Jiang
 
J
,
Al-Khalidi
 
HR
, et al.  
A multifaceted intervention to improve treatment with oral anticoagulants in atrial fibrillation (IMPACT-AF): an international, cluster-randomised trial
.
Lancet
 
2017
;
390
:
1737
46
.

104

Franchi
 
C
,
Antoniazzi
 
S
,
Ardoino
 
I
,
Proietti
 
M
,
Marcucci
 
M
,
Santalucia
 
P
, et al.  
Simulation-based education for physicians to increase oral anticoagulants in hospitalized elderly patients with atrial fibrillation
.
Am J Med
 
2019
;
132
:
e634
47
.

105

Baicus
 
C
,
Delcea
 
C
,
Dima
 
A
,
Oprisan
 
E
,
Jurcut
 
C
,
Dan
 
GA
.
Influence of decision aids on oral anticoagulant prescribing among physicians: a randomised trial
.
Eur J Clin Invest
 
2017
;
47
:
649
58
.

106

Ono
 
F
,
Akiyama
 
S
,
Suzuki
 
A
,
Ikeda
 
Y
,
Takahashi
 
A
,
Matsuoka
 
H
, et al.  
Impact of care coordination on oral anticoagulant therapy among patients with atrial fibrillation in routine clinical practice in Japan: a prospective, observational study
.
BMC Cardiovasc Disord
 
2019
;
19
:
235
.

107

Ferguson
 
C
,
Hickman
 
LD
,
Phillips
 
J
,
Newton
 
PJ
,
Inglis
 
SC
,
Lam
 
L
, et al.  
An mHealth intervention to improve nurses’ atrial fibrillation and anticoagulation knowledge and practice: the EVICOAG study
.
Eur J Cardiovasc Nurs
 
2019
;
18
:
7
15
.

108

Lip
 
GY
,
Laroche
 
C
,
Popescu
 
MI
,
Rasmussen
 
LH
,
Vitali-Serdoz
 
L
,
Dan
 
GA
, et al.  
Improved outcomes with European Society of Cardiology guideline-adherent antithrombotic treatment in high-risk patients with atrial fibrillation: a report from the EORP-AF general pilot registry
.
Europace
 
2015
;
17
:
1777
86
.

109

Linde
 
C
,
Bongiorni
 
MG
,
Birgersdotter-Green
 
U
,
Curtis
 
AB
,
Deisenhofer
 
I
,
Furokawa
 
T
, et al.  
Sex differences in cardiac arrhythmia: a consensus document of the European Heart Rhythm Association, endorsed by the Heart Rhythm Society and Asia Pacific Heart Rhythm Society
.
Europace
 
2018
;
20
:
1565
1565ao
.

110

Camm
 
AJ
,
Accetta
 
G
,
Al Mahmeed
 
W
,
Ambrosio
 
G
,
Goldhaber
 
SZ
,
Haas
 
S
, et al.  
Impact of gender on event rates at 1 year in patients with newly diagnosed non-valvular atrial fibrillation: contemporary perspective from the GARFIELD-AF registry
.
BMJ Open
 
2017
;
7
:
e014579
.

111

Emdin
 
CA
,
Wong
 
CX
,
Hsiao
 
AJ
,
Altman
 
DG
,
Peters
 
SA
,
Woodward
 
M
, et al.  
Atrial fibrillation as risk factor for cardiovascular disease and death in women compared with men: systematic review and meta-analysis of cohort studies
.
BMJ
 
2016
;
532
:
h7013
.

112

Tomasdottir
 
M
,
Friberg
 
L
,
Hijazi
 
Z
,
Lindback
 
J
,
Oldgren
 
J
.
Risk of ischemic stroke and utility of CHA2 DS2 -VASc score in women and men with atrial fibrillation
.
Clin Cardiol
 
2019
;
42
:
1003
9
.

113

Kloosterman
 
M
,
Chua
 
W
,
Fabritz
 
L
,
Al-Khalidi
 
HR
,
Schotten
 
U
,
Nielsen
 
JC
, et al.  
Sex differences in catheter ablation of atrial fibrillation: results from AXAFA-AFNET 5
.
Europace
 
2020
;
22
:
1026
35
.

114

Benjamin
 
EJ
,
Thomas
 
KL
,
Go
 
AS
,
Desvigne-Nickens
 
P
,
Albert
 
CM
,
Alonso
 
A
, et al.  
Transforming atrial fibrillation research to integrate social determinants of health: a national heart, lung, and blood institute workshop report
.
JAMA Cardiol
 
2023
;
8
:
182
91
.

115

Karlsson
 
LO
,
Nilsson
 
S
,
Bang
 
M
,
Nilsson
 
L
,
Charitakis
 
E
,
Janzon
 
M
.
A clinical decision support tool for improving adherence to guidelines on anticoagulant therapy in patients with atrial fibrillation at risk of stroke: a cluster-randomized trial in a Swedish primary care setting (the CDS-AF study)
.
PLoS Med
 
2018
;
15
:
e1002528
.

116

Biersteker
 
TE
,
Schalij
 
MJ
,
Treskes
 
RW
.
Impact of mobile health devices for the detection of atrial fibrillation: systematic review
.
JMIR Mhealth Uhealth
 
2021
;
9
:
e26161
.

117

Romiti
 
GF
,
Pastori
 
D
,
Rivera-Caravaca
 
JM
,
Ding
 
WY
,
Gue
 
YX
,
Menichelli
 
D
, et al.  
Adherence to the ‘atrial fibrillation better care’ pathway in patients with atrial fibrillation: impact on clinical outcomes—a systematic review and meta-analysis of 285,000 patients
.
Thromb Haemost
 
2022
;
122
:
406
14
.

118

Gallagher
 
C
,
Elliott
 
AD
,
Wong
 
CX
,
Rangnekar
 
G
,
Middeldorp
 
ME
,
Mahajan
 
R
, et al.  
Integrated care in atrial fibrillation: a systematic review and meta-analysis
.
Heart
 
2017
;
103
:
1947
53
.

119

Kirchhof
 
P
,
Benussi
 
S
,
Kotecha
 
D
,
Ahlsson
 
A
,
Atar
 
D
,
Casadei
 
B
, et al.  
2016 ESC Guidelines for the management of atrial fibrillation developed in collaboration with EACTS
.
Eur Heart J
 
2016
;
37
:
2893
962
.

120

Hindricks
 
G
,
Potpara
 
T
,
Dagres
 
N
,
Arbelo
 
E
,
Bax
 
JJ
,
Blomström-Lundqvist
 
C
, et al.  
2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): the task force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC
.
Eur Heart J
 
2021
;
42
:
373
498
.

121

Qvist
 
I
,
Hendriks
 
JM
,
Møller
 
DS
,
Albertsen
 
AE
,
Mogensen
 
HM
,
Oddershede
 
GD
, et al.  
Effectiveness of structured, hospital-based, nurse-led atrial fibrillation clinics: a comparison between a real-world population and a clinical trial population
.
Open Heart
 
2016
;
3
:
e000335
.

122

Hendriks
 
JM
,
de Wit
 
R
,
Crijns
 
HJ
,
Vrijhoef
 
HJ
,
Prins
 
MH
,
Pisters
 
R
, et al.  
Nurse-led care vs. usual care for patients with atrial fibrillation: results of a randomized trial of integrated chronic care vs. routine clinical care in ambulatory patients with atrial fibrillation
.
Eur Heart J
 
2012
;
33
:
2692
9
.

123

Carter
 
L
,
Gardner
 
M
,
Magee
 
K
,
Fearon
 
A
,
Morgulis
 
I
,
Doucette
 
S
, et al.  
An integrated management approach to atrial fibrillation
.
J Am Heart Assoc
 
2016
;
5
:
e002950
.

124

van den Dries
 
CJ
,
van Doorn
 
S
,
Rutten
 
FH
,
Oudega
 
R
,
van de Leur
 
S
,
Elvan
 
A
, et al.  
Integrated management of atrial fibrillation in primary care: results of the ALL-IN cluster randomized trial
.
Eur Heart J
 
2020
;
41
:
2836
44
.

125

Abed
 
HS
,
Wittert
 
GA
,
Leong
 
DP
,
Shirazi
 
MG
,
Bahrami
 
B
,
Middeldorp
 
ME
, et al.  
Effect of weight reduction and cardiometabolic risk factor management on symptom burden and severity in patients with atrial fibrillation: a randomized clinical trial
.
JAMA
 
2013
;
310
:
2050
60
.

126

Pathak
 
RK
,
Middeldorp
 
ME
,
Meredith
 
M
,
Mehta
 
AB
,
Mahajan
 
R
,
Wong
 
CX
, et al.  
Long-term effect of goal-directed weight management in an atrial fibrillation cohort: a long-term follow-up study (LEGACY)
.
J Am Coll Cardiol
 
2015
;
65
:
2159
69
.

127

Middeldorp
 
ME
,
Pathak
 
RK
,
Meredith
 
M
,
Mehta
 
AB
,
Elliott
 
AD
,
Mahajan
 
R
, et al.  
PREVEntion and regReSsive effect of weight-loss and risk factor modification on atrial fibrillation: the REVERSE-AF study
.
Europace
 
2018
;
20
:
1929
35
.

128

Pathak
 
RK
,
Middeldorp
 
ME
,
Lau
 
DH
,
Mehta
 
AB
,
Mahajan
 
R
,
Twomey
 
D
, et al.  
Aggressive risk factor reduction study for atrial fibrillation and implications for the outcome of ablation: the ARREST-AF cohort study
.
J Am Coll Cardiol
 
2014
;
64
:
2222
31
.

129

Pinho-Gomes
 
AC
,
Azevedo
 
L
,
Copland
 
E
,
Canoy
 
D
,
Nazarzadeh
 
M
,
Ramakrishnan
 
R
, et al.  
Blood pressure-lowering treatment for the prevention of cardiovascular events in patients with atrial fibrillation: an individual participant data meta-analysis
.
PLoS Med
 
2021
;
18
:
e1003599
.

130

Parkash
 
R
,
Wells
 
GA
,
Sapp
 
JL
,
Healey
 
JS
,
Tardif
 
J-C
,
Greiss
 
I
, et al.  
Effect of aggressive blood pressure control on the recurrence of atrial fibrillation after catheter ablation: a randomized, open-label clinical trial (SMAC-AF [Substrate Modification with Aggressive Blood Pressure Control])
.
Circulation
 
2017
;
135
:
1788
98
.

131

McMurray
 
JJ
,
Adamopoulos
 
S
,
Anker
 
SD
,
Auricchio
 
A
,
Bohm
 
M
,
Dickstein
 
K
, et al.  
ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: the task force for the diagnosis and treatment of acute and chronic heart failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC
.
Eur Heart J
 
2012
;
33
:
1787
847
.

132

Olsson
 
LG
,
Swedberg
 
K
,
Ducharme
 
A
,
Granger
 
CB
,
Michelson
 
EL
,
McMurray
 
JJ
, et al.  
Atrial fibrillation and risk of clinical events in chronic heart failure with and without left ventricular systolic dysfunction: results from the Candesartan in Heart failure-Assessment of Reduction in Mortality and morbidity (CHARM) program
.
J Am Coll Cardiol
 
2006
;
47
:
1997
2004
.

133

Kotecha
 
D
,
Holmes
 
J
,
Krum
 
H
,
Altman
 
DG
,
Manzano
 
L
,
Cleland
 
JG
, et al.  
Efficacy of beta blockers in patients with heart failure plus atrial fibrillation: an individual-patient data meta-analysis
.
Lancet
 
2014
;
384
:
2235
43
.

134

Zannad
 
F
,
McMurray
 
JJ
,
Krum
 
H
,
van Veldhuisen
 
DJ
,
Swedberg
 
K
,
Shi
 
H
, et al.  
Eplerenone in patients with systolic heart failure and mild symptoms
.
N Engl J Med
 
2011
;
364
:
11
21
.

135

McMurray
 
JJ
,
Packer
 
M
,
Desai
 
AS
,
Gong
 
J
,
Lefkowitz
 
MP
,
Rizkala
 
AR
, et al.  
Angiotensin-neprilysin inhibition versus enalapril in heart failure
.
N Engl J Med
 
2014
;
371
:
993
1004
.

136

Pandey
 
AK
,
Okaj
 
I
,
Kaur
 
H
,
Belley-Cote
 
EP
,
Wang
 
J
,
Oraii
 
A
, et al.  
Sodium-glucose co-transporter inhibitors and atrial fibrillation: a systematic review and meta-analysis of randomized controlled trials
.
J Am Heart Assoc
 
2021
;
10
:
e022222
.

137

McDonagh
 
TA
,
Metra
 
M
,
Adamo
 
M
,
Gardner
 
RS
,
Baumbach
 
A
,
Bohm
 
M
, et al.  
2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure
.
Eur Heart J
 
2021
;
42
:
3599
726
.

138

Solomon
 
SD
,
McMurray
 
JJV
,
Claggett
 
B
,
de Boer
 
RA
,
DeMets
 
D
,
Hernandez
 
AF
, et al.  
Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction
.
N Engl J Med
 
2022
;
387
:
1089
98
.

139

Anker
 
SD
,
Butler
 
J
,
Filippatos
 
G
,
Ferreira
 
JP
,
Bocchi
 
E
,
Böhm
 
M
, et al.  
Empagliflozin in heart failure with a preserved ejection fraction
.
N Engl J Med
 
2021
;
385
:
1451
61
.

140

Bhatt
 
DL
,
Szarek
 
M
,
Steg
 
PG
,
Cannon
 
CP
,
Leiter
 
LA
,
McGuire
 
DK
, et al.  
Sotagliflozin in patients with diabetes and recent worsening heart failure
.
N Engl J Med
 
2021
;
384
:
117
28
.

141

Pathak
 
RK
,
Elliott
 
A
,
Middeldorp
 
ME
,
Meredith
 
M
,
Mehta
 
AB
,
Mahajan
 
R
, et al.  
Impact of CARDIOrespiratory FITness on arrhythmia recurrence in obese individuals with atrial fibrillation: the CARDIO-FIT study
.
J Am Coll Cardiol
 
2015
;
66
:
985
96
.

142

Hegbom
 
F
,
Stavem
 
K
,
Sire
 
S
,
Heldal
 
M
,
Orning
 
OM
,
Gjesdal
 
K
.
Effects of short-term exercise training on symptoms and quality of life in patients with chronic atrial fibrillation
.
Int J Cardiol
 
2007
;
116
:
86
92
.

143

Osbak
 
PS
,
Mourier
 
M
,
Kjaer
 
A
,
Henriksen
 
JH
,
Kofoed
 
KF
,
Jensen
 
GB
.
A randomized study of the effects of exercise training on patients with atrial fibrillation
.
Am Heart J
 
2011
;
162
:
1080
7
.

144

Malmo
 
V
,
Nes
 
BM
,
Amundsen
 
BH
,
Tjonna
 
AE
,
Stoylen
 
A
,
Rossvoll
 
O
, et al.  
Aerobic interval training reduces the burden of atrial fibrillation in the short term: a randomized trial
.
Circulation
 
2016
;
133
:
466
73
.

145

Oesterle
 
A
,
Giancaterino
 
S
,
Van Noord
 
MG
,
Pellegrini
 
CN
,
Fan
 
D
,
Srivatsa
 
UN
, et al.  
Effects of supervised exercise training on atrial fibrillation: a meta-analysis of randomized controlled trials
.
J Cardiopulm Rehabil Prev
 
2022
;
42
:
258
65
.

146

Elliott
 
AD
,
Verdicchio
 
CV
,
Mahajan
 
R
,
Middeldorp
 
ME
,
Gallagher
 
C
,
Mishima
 
RS
, et al.  
An exercise and physical activity program in patients with atrial fibrillation: the ACTIVE-AF randomized controlled trial
.
JACC Clin Electrophysiol
 
2023
;
9
:
455
65
.

147

Voskoboinik
 
A
,
Kalman
 
JM
,
De Silva
 
A
,
Nicholls
 
T
,
Costello
 
B
,
Nanayakkara
 
S
, et al.  
Alcohol abstinence in drinkers with atrial fibrillation
.
N Engl J Med
 
2020
;
382
:
20
8
.

148

Holmqvist
 
F
,
Guan
 
N
,
Zhu
 
Z
,
Kowey
 
PR
,
Allen
 
LA
,
Fonarow
 
GC
, et al.  
Impact of obstructive sleep apnea and continuous positive airway pressure therapy on outcomes in patients with atrial fibrillation-results from the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation (ORBIT-AF)
.
Am Heart J
 
2015
;
169
:
647
654.e2
.

149

Fein
 
AS
,
Shvilkin
 
A
,
Shah
 
D
,
Haffajee
 
CI
,
Das
 
S
,
Kumar
 
K
, et al.  
Treatment of obstructive sleep apnea reduces the risk of atrial fibrillation recurrence after catheter ablation
.
J Am Coll Cardiol
 
2013
;
62
:
300
5
.

150

Li
 
L
,
Wang
 
ZW
,
Li
 
J
,
Ge
 
X
,
Guo
 
LZ
,
Wang
 
Y
, et al.  
Efficacy of catheter ablation of atrial fibrillation in patients with obstructive sleep apnoea with and without continuous positive airway pressure treatment: a meta-analysis of observational studies
.
Europace
 
2014
;
16
:
1309
14
.

151

Naruse
 
Y
,
Tada
 
H
,
Satoh
 
M
,
Yanagihara
 
M
,
Tsuneoka
 
H
,
Hirata
 
Y
, et al.  
Concomitant obstructive sleep apnea increases the recurrence of atrial fibrillation following radiofrequency catheter ablation of atrial fibrillation: clinical impact of continuous positive airway pressure therapy
.
Heart Rhythm
 
2013
;
10
:
331
7
.

152

Qureshi
 
WT
,
Nasir
 
UB
,
Alqalyoobi
 
S
,
O’Neal
 
WT
,
Mawri
 
S
,
Sabbagh
 
S
, et al.  
Meta-analysis of continuous positive airway pressure as a therapy of atrial fibrillation in obstructive sleep apnea
.
Am J Cardiol
 
2015
;
116
:
1767
73
.

153

Shukla
 
A
,
Aizer
 
A
,
Holmes
 
D
,
Fowler
 
S
,
Park
 
DS
,
Bernstein
 
S
, et al.  
Effect of obstructive sleep apnea treatment on atrial fibrillation recurrence: a meta-analysis
.
JACC Clin Electrophysiol
 
2015
;
1
:
41
51
.

154

Nalliah
 
CJ
,
Wong
 
GR
,
Lee
 
G
,
Voskoboinik
 
A
,
Kee
 
K
,
Goldin
 
J
, et al.  
Impact of CPAP on the atrial fibrillation substrate in obstructive sleep apnea: the SLEEP-AF study
.
JACC Clin Electrophysiol
 
2022
;
8
:
869
77
.

155

Kadhim
 
K
,
Middeldorp
 
ME
,
Elliott
 
AD
,
Jones
 
D
,
Hendriks
 
JML
,
Gallagher
 
C
, et al.  
Self-reported daytime sleepiness and sleep-disordered breathing in patients with atrial fibrillation: SNOozE-AF
.
Can J Cardiol
 
2019
;
35
:
1457
64
.

156

Traaen
 
GM
,
Overland
 
B
,
Aakeroy
 
L
,
Hunt
 
TE
,
Bendz
 
C
,
Sande
 
L
, et al.  
Prevalence, risk factors, and type of sleep apnea in patients with paroxysmal atrial fibrillation
.
Int J Cardiol Heart Vasc
 
2020
;
26
:
100447
.

157

Kadhim
 
K
,
Middeldorp
 
ME
,
Elliott
 
AD
,
Agbaedeng
 
T
,
Gallagher
 
C
,
Malik
 
V
, et al.  
Prevalence and assessment of sleep-disordered breathing in patients with atrial fibrillation: a systematic review and meta-analysis
.
Can J Cardiol
 
2021
;
37
:
1846
56
.

158

Friberg
 
L
,
Rosenqvist
 
M
,
Lip
 
GY
.
Evaluation of risk stratification schemes for ischaemic stroke and bleeding in 182 678 patients with atrial fibrillation: the Swedish atrial fibrillation cohort study
.
Eur Heart J
 
2012
;
33
:
1500
10
.

159

Lopes
 
LC
,
Spencer
 
FA
,
Neumann
 
I
,
Ventresca
 
M
,
Ebrahim
 
S
,
Zhou
 
Q
, et al.  
Systematic review of observational studies assessing bleeding risk in patients with atrial fibrillation not using anticoagulants
.
PLoS One
 
2014
;
9
:
e88131
.

160

Potpara
 
TS
,
Polovina
 
MM
,
Licina
 
MM
,
Marinkovic
 
JM
,
Lip
 
GY
.
Predictors and prognostic implications of incident heart failure following the first diagnosis of atrial fibrillation in patients with structurally normal hearts: the Belgrade atrial fibrillation study
.
Eur J Heart Fail
 
2013
;
15
:
415
24
.

161

Noubiap
 
JJ
,
Feteh
 
VF
,
Middeldorp
 
ME
,
Fitzgerald
 
JL
,
Thomas
 
G
,
Kleinig
 
T
, et al.  
A meta-analysis of clinical risk factors for stroke in anticoagulant-naive patients with atrial fibrillation
.
Europace
 
2021
;
23
:
1528
38
.

162

McEvoy JW, Touyz RM, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, et al. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension. Eur Heart J 2024.

163

Santoro
 
F
,
Di Biase
 
L
,
Trivedi
 
C
,
Burkhardt
 
JD
,
Paoletti Perini
 
A
,
Sanchez
 
J
, et al.  
Impact of uncontrolled hypertension on atrial fibrillation ablation outcome
.
JACC Clin Electrophysiol
 
2015
;
1
:
164
73
.

164

Trines
 
SA
,
Stabile
 
G
,
Arbelo
 
E
,
Dagres
 
N
,
Brugada
 
J
,
Kautzner
 
J
, et al.  
Influence of risk factors in the ESC-EHRA EORP atrial fibrillation ablation long-term registry
.
Pacing Clin Electrophysiol
 
2019
;
42
:
1365
73
.

165

Shah
 
AN
,
Mittal
 
S
,
Sichrovsky
 
TC
,
Cotiga
 
D
,
Arshad
 
A
,
Maleki
 
K
, et al.  
Long-term outcome following successful pulmonary vein isolation: pattern and prediction of very late recurrence
.
J Cardiovasc Electrophysiol
 
2008
;
19
:
661
7
.

166

Berruezo
 
A
,
Tamborero
 
D
,
Mont
 
L
,
Benito
 
B
,
Tolosana
 
JM
,
Sitges
 
M
, et al.  
Pre-procedural predictors of atrial fibrillation recurrence after circumferential pulmonary vein ablation
.
Eur Heart J
 
2007
;
28
:
836
41
.

167

Themistoclakis
 
S
,
Schweikert
 
RA
,
Saliba
 
WI
,
Bonso
 
A
,
Rossillo
 
A
,
Bader
 
G
, et al.  
Clinical predictors and relationship between early and late atrial tachyarrhythmias after pulmonary vein antrum isolation
.
Heart Rhythm
 
2008
;
5
:
679
85
.

168

Letsas
 
KP
,
Weber
 
R
,
Burkle
 
G
,
Mihas
 
CC
,
Minners
 
J
,
Kalusche
 
D
, et al.  
Pre-ablative predictors of atrial fibrillation recurrence following pulmonary vein isolation: the potential role of inflammation
.
Europace
 
2009
;
11
:
158
63
.

169

Khaykin
 
Y
,
Oosthuizen
 
R
,
Zarnett
 
L
,
Essebag
 
V
,
Parkash
 
R
,
Seabrook
 
C
, et al.  
Clinical predictors of arrhythmia recurrences following pulmonary vein antrum isolation for atrial fibrillation: predicting arrhythmia recurrence post-PVAI
.
J Cardiovasc Electrophysiol
 
2011
;
22
:
1206
14
.

170

Kamioka
 
M
,
Hijioka
 
N
,
Matsumoto
 
Y
,
Nodera
 
M
,
Kaneshiro
 
T
,
Suzuki
 
H
, et al.  
Uncontrolled blood pressure affects atrial remodeling and adverse clinical outcome in paroxysmal atrial fibrillation
.
Pacing Clin Electrophysiol
 
2018
;
41
:
402
10
.

171

Zylla
 
MM
,
Hochadel
 
M
,
Andresen
 
D
,
Brachmann
 
J
,
Eckardt
 
L
,
Hoffmann
 
E
, et al.  
Ablation of atrial fibrillation in patients with hypertension—an analysis from the German ablation registry
.
J Clin Med
 
2020
;
9
:
1
14
.

172

Galzerano
 
D
,
Di Michele
 
S
,
Paolisso
 
G
,
Tuccillo
 
B
,
Lama
 
D
,
Carbotta
 
S
, et al.  
A multicentre, randomized study of telmisartan versus carvedilol for prevention of atrial fibrillation recurrence in hypertensive patients
.
J Renin Angiotensin Aldosterone Syst
 
2012
;
13
:
496
503
.

173

Du
 
H
,
Fan
 
J
,
Ling
 
Z
,
Woo
 
K
,
Su
 
L
,
Chen
 
S
, et al.  
Effect of nifedipine versus telmisartan on prevention of atrial fibrillation recurrence in hypertensive patients
.
Hypertension
 
2013
;
61
:
786
92
.

174

Giannopoulos
 
G
,
Kossyvakis
 
C
,
Efremidis
 
M
,
Katsivas
 
A
,
Panagopoulou
 
V
,
Doudoumis
 
K
, et al.  
Central sympathetic inhibition to reduce postablation atrial fibrillation recurrences in hypertensive patients: a randomized, controlled study
.
Circulation
 
2014
;
130
:
1346
52
.

175

Schneider
 
MP
,
Hua
 
TA
,
Bohm
 
M
,
Wachtell
 
K
,
Kjeldsen
 
SE
,
Schmieder
 
RE
.
Prevention of atrial fibrillation by renin-angiotensin system inhibition a meta-analysis
.
J Am Coll Cardiol
 
2010
;
55
:
2299
307
.

176

Blum
 
S
,
Aeschbacher
 
S
,
Meyre
 
P
,
Zwimpfer
 
L
,
Reichlin
 
T
,
Beer
 
JH
, et al.  
Incidence and predictors of atrial fibrillation progression
.
J Am Heart Assoc
 
2019
;
8
:
e012554
.

177

Kotecha
 
D
,
Piccini
 
JP
.
Atrial fibrillation in heart failure: what should we do?
 
Eur Heart J
 
2015
;
36
:
3250
7
.

178

Santhanakrishnan
 
R
,
Wang
 
N
,
Larson
 
MG
,
Magnani
 
JW
,
McManus
 
DD
,
Lubitz
 
SA
, et al.  
Atrial fibrillation begets heart failure and vice versa: temporal associations and differences in preserved versus reduced ejection fraction
.
Circulation
 
2016
;
133
:
484
92
.

179

Rossello
 
X
,
Gil
 
V
,
Escoda
 
R
,
Jacob
 
J
,
Aguirre
 
A
,
Martín-Sánchez
 
FJ
, et al.  
Editor’s choice – impact of identifying precipitating factors on 30-day mortality in acute heart failure patients
.
Eur Heart J Acute Cardiovasc Care
 
2019
;
8
:
667
80
.

180

Atrial Fibrillation Investigators
.
Echocardiographic predictors of stroke in patients with atrial fibrillation: a prospective study of 1066 patients from 3 clinical trials
.
Arch Intern Med
 
1998
;
158
:
1316
20
.

181

Rohla
 
M
,
Weiss
 
TW
,
Pecen
 
L
,
Patti
 
G
,
Siller-Matula
 
JM
,
Schnabel
 
RB
, et al.  
Risk factors for thromboembolic and bleeding events in anticoagulated patients with atrial fibrillation: the prospective, multicentre observational PREvention oF thromboembolic events—European Registry in Atrial Fibrillation (PREFER in AF)
.
BMJ Open
 
2019
;
9
:
e022478
.

182

Kotecha
 
D
,
Chudasama
 
R
,
Lane
 
DA
,
Kirchhof
 
P
,
Lip
 
GY
.
Atrial fibrillation and heart failure due to reduced versus preserved ejection fraction: a systematic review and meta-analysis of death and adverse outcomes
.
Int J Cardiol
 
2016
;
203
:
660
6
.

183

McDonagh
 
TA
,
Metra
 
M
,
Adamo
 
M
,
Gardner
 
RS
,
Baumbach
 
A
,
Böhm
 
M
, et al.  
2023 focused update of the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure
.
Eur Heart J
 
2023
;
44
:
3627
39
.

184

ACTIVE I Investigators
;
Yusuf
 
S
,
Healey
 
JS
,
Pogue
 
J
,
Chrolavicius
 
S
,
Flather
 
M
, et al.  
Irbesartan in patients with atrial fibrillation
.
N Engl J Med
 
2011
;
364
:
928
38
.

185

Ziff
 
OJ
,
Lane
 
DA
,
Samra
 
M
,
Griffith
 
M
,
Kirchhof
 
P
,
Lip
 
GY
, et al.  
Safety and efficacy of digoxin: systematic review and meta-analysis of observational and controlled trial data
.
BMJ
 
2015
;
351
:
h4451
.

186

Groenveld
 
HF
,
Crijns
 
HJ
,
Van den Berg
 
MP
,
Van Sonderen
 
E
,
Alings
 
AM
,
Tijssen
 
JG
, et al.  
The effect of rate control on quality of life in patients with permanent atrial fibrillation: data from the RACE II (Rate Control Efficacy in Permanent Atrial Fibrillation II) study
.
J Am Coll Cardiol
 
2011
;
58
:
1795
803
.

187

Glikson
 
M
,
Nielsen
 
JC
,
Kronborg
 
MB
,
Michowitz
 
Y
,
Auricchio
 
A
,
Barbash
 
IM
, et al.  
2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy
.
Eur Heart J
 
2021
;
42
:
3427
520
.

188

Solomon
 
SD
,
Claggett
 
B
,
Lewis
 
EF
,
Desai
 
A
,
Anand
 
I
,
Sweitzer
 
NK
, et al.  
Influence of ejection fraction on outcomes and efficacy of spironolactone in patients with heart failure with preserved ejection fraction
.
Eur Heart J
 
2016
;
37
:
455
62
.

189

Lund
 
LH
,
Claggett
 
B
,
Liu
 
J
,
Lam
 
CS
,
Jhund
 
PS
,
Rosano
 
GM
, et al.  
Heart failure with mid-range ejection fraction in CHARM: characteristics, outcomes and effect of candesartan across the entire ejection fraction spectrum
.
Eur J Heart Fail
 
2018
;
20
:
1230
9
.

190

Cleland
 
JGF
,
Bunting
 
KV
,
Flather
 
MD
,
Altman
 
DG
,
Holmes
 
J
,
Coats
 
AJS
, et al.  
Beta-blockers for heart failure with reduced, mid-range, and preserved ejection fraction: an individual patient-level analysis of double-blind randomized trials
.
Eur Heart J
 
2018
;
39
:
26
35
.

191

Țica
 
O
,
Khamboo
 
W
,
Kotecha
 
D
.
Breaking the cycle of heart failure with preserved ejection fraction and atrial fibrillation
.
Card Fail Rev
 
2022
;
8
:
e32
.

192

Nguyen
 
BO
,
Crijns
 
H
,
Tijssen
 
JGP
,
Geelhoed
 
B
,
Hobbelt
 
AH
,
Hemels
 
MEW
, et al.  
Long-term outcome of targeted therapy of underlying conditions in patients with early persistent atrial fibrillation and heart failure: data of the RACE 3 trial
.
Europace
 
2022
;
24
:
910
20
.

193

Wang
 
A
,
Green
 
JB
,
Halperin
 
JL
,
Piccini
 
JP
, Sr
.
Atrial fibrillation and diabetes mellitus: JACC review topic of the week
.
J Am Coll Cardiol
 
2019
;
74
:
1107
15
.

194

Alijla
 
F
,
Buttia
 
C
,
Reichlin
 
T
,
Razvi
 
S
,
Minder
 
B
,
Wilhelm
 
M
, et al.  
Association of diabetes with atrial fibrillation types: a systematic review and meta-analysis
.
Cardiovasc Diabetol
 
2021
;
20
:
230
.

195

Ding
 
WY
,
Kotalczyk
 
A
,
Boriani
 
G
,
Marin
 
F
,
Blomstrom-Lundqvist
 
C
,
Potpara
 
TS
, et al.  
Impact of diabetes on the management and outcomes in atrial fibrillation: an analysis from the ESC-EHRA EORP-AF long-term general registry
.
Eur J Intern Med
 
2022
;
103
:
41
9
.

196

Proietti
 
M
,
Romiti
 
GF
,
Basili
 
S
.
The case of diabetes mellitus and atrial fibrillation: underlining the importance of non-cardiovascular comorbidities
.
Eur J Intern Med
 
2022
;
103
:
38
40
.

197

Karayiannides
 
S
,
Norhammar
 
A
,
Landstedt-Hallin
 
L
,
Friberg
 
L
,
Lundman
 
P
.
Prognostic impact of type 1 and type 2 diabetes mellitus in atrial fibrillation and the effect of severe hypoglycaemia: a nationwide cohort study
.
Eur J Prev Cardiol
 
2022
;
29
:
1759
69
.

198

Lip
 
GY
,
Nieuwlaat
 
R
,
Pisters
 
R
,
Lane
 
DA
,
Crijns
 
HJ
.
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
72
.

199

Abdel-Qadir
 
H
,
Gunn
 
M
,
Lega
 
IC
,
Pang
 
A
,
Austin
 
PC
,
Singh
 
SM
, et al.  
Association of diabetes duration and glycemic control with stroke rate in patients with atrial fibrillation and diabetes: a population-based cohort study
.
J Am Heart Assoc
 
2022
;
11
:
e023643
.

200

Donnellan
 
E
,
Aagaard
 
P
,
Kanj
 
M
,
Jaber
 
W
,
Elshazly
 
M
,
Hoosien
 
M
, et al.  
Association between pre-ablation glycemic control and outcomes among patients with diabetes undergoing atrial fibrillation ablation
.
JACC Clin Electrophysiol
 
2019
;
5
:
897
903
.

201

D’Souza
 
S
,
Elshazly
 
MB
,
Dargham
 
SR
,
Donnellan
 
E
,
Asaad
 
N
,
Hayat
 
S
, et al.  
Atrial fibrillation catheter ablation complications in obese and diabetic patients: insights from the US nationwide inpatient sample 2005–2013
.
Clin Cardiol
 
2021
;
44
:
1151
60
.

202

Creta
 
A
,
Providencia
 
R
,
Adragao
 
P
,
de Asmundis
 
C
,
Chun
 
J
,
Chierchia
 
G
, et al.  
Impact of type-2 diabetes mellitus on the outcomes of catheter ablation of atrial fibrillation (European Observational Multicentre Study)
.
Am J Cardiol
 
2020
;
125
:
901
6
.

203

Wang
 
Z
,
Wang
 
YJ
,
Liu
 
ZY
,
Li
 
Q
,
Kong
 
YW
,
Chen
 
YW
, et al.  
Effect of insulin resistance on recurrence after radiofrequency catheter ablation in patients with atrial fibrillation
.
Cardiovasc Drugs Ther
 
2023
;
37
:
705
13
.

204

Papazoglou
 
AS
,
Kartas
 
A
,
Moysidis
 
DV
,
Tsagkaris
 
C
,
Papadakos
 
SP
,
Bekiaridou
 
A
, et al.  
Glycemic control and atrial fibrillation: an intricate relationship, yet under investigation
.
Cardiovasc Diabetol
 
2022
;
21
:
39
.

205

Zhang
 
Z
,
Zhang
 
X
,
Korantzopoulos
 
P
,
Letsas
 
KP
,
Tse
 
G
,
Gong
 
M
, et al.  
Thiazolidinedione use and atrial fibrillation in diabetic patients: a meta-analysis
.
BMC Cardiovasc Disord
 
2017
;
17
:
96
.

206

Bell
 
DSH
,
Goncalves
 
E
.
Atrial fibrillation and type 2 diabetes: prevalence, etiology, pathophysiology and effect of anti-diabetic therapies
.
Diabetes Obes Metab
 
2019
;
21
:
210
7
.

207

Marx
 
N
,
Federici
 
M
,
Schütt
 
K
,
Müller-Wieland
 
D
,
Ajjan
 
RA
,
Antunes
 
MJ
, et al.  
2023 ESC Guidelines for the management of cardiovascular disease in patients with diabetes
.
Eur Heart J
 
2023
;
44
:
4043
140
.

208

Di Benedetto
 
L
,
Michels
 
G
,
Luben
 
R
,
Khaw
 
KT
,
Pfister
 
R
.
Individual and combined impact of lifestyle factors on atrial fibrillation in apparently healthy men and women: the EPIC-Norfolk prospective population study
.
Eur J Prev Cardiol
 
2018
;
25
:
1374
83
.

209

Grundvold
 
I
,
Bodegard
 
J
,
Nilsson
 
PM
,
Svennblad
 
B
,
Johansson
 
G
,
Ostgren
 
CJ
, et al.  
Body weight and risk of atrial fibrillation in 7,169 patients with newly diagnosed type 2 diabetes; an observational study
.
Cardiovasc Diabetol
 
2015
;
14
:
5
.

210

Wong
 
CX
,
Sullivan
 
T
,
Sun
 
MT
,
Mahajan
 
R
,
Pathak
 
RK
,
Middeldorp
 
M
, et al.  
Obesity and the risk of incident, post-operative, and post-ablation atrial fibrillation: a meta-analysis of 626,603 individuals in 51 studies
.
JACC Clin Electrophysiol
 
2015
;
1
:
139
52
.

211

Providencia
 
R
,
Adragao
 
P
,
de Asmundis
 
C
,
Chun
 
J
,
Chierchia
 
G
,
Defaye
 
P
, et al.  
Impact of body mass index on the outcomes of catheter ablation of atrial fibrillation: a European observational multicenter study
.
J Am Heart Assoc
 
2019
;
8
:
e012253
.

212

Glover
 
BM
,
Hong
 
KL
,
Dagres
 
N
,
Arbelo
 
E
,
Laroche
 
C
,
Riahi
 
S
, et al.  
Impact of body mass index on the outcome of catheter ablation of atrial fibrillation
.
Heart
 
2019
;
105
:
244
50
.

213

Gessler
 
N
,
Willems
 
S
,
Steven
 
D
,
Aberle
 
J
,
Akbulak
 
RO
,
Gosau
 
N
, et al.  
Supervised obesity reduction trial for AF ablation patients: results from the SORT-AF trial
.
Europace
 
2021
;
23
:
1548
58
.

214

Mohanty
 
S
,
Mohanty
 
P
,
Natale
 
V
,
Trivedi
 
C
,
Gianni
 
C
,
Burkhardt
 
JD
, et al.  
Impact of weight loss on ablation outcome in obese patients with longstanding persistent atrial fibrillation
.
J Cardiovasc Electrophysiol
 
2018
;
29
:
246
53
.

215

Donnellan
 
E
,
Wazni
 
OM
,
Kanj
 
M
,
Elshazly
 
M
,
Hussein
 
AA
,
Patel
 
DR
, et al.  
Impact of risk-factor modification on arrhythmia recurrence among morbidly obese patients undergoing atrial fibrillation ablation
.
J Cardiovasc Electrophysiol
 
2020
;
31
:
1979
86
.

216

Donnellan
 
E
,
Wazni
 
OM
,
Kanj
 
M
,
Baranowski
 
B
,
Cremer
 
P
,
Harb
 
S
, et al.  
Association between pre-ablation bariatric surgery and atrial fibrillation recurrence in morbidly obese patients undergoing atrial fibrillation ablation
.
Europace
 
2019
;
21
:
1476
83
.

217

Donnellan
 
E
,
Wazni
 
O
,
Kanj
 
M
,
Hussein
 
A
,
Baranowski
 
B
,
Lindsay
 
B
, et al.  
Outcomes of atrial fibrillation ablation in morbidly obese patients following bariatric surgery compared with a nonobese cohort
.
Circ Arrhythm Electrophysiol
 
2019
;
12
:
e007598
.

218

Moula
 
AI
,
Parrini
 
I
,
Tetta
 
C
,
Lucà
 
F
,
Parise
 
G
,
Rao
 
CM
, et al.  
Obstructive sleep apnea and atrial fibrillation
.
J Clin Med
 
2022
;
11
:
1242
.

219

Kapur
 
VK
,
Auckley
 
DH
,
Chowdhuri
 
S
,
Kuhlmann
 
DC
,
Mehra
 
R
,
Ramar
 
K
, et al.  
Clinical practice guideline for diagnostic testing for adult obstructive sleep apnea: an American academy of sleep medicine clinical practice guideline
.
J Clin Sleep Med
 
2017
;
13
:
479
504
.

220

Linz
 
D
,
Brooks
 
AG
,
Elliott
 
AD
,
Nalliah
 
CJ
,
Hendriks
 
JML
,
Middeldorp
 
ME
, et al.  
Variability of sleep apnea severity and risk of atrial fibrillation: the VARIOSA-AF study
.
JACC Clin Electrophysiol
 
2019
;
5
:
692
701
.

221

Linz
 
D
,
Linz
 
B
,
Dobrev
 
D
,
Baumert
 
M
,
Hendriks
 
JM
,
Pepin
 
JL
, et al.  
Personalized management of sleep apnea in patients with atrial fibrillation: an interdisciplinary and translational challenge
.
Int J Cardiol Heart Vasc
 
2021
;
35
:
100843
. .

222

Kanagala
 
R
,
Murali
 
NS
,
Friedman
 
PA
,
Ammash
 
NM
,
Gersh
 
BJ
,
Ballman
 
KV
, et al.  
Obstructive sleep apnea and the recurrence of atrial fibrillation
.
Circulation
 
2003
;
107
:
2589
94
.

223

Abumuamar
 
AM
,
Newman
 
D
,
Dorian
 
P
,
Shapiro
 
CM
.
Cardiac effects of CPAP treatment in patients with obstructive sleep apnea and atrial fibrillation
.
J Interv Card Electrophysiol
 
2019
;
54
:
289
97
.

224

Mittal
 
S
,
Golombeck
 
D
,
Pimienta
 
J
.
Sleep apnoea and AF: where do we stand? Practical advice for clinicians
.
Arrhythm Electrophysiol Rev
 
2021
;
10
:
140
6
.

225

Hunt
 
TE
,
Traaen
 
GM
,
Aakeroy
 
L
,
Bendz
 
C
,
Overland
 
B
,
Akre
 
H
, et al.  
Effect of continuous positive airway pressure therapy on recurrence of atrial fibrillation after pulmonary vein isolation in patients with obstructive sleep apnea: a randomized controlled trial
.
Heart Rhythm
 
2022
;
19
:
1433
41
.

226

Caples
 
SM
,
Mansukhani
 
MP
,
Friedman
 
PA
,
Somers
 
VK
.
The impact of continuous positive airway pressure treatment on the recurrence of atrial fibrillation post cardioversion: a randomized controlled trial
.
Int J Cardiol
 
2019
;
278
:
133
6
.

227

Labarca
 
G
,
Dreyse
 
J
,
Drake
 
L
,
Jorquera
 
J
,
Barbe
 
F
.
Efficacy of continuous positive airway pressure (CPAP) in the prevention of cardiovascular events in patients with obstructive sleep apnea: systematic review and meta-analysis
.
Sleep Med Rev
 
2020
;
52
:
101312
.

228

Abuzaid
 
AS
,
Al Ashry
 
HS
,
Elbadawi
 
A
,
Ld
 
H
,
Saad
 
M
,
Elgendy
 
IY
, et al.  
Meta-analysis of cardiovascular outcomes with continuous positive airway pressure therapy in patients with obstructive sleep apnea
.
Am J Cardiol
 
2017
;
120
:
693
9
.

229

Yu
 
J
,
Zhou
 
Z
,
McEvoy
 
RD
,
Anderson
 
CS
,
Rodgers
 
A
,
Perkovic
 
V
, et al.  
Association of positive airway pressure with cardiovascular events and death in adults with sleep apnea: a systematic review and meta-analysis
.
JAMA
 
2017
;
318
:
156
66
.

230

McEvoy
 
RD
,
Antic
 
NA
,
Heeley
 
E
,
Luo
 
Y
,
Ou
 
Q
,
Zhang
 
X
, et al.  
CPAP for prevention of cardiovascular events in obstructive sleep apnea
.
N Engl J Med
 
2016
;
375
:
919
31
.

231

Overvad
 
TF
,
Rasmussen
 
LH
,
Skjøth
 
F
,
Overvad
 
K
,
Albertsen
 
IE
,
Lane
 
DA
, et al.  
Alcohol intake and prognosis of atrial fibrillation
.
Heart
 
2013
;
99
:
1093
9
.

232

Lim
 
C
,
Kim
 
T-H
,
Yu
 
HT
,
Lee
 
S-R
,
Cha
 
M-J
,
Lee
 
J-M
, et al.  
Effect of alcohol consumption on the risk of adverse events in atrial fibrillation: from the COmparison study of Drugs for symptom control and complication prEvention of Atrial Fibrillation (CODE-AF) registry
.
EP Europace
 
2021
;
23
:
548
56
.

233

Lee
 
SR
,
Choi
 
EK
,
Jung
 
JH
,
Han
 
KD
,
Oh
 
S
,
Lip
 
GYH
.
Lower risk of stroke after alcohol abstinence in patients with incident atrial fibrillation: a nationwide population-based cohort study
.
Eur Heart J
 
2021
;
42
:
4759
68
.

234

Pisters
 
R
,
Lane
 
DA
,
Nieuwlaat
 
R
,
de Vos
 
CB
,
Crijns
 
HJ
,
Lip
 
GY
.
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
100
.

235

Takahashi
 
Y
,
Nitta
 
J
,
Kobori
 
A
,
Sakamoto
 
Y
,
Nagata
 
Y
,
Tanimoto
 
K
, et al.  
Alcohol consumption reduction and clinical outcomes of catheter ablation for atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2021
;
14
:
e009770
.

236

Friberg
 
L
,
Hammar
 
N
,
Rosenqvist
 
M
.
Stroke in paroxysmal atrial fibrillation: report from the Stockholm cohort of atrial fibrillation
.
Eur Heart J
 
2010
;
31
:
967
75
.

237

Banerjee
 
A
,
Taillandier
 
S
,
Olesen
 
JB
,
Lane
 
DA
,
Lallemand
 
B
,
Lip
 
GY
, et al.  
Pattern of atrial fibrillation and risk of outcomes: the Loire valley atrial fibrillation project
.
Int J Cardiol
 
2013
;
167
:
2682
7
.

238

Wolf
 
PA
,
Abbott
 
RD
,
Kannel
 
WB
.
Atrial fibrillation as an independent risk factor for stroke: the Framingham study
.
Stroke
 
1991
;
22
:
983
8
.

239

Hart
 
RG
,
Pearce
 
LA
,
Aguilar
 
MI
.
Meta-analysis: antithrombotic therapy to prevent stroke in patients who have nonvalvular atrial fibrillation
.
Ann Intern Med
 
2007
;
146
:
857
67
.

240

Ruff
 
CT
,
Giugliano
 
RP
,
Braunwald
 
E
,
Hoffman
 
EB
,
Deenadayalu
 
N
,
Ezekowitz
 
MD
, et al.  
Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials
.
Lancet
 
2014
;
383
:
955
62
.

241

Sjalander
 
S
,
Sjalander
 
A
,
Svensson
 
PJ
,
Friberg
 
L
.
Atrial fibrillation patients do not benefit from acetylsalicylic acid
.
Europace
 
2014
;
16
:
631
8
.

242

Connolly
 
SJ
,
Eikelboom
 
J
,
Joyner
 
C
,
Diener
 
HC
,
Hart
 
R
,
Golitsyn
 
S
, et al.  
Apixaban in patients with atrial fibrillation
.
N Engl J Med
 
2011
;
364
:
806
17
.

243

van Doorn
 
S
,
Rutten
 
FH
,
O’Flynn
 
CM
,
Oudega
 
R
,
Hoes
 
AW
,
Moons
 
KGM
, et al.  
Effectiveness of CHA2DS2-VASc based decision support on stroke prevention in atrial fibrillation: a cluster randomised trial in general practice
.
Int J Cardiol
 
2018
;
273
:
123
9
.

244

Borre
 
ED
,
Goode
 
A
,
Raitz
 
G
,
Shah
 
B
,
Lowenstern
 
A
,
Chatterjee
 
R
, et al.  
Predicting thromboembolic and bleeding event risk in patients with non-valvular atrial fibrillation: a systematic review
.
Thromb Haemost
 
2018
;
118
:
2171
87
.

245

van der Endt
 
VHW
,
Milders
 
J
,
de Vries
 
BBLP
,
Trines
 
SA
,
Groenwold
 
RHH
,
Dekkers
 
OM
, et al.  
Comprehensive comparison of stroke risk score performance: a systematic review and meta-analysis among 6 267 728 patients with atrial fibrillation
.
Europace
 
2022
;
24
:
1739
53
.

246

Quinn
 
GR
,
Severdija
 
ON
,
Chang
 
Y
,
Singer
 
DE
.
Wide variation in reported rates of stroke across cohorts of patients with atrial fibrillation
.
Circulation
 
2017
;
135
:
208
19
.

247

Pisters
 
R
,
Lane
 
DA
,
Marin
 
F
,
Camm
 
AJ
,
Lip
 
GY
.
Stroke and thromboembolism in atrial fibrillation
.
Circ J
 
2012
;
76
:
2289
304
.

248

Hohnloser
 
SH
,
Hijazi
 
Z
,
Thomas
 
L
,
Alexander
 
JH
,
Amerena
 
J
,
Hanna
 
M
, et al.  
Efficacy of apixaban when compared with warfarin in relation to renal function in patients with atrial fibrillation: insights from the ARISTOTLE trial
.
Eur Heart J
 
2012
;
33
:
2821
30
.

249

Fox
 
KA
,
Piccini
 
JP
,
Wojdyla
 
D
,
Becker
 
RC
,
Halperin
 
JL
,
Nessel
 
CC
, et al.  
Prevention of stroke and systemic embolism with rivaroxaban compared with warfarin in patients with non-valvular atrial fibrillation and moderate renal impairment
.
Eur Heart J
 
2011
;
32
:
2387
94
.

250

Yaghi
 
S
,
Henninger
 
N
,
Giles
 
JA
,
Leon Guerrero
 
C
,
Mistry
 
E
,
Liberman
 
AL
, et al.  
Ischaemic stroke on anticoagulation therapy and early recurrence in acute cardioembolic stroke: the IAC study
.
J Neurol Neurosurg Psychiatry
 
2021
;
92
:
1062
7
.

251

Ocak
 
G
,
Khairoun
 
M
,
Khairoun
 
O
,
Bos
 
WJW
,
Fu
 
EL
,
Cramer
 
MJ
, et al.  
Chronic kidney disease and atrial fibrillation: a dangerous combination
.
PLoS One
 
2022
;
17
:
e0266046
.

252

Seiffge
 
DJ
,
De Marchis
 
GM
,
Koga
 
M
,
Paciaroni
 
M
,
Wilson
 
D
,
Cappellari
 
M
, et al.  
Ischemic stroke despite oral anticoagulant therapy in patients with atrial fibrillation
.
Ann Neurol
 
2020
;
87
:
677
87
.

253

Paciaroni
 
M
,
Agnelli
 
G
,
Falocci
 
N
,
Caso
 
V
,
Becattini
 
C
,
Marcheselli
 
S
, et al.  
Prognostic value of trans-thoracic echocardiography in patients with acute stroke and atrial fibrillation: findings from the RAF study
.
J Neurol
 
2016
;
263
:
231
7
.

254

Hijazi
 
Z
,
Oldgren
 
J
,
Siegbahn
 
A
,
Wallentin
 
L
.
Application of biomarkers for risk stratification in patients with atrial fibrillation
.
Clin Chem
 
2017
;
63
:
152
64
.

255

Singleton
 
MJ
,
Yuan
 
Y
,
Dawood
 
FZ
,
Howard
 
G
,
Judd
 
SE
,
Zakai
 
NA
, et al.  
Multiple blood biomarkers and stroke risk in atrial fibrillation: the REGARDS study
.
J Am Heart Assoc
 
2021
;
10
:
e020157
.

256

Wu
 
VC
,
Wu
 
M
,
Aboyans
 
V
,
Chang
 
SH
,
Chen
 
SW
,
Chen
 
MC
, et al.  
Female sex as a risk factor for ischaemic stroke varies with age in patients with atrial fibrillation
.
Heart
 
2020
;
106
:
534
40
.

257

Mikkelsen
 
AP
,
Lindhardsen
 
J
,
Lip
 
GY
,
Gislason
 
GH
,
Torp-Pedersen
 
C
,
Olesen
 
JB
.
Female sex as a risk factor for stroke in atrial fibrillation: a nationwide cohort study
.
J Thromb Haemost
 
2012
;
10
:
1745
51
.

258

Antonenko
 
K
,
Paciaroni
 
M
,
Agnelli
 
G
,
Falocci
 
N
,
Becattini
 
C
,
Marcheselli
 
S
, et al.  
Sex-related differences in risk factors, type of treatment received and outcomes in patients with atrial fibrillation and acute stroke: results from the RAF study (early recurrence and cerebral bleeding in patients with acute ischemic stroke and atrial fibrillation)
.
Eur Stroke J
 
2017
;
2
:
46
53
.

259

Wang
 
X
,
Mobley
 
AR
,
Tica
 
O
,
Okoth
 
K
,
Ghosh
 
RE
,
Myles
 
P
, et al.  
Systematic approach to outcome assessment from coded electronic healthcare records in the DaRe2THINK NHS-embedded randomized trial
.
Eur Heart J - Dig Health
 
2022
;
3
:
426
36
.

260

Rivard
 
L
,
Khairy
 
P
,
Talajic
 
M
,
Tardif
 
JC
,
Nattel
 
S
,
Bherer
 
L
, et al.  
Blinded randomized trial of anticoagulation to prevent ischemic stroke and neurocognitive impairment in atrial fibrillation (BRAIN-AF): methods and design
.
Can J Cardiol
 
2019
;
35
:
1069
77
.

261

Chung
 
S
,
Kim
 
TH
,
Uhm
 
JS
,
Cha
 
MJ
,
Lee
 
JM
,
Park
 
J
, et al.  
Stroke and systemic embolism and other adverse outcomes of heart failure with preserved and reduced ejection fraction in patients with atrial fibrillation (from the COmparison study of Drugs for symptom control and complication prEvention of Atrial Fibrillation [CODE-AF])
.
Am J Cardiol
 
2020
;
125
:
68
75
.

262

Uhm
 
JS
,
Kim
 
J
,
Yu
 
HT
,
Kim
 
TH
,
Lee
 
SR
,
Cha
 
MJ
, et al.  
Stroke and systemic embolism in patients with atrial fibrillation and heart failure according to heart failure type
.
ESC Heart Fail
 
2021
;
8
:
1582
9
.

263

McMurray
 
JJ
,
Ezekowitz
 
JA
,
Lewis
 
BS
,
Gersh
 
BJ
,
van Diepen
 
S
,
Amerena
 
J
, et al.  
Left ventricular systolic dysfunction, heart failure, and the risk of stroke and systemic embolism in patients with atrial fibrillation: insights from the ARISTOTLE trial
.
Circ Heart Fail
 
2013
;
6
:
451
60
.

264

Kim
 
D
,
Yang
 
PS
,
Kim
 
TH
,
Jang
 
E
,
Shin
 
H
,
Kim
 
HY
, et al.  
Ideal blood pressure in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2018
;
72
:
1233
45
.

265

Lip
 
GY
,
Clementy
 
N
,
Pericart
 
L
,
Banerjee
 
A
,
Fauchier
 
L
.
Stroke and major bleeding risk in elderly patients aged ≥75 years with atrial fibrillation: the Loire valley atrial fibrillation project
.
Stroke
 
2015
;
46
:
143
50
.

266

American Diabetes Association Professional Practice Committee
.
2. Classification and diagnosis of diabetes: standards of medical care in diabetes—2022
.
Diabetes Care
 
2022
;
45
:
S17
38
.

267

Steensig
 
K
,
Olesen
 
KKW
,
Thim
 
T
,
Nielsen
 
JC
,
Jensen
 
SE
,
Jensen
 
LO
, et al.  
Should the presence or extent of coronary artery disease be quantified in the CHA2DS2-VASc score in atrial fibrillation? A report from the western Denmark heart registry
.
Thromb Haemost
 
2018
;
118
:
2162
70
.

268

Zabalgoitia
 
M
,
Halperin
 
JL
,
Pearce
 
LA
,
Blackshear
 
JL
,
Asinger
 
RW
,
Hart
 
RG
.
Transesophageal echocardiographic correlates of clinical risk of thromboembolism in nonvalvular atrial fibrillation. Stroke prevention in atrial fibrillation III investigators
.
J Am Coll Cardiol
 
1998
;
31
:
1622
6
.

269

Stroke Prevention in Atrial Fibrillation Investigators Committee on Echocardiography
.
Transesophageal echocardiography in atrial fibrillation: standards for acquisition and interpretation and assessment of interobserver variability. Stroke prevention in atrial fibrillation investigators committee on echocardiography
.
J Am Soc Echocardiogr
 
1996
;
9
:
556
66
.

270

Lozier
 
MR
,
Sanchez
 
AM
,
Lee
 
JJ
,
Donath
 
EM
,
Font
 
VE
,
Escolar
 
E
.
Thromboembolic outcomes of different anticoagulation strategies for patients with atrial fibrillation in the setting of hypertrophic cardiomyopathy: a systematic review
.
J Atr Fibrillation
 
2019
;
12
:
2207
.

271

Guttmann
 
OP
,
Rahman
 
MS
,
O’Mahony
 
C
,
Anastasakis
 
A
,
Elliott
 
PM
.
Atrial fibrillation and thromboembolism in patients with hypertrophic cardiomyopathy: systematic review
.
Heart
 
2014
;
100
:
465
72
.

272

Guttmann
 
OP
,
Pavlou
 
M
,
O’Mahony
 
C
,
Monserrat
 
L
,
Anastasakis
 
A
,
Rapezzi
 
C
, et al.  
Prediction of thromboembolic risk in patients with hypertrophic cardiomyopathy (HCM risk-CVA)
.
Eur J Heart Fail
 
2015
;
17
:
837
45
.

273

Vilches
 
S
,
Fontana
 
M
,
Gonzalez-Lopez
 
E
,
Mitrani
 
L
,
Saturi
 
G
,
Renju
 
M
, et al.  
Systemic embolism in amyloid transthyretin cardiomyopathy
.
Eur J Heart Fail
 
2022
;
24
:
1387
96
.

274

Lee
 
SE
,
Park
 
JK
,
Uhm
 
JS
,
Kim
 
JY
,
Pak
 
HN
,
Lee
 
MH
, et al.  
Impact of atrial fibrillation on the clinical course of apical hypertrophic cardiomyopathy
.
Heart
 
2017
;
103
:
1496
501
.

275

Hirota
 
T
,
Kubo
 
T
,
Baba
 
Y
,
Ochi
 
Y
,
Takahashi
 
A
,
Yamasaki
 
N
, et al.  
Clinical profile of thromboembolic events in patients with hypertrophic cardiomyopathy in a regional Japanese cohort–results from Kochi RYOMA study
.
Circ J
 
2019
;
83
:
1747
54
.

276

Hsu
 
JC
,
Huang
 
YT
,
Lin
 
LY
.
Stroke risk in hypertrophic cardiomyopathy patients with atrial fibrillation: a nationwide database study
.
Aging (Albany NY)
 
2020
;
12
:
24219
27
.

277

Chao
 
TF
,
Lip
 
GYH
,
Liu
 
CJ
,
Lin
 
YJ
,
Chang
 
SL
,
Lo
 
LW
, et al.  
Relationship of aging and incident comorbidities to stroke risk in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2018
;
71
:
122
32
.

278

Weijs
 
B
,
Dudink
 
E
,
de Vos
 
CB
,
Limantoro
 
I
,
Tieleman
 
RG
,
Pisters
 
R
, et al.  
Idiopathic atrial fibrillation patients rapidly outgrow their low thromboembolic risk: a 10-year follow-up study
.
Neth Heart J
 
2019
;
27
:
487
97
.

279

Bezabhe
 
WM
,
Bereznicki
 
LR
,
Radford
 
J
,
Wimmer
 
BC
,
Salahudeen
 
MS
,
Garrahy
 
E
, et al.  
Stroke risk reassessment and oral anticoagulant initiation in primary care patients with atrial fibrillation: A ten-year follow-up
.
Eur J Clin Invest
 
2021
;
51
:
e13489
.

280

Fauchier
 
L
,
Bodin
 
A
,
Bisson
 
A
,
Herbert
 
J
,
Spiesser
 
P
,
Clementy
 
N
, et al.  
Incident comorbidities, aging and the risk of stroke in 608,108 patients with atrial fibrillation: a nationwide analysis
.
J Clin Med
 
2020
;
9
:
1234
.

281

Kirchhof
 
P
,
Toennis
 
T
,
Goette
 
A
,
Camm
 
AJ
,
Diener
 
HC
,
Becher
 
N
, et al.  
Anticoagulation with edoxaban in patients with atrial high-rate episodes
.
N Engl J Med
 
2023
;
389
:
1167
79
.

282

Healey
 
JS
,
Lopes
 
RD
,
Granger
 
CB
,
Alings
 
M
,
Rivard
 
L
,
McIntyre
 
WF
, et al.  
Apixaban for stroke prevention in subclinical atrial fibrillation
.
N Engl J Med
 
2024
;
390
:
107
17
.

283

van Walraven
 
C
,
Hart
 
RG
,
Singer
 
DE
,
Laupacis
 
A
,
Connolly
 
S
,
Petersen
 
P
, et al.  
Oral anticoagulants vs aspirin in nonvalvular atrial fibrillation: an individual patient meta-analysis
.
JAMA
 
2002
;
288
:
2441
8
.

284

Hart
 
RG
,
Pearce
 
LA
,
Rothbart
 
RM
,
McAnulty
 
JH
,
Asinger
 
RW
,
Halperin
 
JL
.
Stroke with intermittent atrial fibrillation: incidence and predictors during aspirin therapy. Stroke prevention in atrial fibrillation investigators
.
J Am Coll Cardiol
 
2000
;
35
:
183
7
.

285

Nieuwlaat
 
R
,
Dinh
 
T
,
Olsson
 
SB
,
Camm
 
AJ
,
Capucci
 
A
,
Tieleman
 
RG
, et al.  
Should we abandon the common practice of withholding oral anticoagulation in paroxysmal atrial fibrillation?
 
Eur Heart J
 
2008
;
29
:
915
22
.

286

Ruff CT. AZALEA-TIMI 71 Steering Committee. Abelacimab, a novel factor XI/XIa inhibitor, vs rivaroxaban in patients with atrial fibrillation: primary results of the AZALEA-TIMI 71 randomized trial. Circulation 2024;148:e282–317.

287

Piccini
 
JP
,
Caso
 
V
,
Connolly
 
SJ
,
Fox
 
KAA
,
Oldgren
 
J
,
Jones
 
WS
, et al.  
Safety of the oral factor XIa inhibitor asundexian compared with apixaban in patients with atrial fibrillation (PACIFIC-AF): a multicentre, randomised, double-blind, double-dummy, dose-finding phase 2 study
.
Lancet
 
2022
;
399
:
1383
90
.

288

Tan
 
CSS
,
Lee
 
SWH
.
Warfarin and food, herbal or dietary supplement interactions: a systematic review
.
Br J Clin Pharmacol
 
2021
;
87
:
352
74
.

289

Holbrook
 
AM
,
Pereira
 
JA
,
Labiris
 
R
,
McDonald
 
H
,
Douketis
 
JD
,
Crowther
 
M
, et al.  
Systematic overview of warfarin and its drug and food interactions
.
Arch Intern Med
 
2005
;
165
:
1095
106
.

290

Ferri
 
N
,
Colombo
 
E
,
Tenconi
 
M
,
Baldessin
 
L
,
Corsini
 
A
.
Drug-drug interactions of direct oral anticoagulants (DOACs): from pharmacological to clinical practice
.
Pharmaceutics
 
2022
;
14
:
1120
.

291

Mar
 
PL
,
Gopinathannair
 
R
,
Gengler
 
BE
,
Chung
 
MK
,
Perez
 
A
,
Dukes
 
J
, et al.  
Drug interactions affecting oral anticoagulant use
.
Circ Arrhythm Electrophysiol
 
2022
;
15
:
e007956
.

292

Carnicelli
 
AP
,
Hong
 
H
,
Connolly
 
SJ
,
Eikelboom
 
J
,
Giugliano
 
RP
,
Morrow
 
DA
, et al.  
Direct oral anticoagulants versus warfarin in patients with atrial fibrillation: patient-level network meta-analyses of randomized clinical trials with interaction testing by age and sex
.
Circulation
 
2022
;
145
:
242
55
.

293

Kotecha
 
D
,
Pollack
 
CV
, Jr
,
De Caterina
 
R
,
Renda
 
G
,
Kirchhof
 
P
.
Direct oral anticoagulants halve thromboembolic events after cardioversion of AF compared with warfarin
.
J Am Coll Cardiol
 
2018
;
72
:
1984
6
.

294

Connolly
 
SJ
,
Karthikeyan
 
G
,
Ntsekhe
 
M
,
Haileamlak
 
A
,
El Sayed
 
A
,
El Ghamrawy
 
A
, et al.  
Rivaroxaban in rheumatic heart disease-associated atrial fibrillation
.
N Engl J Med
 
2022
;
387
:
978
88
.

295

Halperin JL, Hart RG, Kronmal RA, McBride R.

Warfarin versus aspirin for prevention of thromboembolism in atrial fibrillation: stroke prevention in atrial fibrillation II study
.
Lancet
 
1994
;
343
:
687
91
.

296

Singer
 
DE
,
Hughes
 
RA
,
Gress
 
DR
,
Sheehan
 
MA
,
Oertel
 
LB
,
Maraventano
 
SW
, et al.  
The effect of low-dose warfarin on the risk of stroke in patients with nonrheumatic atrial fibrillation
.
N Engl J Med
 
1990
;
323
:
1505
11
.

297

Gulløv
 
AL
,
Koefoed
 
BG
,
Petersen
 
P
,
Pedersen
 
TS
,
Andersen
 
ED
,
Godtfredsen
 
J
, et al.  
Fixed minidose warfarin and aspirin alone and in combination vs adjusted-dose warfarin for stroke prevention in atrial fibrillation: second Copenhagen atrial fibrillation, aspirin, and anticoagulation study
.
Arch Intern Med
 
1998
;
158
:
1513
21
.

298

Blackshear JL, Halperin JL, Hart RG, Laupacis A.

Adjusted-dose warfarin versus low-intensity, fixed-dose warfarin plus aspirin for high-risk patients with atrial fibrillation: stroke prevention in atrial fibrillation III randomised clinical trial
.
Lancet
 
1996
;
348
:
633
8
.

299

Amin
 
A
,
Deitelzweig
 
S
,
Jing
 
Y
,
Makenbaeva
 
D
,
Wiederkehr
 
D
,
Lin
 
J
, et al.  
Estimation of the impact of warfarin’s time-in-therapeutic range on stroke and major bleeding rates and its influence on the medical cost avoidance associated with novel oral anticoagulant use–learnings from ARISTOTLE, ROCKET-AF, and RE-LY trials
.
J Thromb Thrombolysis
 
2014
;
38
:
150
9
.

300

Själander
 
S
,
Sjögren
 
V
,
Renlund
 
H
,
Norrving
 
B
,
Själander
 
A
.
Dabigatran, rivaroxaban and apixaban vs. high TTR warfarin in atrial fibrillation
.
Thromb Res
 
2018
;
167
:
113
8
.

301

van Miert
 
JHA
,
Kooistra
 
HAM
,
Veeger
 
N
,
Westerterp
 
A
,
Piersma-Wichers
 
M
,
Meijer
 
K
.
Choosing between continuing vitamin K antagonists (VKA) or switching to a direct oral anticoagulant in currently well-controlled patients on VKA for atrial fibrillation: a randomised controlled trial (GAInN)
.
Br J Haematol
 
2019
;
186
:
e21
3
.

302

Krittayaphong
 
R
,
Chantrarat
 
T
,
Rojjarekampai
 
R
,
Jittham
 
P
,
Sairat
 
P
,
Lip
 
GYH
.
Poor time in therapeutic range control is associated with adverse clinical outcomes in patients with non-valvular atrial fibrillation: a report from the nationwide COOL-AF registry
.
J Clin Med
 
2020
;
9
:
1698
.

303

Szummer
 
K
,
Gasparini
 
A
,
Eliasson
 
S
,
Ärnlöv
 
J
,
Qureshi
 
AR
,
Bárány
 
P
, et al.  
Time in therapeutic range and outcomes after warfarin initiation in newly diagnosed atrial fibrillation patients with renal dysfunction
.
J Am Heart Assoc
 
2017
;
6
:
e004925
.

304

Cardoso
 
R
,
Ternes
 
CMP
,
Justino
 
GB
,
Fernandes
 
A
,
Rocha
 
AV
,
Knijnik
 
L
, et al.  
Non-vitamin K antagonists versus warfarin in patients with atrial fibrillation and bioprosthetic valves: a systematic review and meta-analysis
.
Am J Med
 
2022
;
135
:
228
234.e1
.

305

Wan
 
Y
,
Heneghan
 
C
,
Perera
 
R
,
Roberts
 
N
,
Hollowell
 
J
,
Glasziou
 
P
, et al.  
Anticoagulation control and prediction of adverse events in patients with atrial fibrillation: a systematic review
.
Circ Cardiovasc Qual Outcomes
 
2008
;
1
:
84
91
.

306

Vestergaard
 
AS
,
Skjøth
 
F
,
Larsen
 
TB
,
Ehlers
 
LH
.
The importance of mean time in therapeutic range for complication rates in warfarin therapy of patients with atrial fibrillation: a systematic review and meta-regression analysis
.
PLoS One
 
2017
;
12
:
e0188482
.

307

Macaluso
 
GP
,
Pagani
 
FD
,
Slaughter
 
MS
,
Milano
 
CA
,
Feller
 
ED
,
Tatooles
 
AJ
, et al.  
Time in therapeutic range significantly impacts survival and adverse events in destination therapy patients
.
ASAIO J
 
2022
;
68
:
14
20
.

308

Heneghan
 
C
,
Ward
 
A
,
Perera
 
R
,
Bankhead
 
C
,
Fuller
 
A
,
Stevens
 
R
, et al.  
Self-monitoring of oral anticoagulation: systematic review and meta-analysis of individual patient data
.
Lancet
 
2012
;
379
:
322
34
.

309

Joosten
 
LPT
,
van Doorn
 
S
,
van de Ven
 
PM
,
Köhlen
 
BTG
,
Nierman
 
MC
,
Koek
 
HL
, et al.  
Safety of switching from a vitamin K antagonist to a non-vitamin K antagonist oral anticoagulant in frail older patients with atrial fibrillation: results of the FRAIL-AF randomized controlled trial
.
Circulation
 
2024
;
149
:
279
89
.

310

Yao
 
X
,
Shah
 
ND
,
Sangaralingham
 
LR
,
Gersh
 
BJ
,
Noseworthy
 
PA
.
Non-vitamin K antagonist oral anticoagulant dosing in patients with atrial fibrillation and renal dysfunction
.
J Am Coll Cardiol
 
2017
;
69
:
2779
90
.

311

Steinberg
 
BA
,
Shrader
 
P
,
Thomas
 
L
,
Ansell
 
J
,
Fonarow
 
GC
,
Gersh
 
BJ
, et al.  
Off-label dosing of non-vitamin K antagonist oral anticoagulants and adverse outcomes: the ORBIT-AF II registry
.
J Am Coll Cardiol
 
2016
;
68
:
2597
604
.

312

Alexander
 
JH
,
Andersson
 
U
,
Lopes
 
RD
,
Hijazi
 
Z
,
Hohnloser
 
SH
,
Ezekowitz
 
JA
, et al.  
Apixaban 5 mg twice daily and clinical outcomes in patients with atrial fibrillation and advanced age, low body weight, or high creatinine: a secondary analysis of a randomized clinical trial
.
JAMA Cardiol
 
2016
;
1
:
673
81
.

313

Carmo
 
J
,
Moscoso Costa
 
F
,
Ferreira
 
J
,
Mendes
 
M
.
Dabigatran in real-world atrial fibrillation. Meta-analysis of observational comparison studies with vitamin K antagonists
.
Thromb Haemost
 
2016
;
116
:
754
63
.

314

Huisman
 
MV
,
Rothman
 
KJ
,
Paquette
 
M
,
Teutsch
 
C
,
Diener
 
HC
,
Dubner
 
SJ
, et al.  
Two-year follow-up of patients treated with dabigatran for stroke prevention in atrial fibrillation: global registry on long-term antithrombotic treatment in patients with atrial fibrillation (GLORIA-AF) registry
.
Am Heart J
 
2018
;
198
:
55
63
.

315

Camm
 
AJ
,
Amarenco
 
P
,
Haas
 
S
,
Hess
 
S
,
Kirchhof
 
P
,
Kuhls
 
S
, et al.  
XANTUS: a real-world, prospective, observational study of patients treated with rivaroxaban for stroke prevention in atrial fibrillation
.
Eur Heart J
 
2016
;
37
:
1145
53
.

316

Martinez
 
CAA
,
Lanas
 
F
,
Radaideh
 
G
,
Kharabsheh
 
SM
,
Lambelet
 
M
,
Viaud
 
MAL
, et al.  
XANTUS-EL: a real-world, prospective, observational study of patients treated with rivaroxaban for stroke prevention in atrial fibrillation in Eastern Europe, Middle East, Africa and Latin America
.
Egypt Heart J
 
2018
;
70
:
307
13
.

317

Li
 
XS
,
Deitelzweig
 
S
,
Keshishian
 
A
,
Hamilton
 
M
,
Horblyuk
 
R
,
Gupta
 
K
, et al.  
Effectiveness and safety of apixaban versus warfarin in non-valvular atrial fibrillation patients in “real-world” clinical practice. A propensity-matched analysis of 76,940 patients
.
Thromb Haemost
 
2017
;
117
:
1072
82
.

318

Lee
 
SR
,
Choi
 
EK
,
Han
 
KD
,
Jung
 
JH
,
Oh
 
S
,
Lip
 
GYH
.
Edoxaban in Asian patients with atrial fibrillation: effectiveness and safety
.
J Am Coll Cardiol
 
2018
;
72
:
838
53
.

319

Cappato
 
R
,
Ezekowitz
 
MD
,
Klein
 
AL
,
Camm
 
AJ
,
Ma
 
CS
,
Le Heuzey
 
JY
, et al.  
Rivaroxaban vs. vitamin K antagonists for cardioversion in atrial fibrillation
.
Eur Heart J
 
2014
;
35
:
3346
55
.

320

Goette
 
A
,
Merino
 
JL
,
Ezekowitz
 
MD
,
Zamoryakhin
 
D
,
Melino
 
M
,
Jin
 
J
, et al.  
Edoxaban versus enoxaparin-warfarin in patients undergoing cardioversion of atrial fibrillation (ENSURE-AF): a randomised, open-label, phase 3b trial
.
Lancet
 
2016
;
388
:
1995
2003
.

321

Ezekowitz
 
MD
,
Pollack
 
CV
, Jr
,
Halperin
 
JL
,
England
 
RD
,
VanPelt Nguyen
 
S
,
Spahr
 
J
, et al.  
Apixaban compared to heparin/vitamin K antagonist in patients with atrial fibrillation scheduled for cardioversion: the EMANATE trial
.
Eur Heart J
 
2018
;
39
:
2959
71
.

322

Savarese
 
G
,
Giugliano
 
RP
,
Rosano
 
GM
,
McMurray
 
J
,
Magnani
 
G
,
Filippatos
 
G
, et al.  
Efficacy and safety of novel oral anticoagulants in patients with atrial fibrillation and heart failure: a meta-analysis
.
JACC Heart Fail
 
2016
;
4
:
870
80
.

323

von Lueder
 
TG
,
Atar
 
D
,
Agewall
 
S
,
Jensen
 
JK
,
Hopper
 
I
,
Kotecha
 
D
, et al.  
All-cause mortality and cardiovascular outcomes with non-vitamin K oral anticoagulants versus warfarin in patients with heart failure in the food and drug administration adverse event reporting system
.
Am J Ther
 
2019
;
26
:
e671
8
.

324

Harrison
 
SL
,
Buckley
 
BJR
,
Ritchie
 
LA
,
Proietti
 
R
,
Underhill
 
P
,
Lane
 
DA
, et al.  
Oral anticoagulants and outcomes in adults >/=80 years with atrial fibrillation: a global federated health network analysis
.
J Am Geriatr Soc
 
2022
;
70
:
2386
92
.

325

Malhotra
 
K
,
Ishfaq
 
MF
,
Goyal
 
N
,
Katsanos
 
AH
,
Parissis
 
J
,
Alexandrov
 
AW
, et al.  
Oral anticoagulation in patients with chronic kidney disease: a systematic review and meta-analysis
.
Neurology
 
2019
;
92
:
e2421
31
.

326

Steffel
 
J
,
Verhamme
 
P
,
Potpara
 
TS
,
Albaladejo
 
P
,
Antz
 
M
,
Desteghe
 
L
, et al.  
The 2018 European heart rhythm association practical guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation
.
Eur Heart J
 
2018
;
39
:
1330
93
.

327

Rhee
 
TM
,
Lee
 
SR
,
Choi
 
EK
,
Oh
 
S
,
Lip
 
GYH
.
Efficacy and safety of oral anticoagulants for atrial fibrillation patients with chronic kidney disease: a systematic review and meta-analysis
.
Front Cardiovasc Med
 
2022
;
9
:
885548
.

328

Reinecke
 
H
,
Engelbertz
 
C
,
Bauersachs
 
R
,
Breithardt
 
G
,
Echterhoff
 
HH
,
Gerß
 
J
, et al.  
A randomized controlled trial comparing apixaban with the vitamin K antagonist phenprocoumon in patients on chronic hemodialysis: the AXADIA-AFNET 8 study
.
Circulation
 
2023
;
147
:
296
309
.

329

Pokorney
 
SD
,
Chertow
 
GM
,
Al-Khalidi
 
HR
,
Gallup
 
D
,
Dignacco
 
P
,
Mussina
 
K
, et al.  
Apixaban for patients with atrial fibrillation on hemodialysis: a multicenter randomized controlled trial
.
Circulation
 
2022
;
146
:
1735
45
.

330

De Vriese
 
AS
,
Caluwé
 
R
,
Van Der Meersch
 
H
,
De Boeck
 
K
,
De Bacquer
 
D
.
Safety and efficacy of vitamin K antagonists versus rivaroxaban in hemodialysis patients with atrial fibrillation: a multicenter randomized controlled trial
.
J Am Soc Nephrol
 
2021
;
32
:
1474
83
.

331

Eikelboom
 
JW
,
Connolly
 
SJ
,
Brueckmann
 
M
,
Granger
 
CB
,
Kappetein
 
AP
,
Mack
 
MJ
, et al.  
Dabigatran versus warfarin in patients with mechanical heart valves
.
N Engl J Med
 
2013
;
369
:
1206
14
.

332

Wang
 
TY
,
Svensson
 
LG
,
Wen
 
J
,
Vekstein
 
A
,
Gerdisch
 
M
,
Rao
 
VU
, et al.  
Apixaban or warfarin in patients with an on-X mechanical aortic valve
.
NEJM Evid
 
2023
;
2
:EVIDoa2300067.

333

Guimarães
 
HP
,
Lopes
 
RD
,
de Barros
 
ESPGM
,
Liporace
 
IL
,
Sampaio
 
RO
,
Tarasoutchi
 
F
, et al.  
Rivaroxaban in patients with atrial fibrillation and a bioprosthetic mitral valve
.
N Engl J Med
 
2020
;
383
:
2117
26
.

334

Collet
 
JP
,
Van Belle
 
E
,
Thiele
 
H
,
Berti
 
S
,
Lhermusier
 
T
,
Manigold
 
T
, et al.  
Apixaban vs. standard of care after transcatheter aortic valve implantation: the ATLANTIS trial
.
Eur Heart J
 
2022
;
43
:
2783
97
.

335

Steffel
 
J
,
Collins
 
R
,
Antz
 
M
,
Cornu
 
P
,
Desteghe
 
L
,
Haeusler
 
KG
, et al.  
2021 European heart rhythm association practical guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation
.
Europace
 
2021
;
23
:
1612
76
.

336

Grymonprez
 
M
,
Carnoy
 
L
,
Capiau
 
A
,
Boussery
 
K
,
Mehuys
 
E
,
De Backer
 
TL
, et al.  
Impact of P-glycoprotein and CYP3A4-interacting drugs on clinical outcomes in patients with atrial fibrillation using non-vitamin K antagonist oral anticoagulants: a nationwide cohort study
.
Eur Heart J Cardiovasc Pharmacother
 
2023
;
9
:
722
30
.

337

Testa
 
S
,
Legnani
 
C
,
Antonucci
 
E
,
Paoletti
 
O
,
Dellanoce
 
C
,
Cosmi
 
B
, et al.  
Drug levels and bleeding complications in atrial fibrillation patients treated with direct oral anticoagulants
.
J Thromb Haemost
 
2019
;
17
:
1064
72
.

338

Suwa
 
M
,
Nohara
 
Y
,
Morii
 
I
,
Kino
 
M
.
Safety and efficacy re-evaluation of edoxaban and rivaroxaban dosing with plasma concentration monitoring in non-valvular atrial fibrillation: with observations of on-label and off-label dosing
.
Circ Rep
 
2023
;
5
:
80
9
.

339

Song
 
D
,
Zhou
 
J
,
Fan
 
T
,
Chang
 
J
,
Qiu
 
Y
,
Zhuang
 
Z
, et al.  
Decision aids for shared decision-making and appropriate anticoagulation therapy in patients with atrial fibrillation: a systematic review and meta-analysis
.
Eur J Cardiovasc Nurs
 
2022
;
21
:
97
106
.

340

Vora
 
P
,
Morgan Stewart
 
H
,
Russell
 
B
,
Asiimwe
 
A
,
Brobert
 
G
.
Time trends and treatment pathways in prescribing individual oral anticoagulants in patients with nonvalvular atrial fibrillation: an observational study of more than three million patients from Europe and the United States
.
Int J Clin Pract
 
2022
;
2022
:
6707985
.

341

Grymonprez
 
M
,
Simoens
 
C
,
Steurbaut
 
S
,
De Backer
 
TL
,
Lahousse
 
L
.
Worldwide trends in oral anticoagulant use in patients with atrial fibrillation from 2010 to 2018: a systematic review and meta-analysis
.
Europace
 
2022
;
24
:
887
98
.

342

De Caterina
 
R
,
Husted
 
S
,
Wallentin
 
L
,
Andreotti
 
F
,
Arnesen
 
H
,
Bachmann
 
F
, et al.  
Vitamin K antagonists in heart disease: current status and perspectives (section III). Position paper of the ESC working group on thrombosis—task force on anticoagulants in heart disease
.
Thromb Haemost
 
2013
;
110
:
1087
107
.

343

Pandey
 
AK
,
Xu
 
K
,
Zhang
 
L
,
Gupta
 
S
,
Eikelboom
 
J
,
Cook
 
O
, et al.  
Lower versus standard INR targets in atrial fibrillation: a systematic review and meta-analysis of randomized controlled trials
.
Thromb Haemost
 
2020
;
120
:
484
94
.

344

Sanders P, Svennberg E, Diederichsen SZ, Crijns HJGM, Lambiase PD, Boriani G, et al. Great debate: device-detected subclinical atrial fibrillation should be treated like clinical atrial fibrillation. Eur Heart J  

2024
;
45
:
2594
603
.

345

ACTIVE Investigators
;
Connolly
 
SJ
,
Pogue
 
J
,
Hart
 
RG
,
Hohnloser
 
SH
,
Pfeffer
 
M
et al.  et al.  
Effect of clopidogrel added to aspirin in patients with atrial fibrillation
.
N Engl J Med
 
2009
;
360
:
2066
78
.

346

Mant
 
J
,
Hobbs
 
FD
,
Fletcher
 
K
,
Roalfe
 
A
,
Fitzmaurice
 
D
,
Lip
 
GY
, et al.  
Warfarin versus aspirin for stroke prevention in an elderly community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised controlled trial
.
Lancet
 
2007
;
370
:
493
503
.

347

Lip
 
GY
.
The role of aspirin for stroke prevention in atrial fibrillation
.
Nat Rev Cardiol
 
2011
;
8
:
602
6
.

348

ACTIVE Writing Group of the ACTIVE Investigators
;
Connolly
 
S
,
Pogue
 
J
,
Hart
 
R
,
Pfeffer
 
M
,
Hohnloser
 
S
, et al.  
Clopidogrel plus aspirin versus oral anticoagulation for atrial fibrillation in the Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events (ACTIVE W): a randomised controlled trial
.
Lancet
 
2006
;
367
:
1903
12
.

349

Fox
 
KAA
,
Velentgas
 
P
,
Camm
 
AJ
,
Bassand
 
JP
,
Fitzmaurice
 
DA
,
Gersh
 
BJ
, et al.  
Outcomes associated with oral anticoagulants plus antiplatelets in patients with newly diagnosed atrial fibrillation
.
JAMA Netw Open
 
2020
;
3
:
e200107
.

350

Verheugt
 
FWA
,
Gao
 
H
,
Al Mahmeed
 
W
,
Ambrosio
 
G
,
Angchaisuksiri
 
P
,
Atar
 
D
, et al.  
Characteristics of patients with atrial fibrillation prescribed antiplatelet monotherapy compared with those on anticoagulants: insights from the GARFIELD-AF registry
.
Eur Heart J
 
2018
;
39
:
464
73
.

351

Steffel
 
J
,
Eikelboom
 
JW
,
Anand
 
SS
,
Shestakovska
 
O
,
Yusuf
 
S
,
Fox
 
KAA
.
The COMPASS trial: net clinical benefit of low-dose rivaroxaban plus aspirin as compared with aspirin in patients with chronic vascular disease
.
Circulation
 
2020
;
142
:
40
8
.

352

Sharma
 
M
,
Hart
 
RG
,
Connolly
 
SJ
,
Bosch
 
J
,
Shestakovska
 
O
,
Ng
 
KKH
, et al.  
Stroke outcomes in the COMPASS trial
.
Circulation
 
2019
;
139
:
1134
45
.

353

Yasuda
 
S
,
Kaikita
 
K
,
Akao
 
M
,
Ako
 
J
,
Matoba
 
T
,
Nakamura
 
M
, et al.  
Antithrombotic therapy for atrial fibrillation with stable coronary disease
.
N Engl J Med
 
2019
;
381
:
1103
13
.

354

Senoo
 
K
,
Lip
 
GY
,
Lane
 
DA
,
Büller
 
HR
,
Kotecha
 
D
.
Residual risk of stroke and death in anticoagulated patients according to the type of atrial fibrillation: AMADEUS trial
.
Stroke
 
2015
;
46
:
2523
8
.

355

Meinel
 
TR
,
Branca
 
M
,
De Marchis
 
GM
,
Nedeltchev
 
K
,
Kahles
 
T
,
Bonati
 
L
, et al.  
Prior anticoagulation in patients with ischemic stroke and atrial fibrillation
.
Ann Neurol
 
2021
;
89
:
42
53
.

356

Polymeris
 
AA
,
Meinel
 
TR
,
Oehler
 
H
,
Hölscher
 
K
,
Zietz
 
A
,
Scheitz
 
JF
, et al.  
Aetiology, secondary prevention strategies and outcomes of ischaemic stroke despite oral anticoagulant therapy in patients with atrial fibrillation
.
J Neurol Neurosurg Psychiatry
 
2022
;
93
:
588
98
.

357

Paciaroni
 
M
,
Agnelli
 
G
,
Caso
 
V
,
Silvestrelli
 
G
,
Seiffge
 
DJ
,
Engelter
 
S
, et al.  
Causes and risk factors of cerebral ischemic events in patients with atrial fibrillation treated with non-vitamin K antagonist oral anticoagulants for stroke prevention
.
Stroke
 
2019
;
50
:
2168
74
.

358

Purrucker
 
JC
,
Hölscher
 
K
,
Kollmer
 
J
,
Ringleb
 
PA
.
Etiology of ischemic strokes of patients with atrial fibrillation and therapy with anticoagulants
.
J Clin Med
 
2020
;
9
:
2938
.

359

Paciaroni
 
M
,
Caso
 
V
,
Agnelli
 
G
,
Mosconi
 
MG
,
Giustozzi
 
M
,
Seiffge
 
DJ
, et al.  
Recurrent ischemic stroke and bleeding in patients with atrial fibrillation who suffered an acute stroke while on treatment with nonvitamin K antagonist oral anticoagulants: the RENO-EXTEND study
.
Stroke
 
2022
;
53
:
2620
7
.

360

Smits
 
E
,
Andreotti
 
F
,
Houben
 
E
,
Crijns
 
H
,
Haas
 
S
,
Spentzouris
 
G
, et al.  
Adherence and persistence with once-daily vs twice-daily direct oral anticoagulants among patients with atrial fibrillation: real-world analyses from The Netherlands, Italy and Germany
.
Drugs Real World Outcomes
 
2022
;
9
:
199
209
.

361

Polymeris
 
AA
,
Zietz
 
A
,
Schaub
 
F
,
Meya
 
L
,
Traenka
 
C
,
Thilemann
 
S
, et al.  
Once versus twice daily direct oral anticoagulants in patients with recent stroke and atrial fibrillation
.
Eur Stroke J
 
2022
;
7
:
221
9
.

362

Glikson
 
M
,
Wolff
 
R
,
Hindricks
 
G
,
Mandrola
 
J
,
Camm
 
AJ
,
Lip
 
GYH
, et al.  
EHRA/EAPCI expert consensus statement on catheter-based left atrial appendage occlusion—an update
.
Europace
 
2020
;
22
:
184
.

363

Blackshear
 
JL
,
Odell
 
JA
.
Appendage obliteration to reduce stroke in cardiac surgical patients with atrial fibrillation
.
Ann Thorac Surg
 
1996
;
61
:
755
9
.

364

Reddy
 
VY
,
Doshi
 
SK
,
Kar
 
S
,
Gibson
 
DN
,
Price
 
MJ
,
Huber
 
K
, et al.  
5-Year outcomes after left atrial appendage closure: from the PREVAIL and PROTECT AF trials
.
J Am Coll Cardiol
 
2017
;
70
:
2964
75
.

365

Lakkireddy
 
D
,
Thaler
 
D
,
Ellis
 
CR
,
Swarup
 
V
,
Sondergaard
 
L
,
Carroll
 
J
, et al.  
Amplatzer amulet left atrial appendage occluder versus watchman device for stroke prophylaxis (amulet IDE): a randomized, controlled trial
.
Circulation
 
2021
;
144
:
1543
52
.

366

Osmancik
 
P
,
Herman
 
D
,
Neuzil
 
P
,
Hala
 
P
,
Taborsky
 
M
,
Kala
 
P
, et al.  
Left atrial appendage closure versus direct oral anticoagulants in high-risk patients with atrial fibrillation
.
J Am Coll Cardiol
 
2020
;
75
:
3122
35
.

367

Osmancik
 
P
,
Herman
 
D
,
Neuzil
 
P
,
Hala
 
P
,
Taborsky
 
M
,
Kala
 
P
, et al.  
4-Year outcomes after left atrial appendage closure versus nonwarfarin oral anticoagulation for atrial fibrillation
.
J Am Coll Cardiol
 
2022
;
79
:
1
14
.

368

Korsholm
 
K
,
Damgaard
 
D
,
Valentin
 
JB
,
Packer
 
EJS
,
Odenstedt
 
J
,
Sinisalo
 
J
, et al.  
Left atrial appendage occlusion vs novel oral anticoagulation for stroke prevention in atrial fibrillation: rationale and design of the multicenter randomized occlusion-AF trial
.
Am Heart J
 
2022
;
243
:
28
38
.

369

Huijboom
 
M
,
Maarse
 
M
,
Aarnink
 
E
,
van Dijk
 
V
,
Swaans
 
M
,
van der Heijden
 
J
, et al.  
COMPARE LAAO: rationale and design of the randomized controlled trial “COMPARing Effectiveness and safety of Left Atrial Appendage Occlusion to standard of care for atrial fibrillation patients at high stroke risk and ineligible to use oral anticoagulation therapy”
.
Am Heart J
 
2022
;
250
:
45
56
.

370

Freeman
 
JV
,
Higgins
 
AY
,
Wang
 
Y
,
Du
 
C
,
Friedman
 
DJ
,
Daimee
 
UA
, et al.  
Antithrombotic therapy after left atrial appendage occlusion in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2022
;
79
:
1785
98
.

371

Patti
 
G
,
Sticchi
 
A
,
Verolino
 
G
,
Pasceri
 
V
,
Vizzi
 
V
,
Brscic
 
E
, et al.  
Safety and efficacy of single versus dual antiplatelet therapy after left atrial appendage occlusion
.
Am J Cardiol
 
2020
;
134
:
83
90
.

372

Boersma
 
LV
,
Schmidt
 
B
,
Betts
 
TR
,
Sievert
 
H
,
Tamburino
 
C
,
Teiger
 
E
, et al.  
Implant success and safety of left atrial appendage closure with the WATCHMAN device: peri-procedural outcomes from the EWOLUTION registry
.
Eur Heart J
 
2016
;
37
:
2465
74
.

373

Garg
 
J
,
Shah
 
S
,
Shah
 
K
,
Turagam
 
MK
,
Tzou
 
W
,
Natale
 
A
, et al.  
Direct oral anticoagulant versus warfarin for watchman left atrial appendage occlusion—systematic review
.
JACC Clin Electrophysiol
 
2020
;
6
:
1735
7
.

374

Osman
 
M
,
Busu
 
T
,
Osman
 
K
,
Khan
 
SU
,
Daniels
 
M
,
Holmes
 
DR
, et al.  
Short-term antiplatelet versus anticoagulant therapy after left atrial appendage occlusion: a systematic review and meta-analysis
.
JACC Clin Electrophysiol
 
2020
;
6
:
494
506
.

375

Hildick-Smith
 
D
,
Landmesser
 
U
,
Camm
 
AJ
,
Diener
 
HC
,
Paul
 
V
,
Schmidt
 
B
, et al.  
Left atrial appendage occlusion with the Amplatzer Amulet device: full results of the prospective global observational study
.
Eur Heart J
 
2020
;
41
:
2894
901
.

376

Reddy
 
VY
,
Mobius-Winkler
 
S
,
Miller
 
MA
,
Neuzil
 
P
,
Schuler
 
G
,
Wiebe
 
J
, et al.  
Left atrial appendage closure with the Watchman device in patients with a contraindication for oral anticoagulation: the ASAP study (ASA Plavix Feasibility Study with Watchman Left Atrial Appendage Closure Technology)
.
J Am Coll Cardiol
 
2013
;
61
:
2551
6
.

377

Sondergaard
 
L
,
Wong
 
YH
,
Reddy
 
VY
,
Boersma
 
LVA
,
Bergmann
 
MW
,
Doshi
 
S
, et al.  
Propensity-matched comparison of oral anticoagulation versus antiplatelet therapy after left atrial appendage closure with Watchman
.
JACC Cardiovasc Interv
 
2019
;
12
:
1055
63
.

378

Flores-Umanzor
 
EJ
,
Cepas-Guillen
 
PL
,
Arzamendi
 
D
,
Cruz-Gonzalez
 
I
,
Regueiro
 
A
,
Freixa
 
X
.
Rationale and design of a randomized clinical trial to compare two antithrombotic strategies after left atrial appendage occlusion: double antiplatelet therapy vs. apixaban (ADALA study)
.
J Interv Card Electrophysiol
 
2020
;
59
:
471
7
.

379

Aminian
 
A
,
Schmidt
 
B
,
Mazzone
 
P
,
Berti
 
S
,
Fischer
 
S
,
Montorfano
 
M
, et al.  
Incidence, characterization, and clinical impact of device-related thrombus following left atrial appendage occlusion in the prospective global AMPLATZER amulet observational study
.
JACC Cardiovasc Interv
 
2019
;
12
:
1003
14
.

380

Kany
 
S
,
Metzner
 
A
,
Lubos
 
E
,
Kirchhof
 
P
.
The atrial fibrillation heart team-guiding therapy in left atrial appendage occlusion with increasingly complex patients and little evidence
.
Eur Heart J
 
2022
;
43
:
1691
2
.

381

Saw
 
J
,
Holmes
 
DR
,
Cavalcante
 
JL
,
Freeman
 
JV
,
Goldsweig
 
AM
,
Kavinsky
 
CJ
, et al.  
SCAI/HRS expert consensus statement on transcatheter left atrial appendage closure
.
Heart Rhythm
 
2023
;
20
:
e1
16
.

382

Cruz-González
 
I
,
González-Ferreiro
 
R
,
Freixa
 
X
,
Gafoor
 
S
,
Shakir
 
S
,
Omran
 
H
, et al.  
Left atrial appendage occlusion for stroke despite oral anticoagulation (resistant stroke). Results from the Amplatzer Cardiac Plug registry
.
Rev Esp Cardiol (Engl Ed)
 
2020
;
73
:
28
34
.

383

Willits
 
I
,
Keltie
 
K
,
Linker
 
N
,
de Belder
 
M
,
Henderson
 
R
,
Patrick
 
H
, et al.  
Left atrial appendage occlusion in the UK: prospective registry and data linkage to hospital episode statistics
.
Eur Heart J Qual Care Clin Outcomes
 
2021
;
7
:
468
75
.

384

Price
 
MJ
,
Valderrabano
 
M
,
Zimmerman
 
S
,
Friedman
 
DJ
,
Kar
 
S
,
Curtis
 
JP
, et al.  
Periprocedural pericardial effusion complicating transcatheter left atrial appendage occlusion: a report from the NCDR LAAO registry
.
Circ Cardiovasc Interv
 
2022
;
15
:
e011718
.

385

Aminian
 
A
,
De Backer
 
O
,
Nielsen-Kudsk
 
JE
,
Mazzone
 
P
,
Berti
 
S
,
Fischer
 
S
, et al.  
Incidence and clinical impact of major bleeding following left atrial appendage occlusion: insights from the amplatzer amulet observational post-market study
.
EuroIntervention
 
2021
;
17
:
774
82
.

386

Boersma
 
LV
,
Ince
 
H
,
Kische
 
S
,
Pokushalov
 
E
,
Schmitz
 
T
,
Schmidt
 
B
, et al.  
Evaluating real-world clinical outcomes in atrial fibrillation patients receiving the WATCHMAN left atrial appendage closure technology: final 2–year outcome data of the EWOLUTION trial focusing on history of stroke and hemorrhage
.
Circ Arrhythm Electrophysiol
 
2019
;
12
:
e006841
.

387

Tzikas
 
A
,
Shakir
 
S
,
Gafoor
 
S
,
Omran
 
H
,
Berti
 
S
,
Santoro
 
G
, et al.  
Left atrial appendage occlusion for stroke prevention in atrial fibrillation: multicentre experience with the AMPLATZER cardiac plug
.
EuroIntervention
 
2016
;
11
:
1170
9
.

388

Nazir
 
S
,
Ahuja
 
KR
,
Kolte
 
D
,
Isogai
 
T
,
Michihata
 
N
,
Saad
 
AM
, et al.  
Association of hospital procedural volume with outcomes of percutaneous left atrial appendage occlusion
.
JACC Cardiovasc Interv
 
2021
;
14
:
554
61
.

389

Freeman
 
JV
,
Varosy
 
P
,
Price
 
MJ
,
Slotwiner
 
D
,
Kusumoto
 
FM
,
Rammohan
 
C
, et al.  
The NCDR left atrial appendage occlusion registry
.
J Am Coll Cardiol
 
2020
;
75
:
1503
18
.

390

Cruz-Gonzalez
 
I
,
Korsholm
 
K
,
Trejo-Velasco
 
B
,
Thambo
 
JB
,
Mazzone
 
P
,
Rioufol
 
G
, et al.  
Procedural and short-term results with the new watchman FLX left atrial appendage occlusion device
.
JACC Cardiovasc Interv
 
2020
;
13
:
2732
41
.

391

Simard
 
T
,
Jung
 
RG
,
Lehenbauer
 
K
,
Piayda
 
K
,
Pracon
 
R
,
Jackson
 
GG
, et al.  
Predictors of device-related thrombus following percutaneous left atrial appendage occlusion
.
J Am Coll Cardiol
 
2021
;
78
:
297
313
.

392

Simard
 
TJ
,
Hibbert
 
B
,
Alkhouli
 
MA
,
Abraham
 
NS
,
Holmes
 
DR
, Jr
.
Device-related thrombus following left atrial appendage occlusion
.
EuroIntervention
 
2022
;
18
:
224
32
.

393

Lempereur
 
M
,
Aminian
 
A
,
Freixa
 
X
,
Gafoor
 
S
,
Kefer
 
J
,
Tzikas
 
A
, et al.  
Device-associated thrombus formation after left atrial appendage occlusion: a systematic review of events reported with the Watchman, the Amplatzer Cardiac Plug and the Amulet
.
Catheter Cardiovasc Interv
 
2017
;
90
:
E111
21
.

394

Saw
 
J
,
Tzikas
 
A
,
Shakir
 
S
,
Gafoor
 
S
,
Omran
 
H
,
Nielsen-Kudsk
 
JE
, et al.  
Incidence and clinical impact of device-associated thrombus and peri-device leak following left atrial appendage closure with the amplatzer cardiac plug
.
JACC Cardiovasc Interv
 
2017
;
10
:
391
9
.

395

Fauchier
 
L
,
Cinaud
 
A
,
Brigadeau
 
F
,
Lepillier
 
A
,
Pierre
 
B
,
Abbey
 
S
, et al.  
Device-related thrombosis after percutaneous left atrial appendage occlusion for atrial fibrillation
.
J Am Coll Cardiol
 
2018
;
71
:
1528
36
.

396

Dukkipati
 
SR
,
Kar
 
S
,
Holmes
 
DR
,
Doshi
 
SK
,
Swarup
 
V
,
Gibson
 
DN
, et al.  
Device-related thrombus after left atrial appendage closure: incidence, predictors, and outcomes
.
Circulation
 
2018
;
138
:
874
85
.

397

Kar
 
S
,
Doshi
 
SK
,
Sadhu
 
A
,
Horton
 
R
,
Osorio
 
J
,
Ellis
 
C
, et al.  
Primary outcome evaluation of a next-generation left atrial appendage closure device: results from the PINNACLE FLX trial
.
Circulation
 
2021
;
143
:
1754
62
.

398

Alkhouli
 
M
,
Du
 
C
,
Killu
 
A
,
Simard
 
T
,
Noseworthy
 
PA
,
Friedman
 
PA
, et al.  
Clinical impact of residual leaks following left atrial appendage occlusion: insights from the NCDR LAAO registry
.
JACC Clin Electrophysiol
 
2022
;
8
:
766
78
.

399

Tsai
 
YC
,
Phan
 
K
,
Munkholm-Larsen
 
S
,
Tian
 
DH
,
La Meir
 
M
,
Yan
 
TD
.
Surgical left atrial appendage occlusion during cardiac surgery for patients with atrial fibrillation: a meta-analysis
.
Eur J Cardiothorac Surg
 
2015
;
47
:
847
54
.

400

Whitlock
 
RP
,
Vincent
 
J
,
Blackall
 
MH
,
Hirsh
 
J
,
Fremes
 
S
,
Novick
 
R
, et al.  
Left atrial appendage occlusion study II (LAAOS II)
.
Can J Cardiol
 
2013
;
29
:
1443
7
.

401

Whitlock
 
RP
,
Belley-Cote
 
EP
,
Paparella
 
D
,
Healey
 
JS
,
Brady
 
K
,
Sharma
 
M
, et al.  
Left atrial appendage occlusion during cardiac surgery to prevent stroke
.
N Engl J Med
 
2021
;
384
:
2081
91
.

402

Zhang
 
S
,
Cui
 
Y
,
Li
 
J
,
Tian
 
H
,
Yun
 
Y
,
Zhou
 
X
, et al.  
Concomitant transcatheter occlusion versus thoracoscopic surgical clipping for left atrial appendage in patients undergoing ablation for atrial fibrillation: a meta-analysis
.
Front Cardiovasc Med
 
2022
;
9
:
970847
.

403

van Laar
 
C
,
Verberkmoes
 
NJ
,
van Es
 
HW
,
Lewalter
 
T
,
Dunnington
 
G
,
Stark
 
S
, et al.  
Thoracoscopic left atrial appendage clipping: a multicenter cohort analysis
.
JACC Clin Electrophysiol
 
2018
;
4
:
893
901
.

404

Kiviniemi
 
T
,
Bustamante-Munguira
 
J
,
Olsson
 
C
,
Jeppsson
 
A
,
Halfwerk
 
FR
,
Hartikainen
 
J
, et al.  
A randomized prospective multicenter trial for stroke prevention by prophylactic surgical closure of the left atrial appendage in patients undergoing bioprosthetic aortic valve surgery–LAA-CLOSURE trial protocol
.
Am Heart J
 
2021
;
237
:
127
34
.

405

Cartledge
 
R
,
Suwalski
 
G
,
Witkowska
 
A
,
Gottlieb
 
G
,
Cioci
 
A
,
Chidiac
 
G
, et al.  
Standalone epicardial left atrial appendage exclusion for thromboembolism prevention in atrial fibrillation
.
Interact Cardiovasc Thorac Surg
 
2022
;
34
:
548
55
.

406

Branzoli
 
S
,
Guarracini
 
F
,
Marini
 
M
,
D’Onghia
 
G
,
Penzo
 
D
,
Piffer
 
S
, et al.  
Heart team for left atrial appendage occlusion: a patient-tailored approach
.
J Clin Med
 
2022
;
11
:
176
.

407

Toale
 
C
,
Fitzmaurice
 
GJ
,
Eaton
 
D
,
Lyne
 
J
,
Redmond
 
KC
.
Outcomes of left atrial appendage occlusion using the AtriClip device: a systematic review
.
Interact Cardiovasc Thorac Surg
 
2019
;
29
:
655
62
.

408

Caliskan
 
E
,
Sahin
 
A
,
Yilmaz
 
M
,
Seifert
 
B
,
Hinzpeter
 
R
,
Alkadhi
 
H
, et al.  
Epicardial left atrial appendage AtriClip occlusion reduces the incidence of stroke in patients with atrial fibrillation undergoing cardiac surgery
.
Europace
 
2018
;
20
:
e105
14
.

409

Nso
 
N
,
Nassar
 
M
,
Zirkiyeva
 
M
,
Lakhdar
 
S
,
Shaukat
 
T
,
Guzman
 
L
, et al.  
Outcomes of cardiac surgery with left atrial appendage occlusion versus no occlusion, direct oral anticoagulants, and vitamin K antagonists: a systematic review with meta-analysis
.
Int J Cardiol Heart Vasc
 
2022
;
40
:
100998
.

410

Ibrahim
 
AM
,
Tandan
 
N
,
Koester
 
C
,
Al-Akchar
 
M
,
Bhandari
 
B
,
Botchway
 
A
, et al.  
Meta-analysis evaluating outcomes of surgical left atrial appendage occlusion during cardiac surgery
.
Am J Cardiol
 
2019
;
124
:
1218
25
.

411

Park-Hansen
 
J
,
Holme
 
SJV
,
Irmukhamedov
 
A
,
Carranza
 
CL
,
Greve
 
AM
,
Al-Farra
 
G
, et al.  
Adding left atrial appendage closure to open heart surgery provides protection from ischemic brain injury six years after surgery independently of atrial fibrillation history: the LAACS randomized study
.
J Cardiothorac Surg
 
2018
;
13
:
53
.

412

Soltesz
 
EG
,
Dewan
 
KC
,
Anderson
 
LH
,
Ferguson
 
MA
,
Gillinov
 
AM
.
Improved outcomes in CABG patients with atrial fibrillation associated with surgical left atrial appendage exclusion
.
J Card Surg
 
2021
;
36
:
1201
8
.

413

Fu
 
M
,
Qin
 
Z
,
Zheng
 
S
,
Li
 
Y
,
Yang
 
S
,
Zhao
 
Y
, et al.  
Thoracoscopic left atrial appendage occlusion for stroke prevention compared with long-term warfarin therapy in patients with nonvalvular atrial fibrillation
.
Am J Cardiol
 
2019
;
123
:
50
6
.

414

Peterson
 
D
,
Geison
 
E
.
Pharmacist interventions to reduce modifiable bleeding risk factors using HAS-BLED in patients taking warfarin
.
Fed Pract
 
2017
;
34
:
S16
20
.

415

Chao
 
TF
,
Lip
 
GYH
,
Lin
 
YJ
,
Chang
 
SL
,
Lo
 
LW
,
Hu
 
YF
, et al.  
Incident risk factors and major bleeding in patients with atrial fibrillation treated with oral anticoagulants: a comparison of baseline, follow-up and Delta HAS-BLED scores with an approach focused on modifiable bleeding risk factors
.
Thromb Haemost
 
2018
;
47
:
768
77
.

416

Linkins
 
LA
,
Choi
 
PT
,
Douketis
 
JD
.
Clinical impact of bleeding in patients taking oral anticoagulant therapy for venous thromboembolism: a meta-analysis
.
Ann Intern Med
 
2003
;
139
:
893
900
.

417

Kirchhof
 
P
,
Haas
 
S
,
Amarenco
 
P
,
Hess
 
S
,
Lambelet
 
M
,
van Eickels
 
M
, et al.  
Impact of modifiable bleeding risk factors on major bleeding in patients with atrial fibrillation anticoagulated with rivaroxaban
.
J Am Heart Assoc
 
2020
;
9
:
e009530
.

418

Guo
 
Y
,
Lane
 
DA
,
Chen
 
Y
,
Lip
 
GYH
;
mAF-App II Trial investigators
.
Regular bleeding risk assessment associated with reduction in bleeding outcomes: the mAFA-II randomized trial
.
Am J Med
 
2020
;
133
:
1195
1202.e2
.

419

Lane
 
DA
,
Lip
 
GYH
.
Stroke and bleeding risk stratification in atrial fibrillation: a critical appraisal
.
Eur Heart J Suppl
 
2020
;
22
:
O14
27
.

420

Lip
 
GYH
,
Lane
 
DA
.
Bleeding risk assessment in atrial fibrillation: observations on the use and misuse of bleeding risk scores
.
J Thromb Haemost
 
2016
;
14
:
1711
4
.

421

Gorog
 
DA
,
Gue
 
YX
,
Chao
 
TF
,
Fauchier
 
L
,
Ferreiro
 
JL
,
Huber
 
K
, 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
:
1844
71
.

422

Nelson
 
WW
,
Laliberté
 
F
,
Patel
 
AA
,
Germain
 
G
,
Pilon
 
D
,
McCormick
 
N
, et al.  
Stroke risk reduction outweighed bleeding risk increase from vitamin K antagonist treatment among nonvalvular atrial fibrillation patients with high stroke risk and low bleeding risk
.
Curr Med Res Opin
 
2017
;
33
:
631
8
.

423

Hijazi
 
Z
,
Lindbäck
 
J
,
Oldgren
 
J
,
Benz
 
AP
,
Alexander
 
JH
,
Connolly
 
SJ
, et al.  
Individual net clinical outcome with oral anticoagulation in atrial fibrillation using the ABC-AF risk scores
.
Am Heart J
 
2023
;
261
:
55
63
.

424

Hijazi
 
Z
,
Lindbäck
 
J
,
Alexander
 
JH
,
Hanna
 
M
,
Held
 
C
,
Hylek
 
EM
, et al.  
The ABC (age, biomarkers, clinical history) stroke risk score: a biomarker-based risk score for predicting stroke in atrial fibrillation
.
Eur Heart J
 
2016
;
37
:
1582
90
.

425

Gao
 
X
,
Cai
 
X
,
Yang
 
Y
,
Zhou
 
Y
,
Zhu
 
W
.
Diagnostic accuracy of the HAS-BLED bleeding score in VKA- or DOAC-treated patients with atrial fibrillation: a systematic review and meta-analysis
.
Front Cardiovasc Med
 
2021
;
8
:
757087
.

426

Zhu
 
W
,
He
 
W
,
Guo
 
L
,
Wang
 
X
,
Hong
 
K
.
The HAS-BLED score for predicting major bleeding risk in anticoagulated patients with atrial fibrillation: a systematic review and meta-analysis
.
Clin Cardiol
 
2015
;
38
:
555
61
.

427

Caldeira
 
D
,
Costa
 
J
,
Fernandes
 
RM
,
Pinto
 
FJ
,
Ferreira
 
JJ
.
Performance of the HAS-BLED high bleeding-risk category, compared to ATRIA and HEMORR2HAGES in patients with atrial fibrillation: a systematic review and meta-analysis
.
J Interv Card Electrophysiol
 
2014
;
40
:
277
84
.

428

Zeng
 
J
,
Yu
 
P
,
Cui
 
W
,
Wang
 
X
,
Ma
 
J
,
Zeng
 
C
.
Comparison of HAS-BLED with other risk models for predicting the bleeding risk in anticoagulated patients with atrial fibrillation: a PRISMA-compliant article
.
Medicine (Baltimore)
 
2020
;
99
:
e20782
.

429

Wang
 
C
,
Yu
 
Y
,
Zhu
 
W
,
Yu
 
J
,
Lip
 
GYH
,
Hong
 
K
.
Comparing the ORBIT and HAS-BLED bleeding risk scores in anticoagulated atrial fibrillation patients: a systematic review and meta-analysis
.
Oncotarget
 
2017
;
8
:
109703
11
.

430

Loewen
 
P
,
Dahri
 
K
.
Risk of bleeding with oral anticoagulants: an updated systematic review and performance analysis of clinical prediction rules
.
Ann Hematol
 
2011
;
90
:
1191
200
.

431

Hilkens
 
NA
,
Algra
 
A
,
Greving
 
JP
.
Predicting major bleeding in ischemic stroke patients with atrial fibrillation
.
Stroke
 
2017
;
48
:
3142
4
.

432

Dalgaard
 
F
,
Pieper
 
K
,
Verheugt
 
F
,
Camm
 
AJ
,
Fox
 
KA
,
Kakkar
 
AK
, et al.  
GARFIELD-AF model for prediction of stroke and major bleeding in atrial fibrillation: a Danish nationwide validation study
.
BMJ Open
 
2019
;
9
:
e033283
.

433

Mori
 
N
,
Sotomi
 
Y
,
Hirata
 
A
,
Hirayama
 
A
,
Sakata
 
Y
,
Higuchi
 
Y
.
External validation of the ORBIT bleeding score and the HAS-BLED score in nonvalvular atrial fibrillation patients using direct oral anticoagulants (Asian Data from the DIRECT Registry)
.
Am J Cardiol
 
2019
;
124
:
1044
8
.

434

Yao
 
X
,
Gersh
 
BJ
,
Sangaralingham
 
LR
,
Kent
 
DM
,
Shah
 
ND
,
Abraham
 
NS
, et al.  
Comparison of the CHA2DS2-VASc, CHADS2, HAS-BLED, ORBIT, and ATRIA risk scores in predicting non-vitamin K antagonist oral anticoagulants-associated bleeding in patients with atrial fibrillation
.
Am J Cardiol
 
2017
;
120
:
1549
56
.

435

Giustozzi
 
M
,
Proietti
 
G
,
Becattini
 
C
,
Roila
 
F
,
Agnelli
 
G
,
Mandalà
 
M
.
ICH in primary or metastatic brain cancer patients with or without anticoagulant treatment: a systematic review and meta-analysis
.
Blood Adv
 
2022
;
6
:
4873
83
.

436

Shoamanesh
 
A
.
Anticoagulation in patients with cerebral amyloid angiopathy
.
Lancet
 
2023
;
402
:
1418
9
.

437

Kurlander
 
JE
,
Barnes
 
GD
,
Fisher
 
A
,
Gonzalez
 
JJ
,
Helminski
 
D
,
Saini
 
SD
, et al.  
Association of antisecretory drugs with upper gastrointestinal bleeding in patients using oral anticoagulants: a systematic review and meta-analysis
.
Am J Med
 
2022
;
135
:
1231
1243.e8
.

438

Moayyedi
 
P
,
Eikelboom
 
JW
,
Bosch
 
J
,
Connolly
 
SJ
,
Dyal
 
L
,
Shestakovska
 
O
, et al.  
Pantoprazole to prevent gastroduodenal events in patients receiving rivaroxaban and/or aspirin in a randomized, double-blind, placebo-controlled trial
.
Gastroenterology
 
2019
;
157
:
403
412.e5
.

439

DiMarco
 
JP
,
Flaker
 
G
,
Waldo
 
AL
,
Corley
 
SD
,
Greene
 
HL
,
Safford
 
RE
, et al.  
Factors affecting bleeding risk during anticoagulant therapy in patients with atrial fibrillation: observations from the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study
.
Am Heart J
 
2005
;
149
:
650
6
.

440

Harskamp
 
RE
,
Lucassen
 
WAM
,
Lopes
 
RD
,
Himmelreich
 
JCL
,
Parati
 
G
,
Weert
 
H
.
Risk of stroke and bleeding in relation to hypertension in anticoagulated patients with atrial fibrillation: a meta-analysis of randomised controlled trials
.
Acta Cardiol
 
2022
;
77
:
191
5
.

441

Schulman
 
S
,
Beyth
 
RJ
,
Kearon
 
C
,
Levine
 
MN
.
Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians evidence-based clinical practice guidelines (8th Edition)
.
Chest
 
2008
;
133
:
257s
98s
.

442

Gallego
 
P
,
Roldán
 
V
,
Torregrosa
 
JM
,
Gálvez
 
J
,
Valdés
 
M
,
Vicente
 
V
, et al.  
Relation of the HAS-BLED bleeding risk score to major bleeding, cardiovascular events, and mortality in anticoagulated patients with atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2012
;
5
:
312
8
.

443

Bouillon
 
K
,
Bertrand
 
M
,
Boudali
 
L
,
Ducimetière
 
P
,
Dray-Spira
 
R
,
Zureik
 
M
.
Short-term risk of bleeding during heparin bridging at initiation of vitamin K antagonist therapy in more than 90 000 patients with nonvalvular atrial fibrillation managed in outpatient care
.
J Am Heart Assoc
 
2016
;
5
:
e004065
.

444

White
 
HD
,
Gruber
 
M
,
Feyzi
 
J
,
Kaatz
 
S
,
Tse
 
HF
,
Husted
 
S
, et al.  
Comparison of outcomes among patients randomized to warfarin therapy according to anticoagulant control: results from SPORTIF III and V
.
Arch Intern Med
 
2007
;
167
:
239
45
.

445

January
 
CT
,
Wann
 
LS
,
Alpert
 
JS
,
Calkins
 
H
,
Cigarroa
 
JE
,
Cleveland
 
JC
, Jr
, et al.  
2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society
.
Circulation
 
2014
;
130
:
2071
104
.

446

Olesen
 
JB
,
Lip
 
GY
,
Lindhardsen
 
J
,
Lane
 
DA
,
Ahlehoff
 
O
,
Hansen
 
ML
, et al.  
Risks of thromboembolism and bleeding with thromboprophylaxis in patients with atrial fibrillation: a net clinical benefit analysis using a ‘real world’ nationwide cohort study
.
Thromb Haemost
 
2011
;
106
:
739
49
.

447

Tomaselli
 
GF
,
Mahaffey
 
KW
,
Cuker
 
A
,
Dobesh
 
PP
,
Doherty
 
JU
,
Eikelboom
 
JW
, et al.  
2020 ACC expert consensus decision pathway on management of bleeding in patients on oral anticoagulants: a report of the American College of Cardiology Solution Set Oversight Committee
.
J Am Coll Cardiol
 
2020
;
76
:
594
622
.

448

Cuker
 
A
.
Laboratory measurement of the non-vitamin K antagonist oral anticoagulants: selecting the optimal assay based on drug, assay availability, and clinical indication
.
J Thromb Thrombolysis
 
2016
;
41
:
241
7
.

449

Douxfils
 
J
,
Ageno
 
W
,
Samama
 
CM
,
Lessire
 
S
,
Ten Cate
 
H
,
Verhamme
 
P
, et al.  
Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians
.
J Thromb Haemost
 
2018
;
16
:
209
19
.

450

Milling
 
TJ
, Jr,
Refaai
 
MA
,
Sarode
 
R
,
Lewis
 
B
,
Mangione
 
A
,
Durn
 
BL
, et al.  
Safety of a four-factor prothrombin complex concentrate versus plasma for vitamin K antagonist reversal: an integrated analysis of two phase IIIb clinical trials
.
Acad Emerg Med
 
2016
;
23
:
466
75
.

451

Pollack
 
CV
, Jr,
Reilly
 
PA
,
van Ryn
 
J
,
Eikelboom
 
JW
,
Glund
 
S
,
Bernstein
 
RA
, et al.  
Idarucizumab for dabigatran reversal—full cohort analysis
.
N Engl J Med
 
2017
;
377
:
431
41
.

452

Connolly SJ, Sharma M, Cohen AT, Demchuk AM, Członkowska A, Lindgren AG, et al. ANNEXA–I investigators. Andexanet for factor Xa inhibitor–associated acute intracerebral hemorrhage. N Engl J Med 2024;390:1745–55.

453

Milioglou
 
I
,
Farmakis
 
I
,
Neudeker
 
M
,
Hussain
 
Z
,
Guha
 
A
,
Giannakoulas
 
G
, et al.  
Prothrombin complex concentrate in major bleeding associated with DOACs; an updated systematic review and meta-analysis
.
J Thromb Thrombolysis
 
2021
;
52
:
1137
50
.

454

Meyre
 
PB
,
Blum
 
S
,
Hennings
 
E
,
Aeschbacher
 
S
,
Reichlin
 
T
,
Rodondi
 
N
, et al.  
Bleeding and ischaemic events after first bleed in anticoagulated atrial fibrillation patients: risk and timing
.
Eur Heart J
 
2022
;
43
:
4899
908
.

455

Connolly
 
SJ
,
Crowther
 
M
,
Eikelboom
 
JW
,
Gibson
 
CM
,
Curnutte
 
JT
,
Lawrence
 
JH
, et al.  
Full study report of andexanet alfa for bleeding associated with factor Xa inhibitors
.
N Engl J Med
 
2019
;
380
:
1326
35
.

456

Fanikos
 
J
,
Murwin
 
D
,
Gruenenfelder
 
F
,
Tartakovsky
 
I
,
França
 
LR
,
Reilly
 
PA
, et al.  
Global use of idarucizumab in clinical practice: outcomes of the RE-VECTO surveillance program
.
Thromb Haemost
 
2020
;
120
:
27
35
.

457

Kotecha
 
D
,
Calvert
 
M
,
Deeks
 
JJ
,
Griffith
 
M
,
Kirchhof
 
P
,
Lip
 
GY
, et al.  
A review of rate control in atrial fibrillation, and the rationale and protocol for the RATE-AF trial
.
BMJ Open
 
2017
;
7
:
e015099
.

458

Hess
 
PL
,
Sheng
 
S
,
Matsouaka
 
R
,
DeVore
 
AD
,
Heidenreich
 
PA
,
Yancy
 
CW
, et al.  
Strict versus lenient versus poor rate control among patients with atrial fibrillation and heart failure (from the get with the guidelines—heart failure program)
.
Am J Cardiol
 
2020
;
125
:
894
900
.

459

Van Gelder
 
IC
,
Groenveld
 
HF
,
Crijns
 
HJ
,
Tuininga
 
YS
,
Tijssen
 
JG
,
Alings
 
AM
, et al.  
Lenient versus strict rate control in patients with atrial fibrillation
.
N Engl J Med
 
2010
;
362
:
1363
73
.

460

Olshansky
 
B
,
Rosenfeld
 
LE
,
Warner
 
AL
,
Solomon
 
AJ
,
O’Neill
 
G
,
Sharma
 
A
, et al.  
The Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study: approaches to control rate in atrial fibrillation
.
J Am Coll Cardiol
 
2004
;
43
:
1201
8
.

461

Ulimoen
 
SR
,
Enger
 
S
,
Carlson
 
J
,
Platonov
 
PG
,
Pripp
 
AH
,
Abdelnoor
 
M
, et al.  
Comparison of four single-drug regimens on ventricular rate and arrhythmia-related symptoms in patients with permanent atrial fibrillation
.
Am J Cardiol
 
2013
;
111
:
225
30
.

462

Tisdale
 
JE
,
Padhi
 
ID
,
Goldberg
 
AD
,
Silverman
 
NA
,
Webb
 
CR
,
Higgins
 
RS
, et al.  
A randomized, double-blind comparison of intravenous diltiazem and digoxin for atrial fibrillation after coronary artery bypass surgery
.
Am Heart J
 
1998
;
135
:
739
47
.

463

Khand
 
AU
,
Rankin
 
AC
,
Martin
 
W
,
Taylor
 
J
,
Gemmell
 
I
,
Cleland
 
JG
.
Carvedilol alone or in combination with digoxin for the management of atrial fibrillation in patients with heart failure?
 
J Am Coll Cardiol
 
2003
;
42
:
1944
51
.

464

Nikolaidou
 
T
,
Channer
 
KS
.
Chronic atrial fibrillation: a systematic review of medical heart rate control management
.
Postgrad Med J
 
2009
;
85
:
303
12
.

465

Figulla
 
HR
,
Gietzen
 
F
,
Zeymer
 
U
,
Raiber
 
M
,
Hegselmann
 
J
,
Soballa
 
R
, et al.  
Diltiazem improves cardiac function and exercise capacity in patients with idiopathic dilated cardiomyopathy. Results of the Diltiazem in Dilated Cardiomyopathy Trial
.
Circulation
 
1996
;
94
:
346
52
.

466

Andrade
 
JG
,
Roy
 
D
,
Wyse
 
DG
,
Tardif
 
JC
,
Talajic
 
M
,
Leduc
 
H
, et al.  
Heart rate and adverse outcomes in patients with atrial fibrillation: a combined AFFIRM and AF-CHF substudy
.
Heart Rhythm
 
2016
;
13
:
54
61
.

467

Weerasooriya
 
R
,
Davis
 
M
,
Powell
 
A
,
Szili-Torok
 
T
,
Shah
 
C
,
Whalley
 
D
, et al.  
The Australian Intervention Randomized Control of Rate in Atrial Fibrillation Trial (AIRCRAFT)
.
J Am Coll Cardiol
 
2003
;
41
:
1697
702
.

468

Lim
 
KT
,
Davis
 
MJ
,
Powell
 
A
,
Arnolda
 
L
,
Moulden
 
K
,
Bulsara
 
M
, et al.  
Ablate and pace strategy for atrial fibrillation: long-term outcome of AIRCRAFT trial
.
Europace
 
2007
;
9
:
498
505
.

469

Vijayaraman
 
P
,
Subzposh
 
FA
,
Naperkowski
 
A
.
Atrioventricular node ablation and His bundle pacing
.
Europace
 
2017
;
19
:
iv10
6
.

470

Brignole
 
M
,
Pokushalov
 
E
,
Pentimalli
 
F
,
Palmisano
 
P
,
Chieffo
 
E
,
Occhetta
 
E
, et al.  
A randomized controlled trial of atrioventricular junction ablation and cardiac resynchronization therapy in patients with permanent atrial fibrillation and narrow QRS
.
Eur Heart J
 
2018
;
39
:
3999
4008
.

471

Brignole
 
M
,
Pentimalli
 
F
,
Palmisano
 
P
,
Landolina
 
M
,
Quartieri
 
F
,
Occhetta
 
E
, et al.  
AV junction ablation and cardiac resynchronization for patients with permanent atrial fibrillation and narrow QRS: the APAF-CRT mortality trial
.
Eur Heart J
 
2021
;
42
:
4731
9
.

472

Delle Karth
 
G
,
Geppert
 
A
,
Neunteufl
 
T
,
Priglinger
 
U
,
Haumer
 
M
,
Gschwandtner
 
M
, et al.  
Amiodarone versus diltiazem for rate control in critically ill patients with atrial tachyarrhythmias
.
Crit Care Med
 
2001
;
29
:
1149
53
.

473

Hou
 
ZY
,
Chang
 
MS
,
Chen
 
CY
,
Tu
 
MS
,
Lin
 
SL
,
Chiang
 
HT
, et al.  
Acute treatment of recent-onset atrial fibrillation and flutter with a tailored dosing regimen of intravenous amiodarone. A randomized, digoxin-controlled study
.
Eur Heart J
 
1995
;
16
:
521
8
.

474

Van Gelder
 
IC
,
Wyse
 
DG
,
Chandler
 
ML
,
Cooper
 
HA
,
Olshansky
 
B
,
Hagens
 
VE
, et al.  
Does intensity of rate-control influence outcome in atrial fibrillation? An analysis of pooled data from the RACE and AFFIRM studies
.
Europace
 
2006
;
8
:
935
42
.

475

Scheuermeyer
 
FX
,
Grafstein
 
E
,
Stenstrom
 
R
,
Christenson
 
J
,
Heslop
 
C
,
Heilbron
 
B
, et al.  
Safety and efficiency of calcium channel blockers versus beta-blockers for rate control in patients with atrial fibrillation and no acute underlying medical illness
.
Acad Emerg Med
 
2013
;
20
:
222
30
.

476

Siu
 
CW
,
Lau
 
CP
,
Lee
 
WL
,
Lam
 
KF
,
Tse
 
HF
.
Intravenous diltiazem is superior to intravenous amiodarone or digoxin for achieving ventricular rate control in patients with acute uncomplicated atrial fibrillation
.
Crit Care Med
 
2009
;
37
:
2174
9
, quiz 2180.

477

Perrett
 
M
,
Gohil
 
N
,
Tica
 
O
,
Bunting
 
KV
,
Kotecha
 
D
.
Efficacy and safety of intravenous beta-blockers in acute atrial fibrillation and flutter is dependent on beta-1 selectivity: a systematic review and meta-analysis of randomised trials
.
Clin Res Cardiol
 
2023
;113:831–41.

478

Darby
 
AE
,
Dimarco
 
JP
.
Management of atrial fibrillation in patients with structural heart disease
.
Circulation
 
2012
;
125
:
945
57
.

479

Imamura
 
T
,
Kinugawa
 
K
.
Novel rate control strategy with landiolol in patients with cardiac dysfunction and atrial fibrillation
.
ESC Heart Fail
 
2020
;
7
:
2208
13
.

480

Ulimoen
 
SR
,
Enger
 
S
,
Pripp
 
AH
,
Abdelnoor
 
M
,
Arnesen
 
H
,
Gjesdal
 
K
, et al.  
Calcium channel blockers improve exercise capacity and reduce N-terminal Pro-B-type natriuretic peptide levels compared with beta-blockers in patients with permanent atrial fibrillation
.
Eur Heart J
 
2014
;
35
:
517
24
.

481

Connolly
 
SJ
,
Camm
 
AJ
,
Halperin
 
JL
,
Joyner
 
C
,
Alings
 
M
,
Amerena
 
J
, et al.  
Dronedarone in high-risk permanent atrial fibrillation
.
N Engl J Med
 
2011
;
365
:
2268
76
.

482

Karwath A, Bunting KV, Gill SK, Tica O, Pendleton S, Aziz F, et al. Redefining betablocker response in heart failure patients with sinus rhythm and atrial fibrillation: a machine learning cluster analysis. Lancet 2021;398:1427–35.

483

Koldenhof
 
T
,
Wijtvliet
 
P
,
Pluymaekers
 
N
,
Rienstra
 
M
,
Folkeringa
 
RJ
,
Bronzwaer
 
P
, et al.  
Rate control drugs differ in the prevention of progression of atrial fibrillation
.
Europace
 
2022
;
24
:
384
9
.

484

Champsi
 
A
,
Mitchell
 
C
,
Tica
 
O
,
Ziff
 
OJ
,
Bunting
 
KV
,
Mobley
 
AR
, et al. Digoxin in patients with heart failure and/or atrial fibrillation: a systematic review and meta-analysis of 5.9 million patient years of follow-up. SSRN preprint. 2023. .

485

Andrews
 
P
,
Anseeuw
 
K
,
Kotecha
 
D
,
Lapostolle
 
F
,
Thanacoody
 
R
.
Diagnosis and practical management of digoxin toxicity: a narrative review and consensus
.
Eur J Emerg Med
 
2023
;
30
:
395
401
.

486

Bavendiek
 
U
,
Berliner
 
D
,
Dávila
 
LA
,
Schwab
 
J
,
Maier
 
L
,
Philipp
 
SA
, et al.  
Rationale and design of the DIGIT-HF trial (DIGitoxin to Improve ouTcomes in patients with advanced chronic Heart Failure): a randomized, double-blind, placebo-controlled study
.
Eur J Heart Fail
 
2019
;
21
:
676
84
.

487

Clemo
 
HF
,
Wood
 
MA
,
Gilligan
 
DM
,
Ellenbogen
 
KA
.
Intravenous amiodarone for acute heart rate control in the critically ill patient with atrial tachyarrhythmias
.
Am J Cardiol
 
1998
;
81
:
594
8
.

488

Queiroga
 
A
,
Marshall
 
HJ
,
Clune
 
M
,
Gammage
 
MD
.
Ablate and pace revisited: long term survival and predictors of permanent atrial fibrillation
.
Heart
 
2003
;
89
:
1035
8
.

489

Geelen
 
P
,
Brugada
 
J
,
Andries
 
E
,
Brugada
 
P
.
Ventricular fibrillation and sudden death after radiofrequency catheter ablation of the atrioventricular junction
.
Pacing Clin Electrophysiol
 
1997
;
20
:
343
8
.

490

Wang
 
RX
,
Lee
 
HC
,
Hodge
 
DO
,
Cha
 
YM
,
Friedman
 
PA
,
Rea
 
RF
, et al.  
Effect of pacing method on risk of sudden death after atrioventricular node ablation and pacemaker implantation in patients with atrial fibrillation
.
Heart Rhythm
 
2013
;
10
:
696
701
.

491

Chatterjee
 
NA
,
Upadhyay
 
GA
,
Ellenbogen
 
KA
,
McAlister
 
FA
,
Choudhry
 
NK
,
Singh
 
JP
.
Atrioventricular nodal ablation in atrial fibrillation: a meta-analysis and systematic review
.
Circ Arrhythm Electrophysiol
 
2012
;
5
:
68
76
.

492

Bradley
 
DJ
,
Shen
 
WK
.
Overview of management of atrial fibrillation in symptomatic elderly patients: pharmacologic therapy versus AV node ablation
.
Clin Pharmacol Ther
 
2007
;
81
:
284
7
.

493

Ozcan
 
C
,
Jahangir
 
A
,
Friedman
 
PA
,
Patel
 
PJ
,
Munger
 
TM
,
Rea
 
RF
, et al.  
Long-term survival after ablation of the atrioventricular node and implantation of a permanent pacemaker in patients with atrial fibrillation
.
N Engl J Med
 
2001
;
344
:
1043
51
.

494

Chatterjee
 
NA
,
Upadhyay
 
GA
,
Ellenbogen
 
KA
,
Hayes
 
DL
,
Singh
 
JP
.
Atrioventricular nodal ablation in atrial fibrillation: a meta-analysis of biventricular vs. right ventricular pacing mode
.
Eur J Heart Fail
 
2012
;
14
:
661
7
.

495

Huang
 
W
,
Su
 
L
,
Wu
 
S
.
Pacing treatment of atrial fibrillation patients with heart failure: His bundle pacing combined with atrioventricular node ablation
.
Card Electrophysiol Clin
 
2018
;
10
:
519
35
.

496

Huang
 
W
,
Su
 
L
,
Wu
 
S
,
Xu
 
L
,
Xiao
 
F
,
Zhou
 
X
, et al.  
Benefits of permanent His bundle pacing combined with atrioventricular node ablation in atrial fibrillation patients with heart failure with both preserved and reduced left ventricular ejection fraction
.
J Am Heart Assoc
 
2017
;
6
:
e005309
.

497

Van Gelder
 
IC
,
Hagens
 
VE
,
Bosker
 
HA
,
Kingma
 
JH
,
Kamp
 
O
,
Kingma
 
T
, et al.  
A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation
.
N Engl J Med
 
2002
;
347
:
1834
40
.

498

Wyse
 
DG
,
Waldo
 
AL
,
DiMarco
 
JP
,
Domanski
 
MJ
,
Rosenberg
 
Y
,
Schron
 
EB
, et al.  
A comparison of rate control and rhythm control in patients with atrial fibrillation
.
N Engl J Med
 
2002
;
347
:
1825
33
.

499

Hohnloser
 
SH
,
Kuck
 
KH
,
Lilienthal
 
J
.
Rhythm or rate control in atrial fibrillation—pharmacological intervention in atrial fibrillation (PIAF): a randomised trial
.
Lancet
 
2000
;
356
:
1789
94
.

500

Roy
 
D
,
Talajic
 
M
,
Nattel
 
S
,
Wyse
 
DG
,
Dorian
 
P
,
Lee
 
KL
, et al.  
Rhythm control versus rate control for atrial fibrillation and heart failure
.
N Engl J Med
 
2008
;
358
:
2667
77
.

501

Blomström-Lundqvist
 
C
,
Gizurarson
 
S
,
Schwieler
 
J
,
Jensen
 
SM
,
Bergfeldt
 
L
,
Kennebäck
 
G
, et al.  
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
68
.

502

Mark
 
DB
,
Anstrom
 
KJ
,
Sheng
 
S
,
Piccini
 
JP
,
Baloch
 
KN
,
Monahan
 
KH
, et al.  
Effect of catheter ablation vs medical therapy on quality of life among patients with atrial fibrillation: the CABANA randomized clinical trial
.
JAMA
 
2019
;
321
:
1275
85
.

503

Wilber
 
DJ
,
Pappone
 
C
,
Neuzil
 
P
,
De Paola
 
A
,
Marchlinski
 
F
,
Natale
 
A
, et al.  
Comparison of antiarrhythmic drug therapy and radiofrequency catheter ablation in patients with paroxysmal atrial fibrillation: a randomized controlled trial
.
JAMA
 
2010
;
303
:
333
40
.

504

Calkins
 
H
,
Reynolds
 
MR
,
Spector
 
P
,
Sondhi
 
M
,
Xu
 
Y
,
Martin
 
A
, et al.  
Treatment of atrial fibrillation with antiarrhythmic drugs or radiofrequency ablation: two systematic literature reviews and meta-analyses
.
Circ Arrhythm Electrophysiol
 
2009
;
2
:
349
61
.

505

Jais
 
P
,
Cauchemez
 
B
,
Macle
 
L
,
Daoud
 
E
,
Khairy
 
P
,
Subbiah
 
R
, et al.  
Catheter ablation versus antiarrhythmic drugs for atrial fibrillation: the A4 study
.
Circulation
 
2008
;
118
:
2498
505
.

506

Packer
 
DL
,
Kowal
 
RC
,
Wheelan
 
KR
,
Irwin
 
JM
,
Champagne
 
J
,
Guerra
 
PG
, et al.  
Cryoballoon ablation of pulmonary veins for paroxysmal atrial fibrillation: first results of the North American Arctic Front (STOP AF) pivotal trial
.
J Am Coll Cardiol
 
2013
;
61
:
1713
23
.

507

Poole
 
JE
,
Bahnson
 
TD
,
Monahan
 
KH
,
Johnson
 
G
,
Rostami
 
H
,
Silverstein
 
AP
, et al.  
Recurrence of atrial fibrillation after catheter ablation or antiarrhythmic drug therapy in the CABANA trial
.
J Am Coll Cardiol
 
2020
;
75
:
3105
18
.

508

Mont
 
L
,
Bisbal
 
F
,
Hernandez-Madrid
 
A
,
Perez-Castellano
 
N
,
Vinolas
 
X
,
Arenal
 
A
, et al.  
Catheter ablation vs. antiarrhythmic drug treatment of persistent atrial fibrillation: a multicentre, randomized, controlled trial (SARA study)
.
Eur Heart J
 
2014
;
35
:
501
7
.

509

Scherr
 
D
,
Khairy
 
P
,
Miyazaki
 
S
,
Aurillac-Lavignolle
 
V
,
Pascale
 
P
,
Wilton
 
SB
, et al.  
Five-year outcome of catheter ablation of persistent atrial fibrillation using termination of atrial fibrillation as a procedural endpoint
.
Circ Arrhythm Electrophysiol
 
2015
;
8
:
18
24
.

510

Lau
 
DH
,
Nattel
 
S
,
Kalman
 
JM
,
Sanders
 
P
.
Modifiable risk factors and atrial fibrillation
.
Circulation
 
2017
;
136
:
583
96
.

511

Sanders
 
P
,
Elliott
 
AD
,
Linz
 
D
.
Upstream targets to treat atrial fibrillation
.
J Am Coll Cardiol
 
2017
;
70
:
2906
8
.

512

Hohnloser
 
SH
,
Crijns
 
HJ
,
van Eickels
 
M
,
Gaudin
 
C
,
Page
 
RL
,
Torp-Pedersen
 
C
, et al.  
Effect of dronedarone on cardiovascular events in atrial fibrillation
.
N Engl J Med
 
2009
;
360
:
668
78
.

513

Sohns
 
C
,
Fox
 
H
,
Marrouche
 
NF
,
Crijns
 
H
,
Costard-Jaeckle
 
A
,
Bergau
 
L
, et al.  
Catheter ablation in end-stage heart failure with atrial fibrillation
.
N Engl J Med
 
2023
;
389
:
1380
9
.

514

Di Biase
 
L
,
Mohanty
 
P
,
Mohanty
 
S
,
Santangeli
 
P
,
Trivedi
 
C
,
Lakkireddy
 
D
, et al.  
Ablation versus amiodarone for treatment of persistent atrial fibrillation in patients with congestive heart failure and an implanted device: results from the AATAC multicenter randomized trial
.
Circulation
 
2016
;
133
:
1637
44
.

515

Nuotio
 
I
,
Hartikainen
 
JE
,
Gronberg
 
T
,
Biancari
 
F
,
Airaksinen
 
KE
.
Time to cardioversion for acute atrial fibrillation and thromboembolic complications
.
JAMA
 
2014
;
312
:
647
9
.

516

Garg
 
A
,
Khunger
 
M
,
Seicean
 
S
,
Chung
 
MK
,
Tchou
 
PJ
.
Incidence of thromboembolic complications within 30 days of electrical cardioversion performed within 48 hours of atrial fibrillation onset
.
JACC Clin Electrophysiol
 
2016
;
2
:
487
94
.

517

Tampieri
 
A
,
Cipriano
 
V
,
Mucci
 
F
,
Rusconi
 
AM
,
Lenzi
 
T
,
Cenni
 
P
.
Safety of cardioversion in atrial fibrillation lasting less than 48 h without post-procedural anticoagulation in patients at low cardioembolic risk
.
Intern Emerg Med
 
2018
;
13
:
87
93
.

518

Airaksinen
 
KE
,
Gronberg
 
T
,
Nuotio
 
I
,
Nikkinen
 
M
,
Ylitalo
 
A
,
Biancari
 
F
, et al.  
Thromboembolic complications after cardioversion of acute atrial fibrillation: the FinCV (Finnish CardioVersion) study
.
J Am Coll Cardiol
 
2013
;
62
:
1187
92
.

519

Hansen
 
ML
,
Jepsen
 
RM
,
Olesen
 
JB
,
Ruwald
 
MH
,
Karasoy
 
D
,
Gislason
 
GH
, et al.  
Thromboembolic risk in 16 274 atrial fibrillation patients undergoing direct current cardioversion with and without oral anticoagulant therapy
.
Europace
 
2015
;
17
:
18
23
.

520

Bonfanti
 
L
,
Annovi
 
A
,
Sanchis-Gomar
 
F
,
Saccenti
 
C
,
Meschi
 
T
,
Ticinesi
 
A
, et al.  
Effectiveness and safety of electrical cardioversion for acute-onset atrial fibrillation in the emergency department: a real-world 10-year single center experience
.
Clin Exp Emerg Med
 
2019
;
6
:
64
9
.

521

Telles-Garcia
 
N
,
Dahal
 
K
,
Kocherla
 
C
,
Lip
 
GYH
,
Reddy
 
P
,
Dominic
 
P
.
Non-vitamin K antagonists oral anticoagulants are as safe and effective as warfarin for cardioversion of atrial fibrillation: a systematic review and meta-analysis
.
Int J Cardiol
 
2018
;
268
:
143
8
.

522

Klein
 
AL
,
Grimm
 
RA
,
Murray
 
RD
,
Apperson-Hansen
 
C
,
Asinger
 
RW
,
Black
 
IW
, et al.  
Use of transesophageal echocardiography to guide cardioversion in patients with atrial fibrillation
.
N Engl J Med
 
2001
;
344
:
1411
20
.

523

Brunetti
 
ND
,
Tarantino
 
N
,
De Gennaro
 
L
,
Correale
 
M
,
Santoro
 
F
,
Di Biase
 
M
.
Direct oral anti-coagulants compared to vitamin-K antagonists in cardioversion of atrial fibrillation: an updated meta-analysis
.
J Thromb Thrombolysis
 
2018
;
45
:
550
6
.

524

Steinberg
 
JS
,
Sadaniantz
 
A
,
Kron
 
J
,
Krahn
 
A
,
Denny
 
DM
,
Daubert
 
J
, et al.  
Analysis of cause-specific mortality in the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study
.
Circulation
 
2004
;
109
:
1973
80
.

525

Crijns
 
HJ
,
Weijs
 
B
,
Fairley
 
AM
,
Lewalter
 
T
,
Maggioni
 
AP
,
Martín
 
A
, et al.  
Contemporary real life cardioversion of atrial fibrillation: results from the multinational RHYTHM-AF study
.
Int J Cardiol
 
2014
;
172
:
588
94
.

526

Kirchhof
 
P
,
Andresen
 
D
,
Bosch
 
R
,
Borggrefe
 
M
,
Meinertz
 
T
,
Parade
 
U
, et al.  
Short-term versus long-term antiarrhythmic drug treatment after cardioversion of atrial fibrillation (Flec-SL): a prospective, randomised, open-label, blinded endpoint assessment trial
.
Lancet
 
2012
;
380
:
238
46
.

527

Zhu
 
W
,
Wu
 
Z
,
Dong
 
Y
,
Lip
 
GYH
,
Liu
 
C
.
Effectiveness of early rhythm control in improving clinical outcomes in patients with atrial fibrillation: a systematic review and meta-analysis
.
BMC Med
 
2022
;
20
:
340
.

528

Stellbrink
 
C
,
Nixdorff
 
U
,
Hofmann
 
T
,
Lehmacher
 
W
,
Daniel
 
WG
,
Hanrath
 
P
, et al.  
Safety and efficacy of enoxaparin compared with unfractionated heparin and oral anticoagulants for prevention of thromboembolic complications in cardioversion of nonvalvular atrial fibrillation: the Anticoagulation in Cardioversion using Enoxaparin (ACE) trial
.
Circulation
 
2004
;
109
:
997
1003
.

529

Lip
 
GY
,
Hammerstingl
 
C
,
Marin
 
F
,
Cappato
 
R
,
Meng
 
IL
,
Kirsch
 
B
, et al.  
Left atrial thrombus resolution in atrial fibrillation or flutter: results of a prospective study with rivaroxaban (X-TRA) and a retrospective observational registry providing baseline data (CLOT-AF)
.
Am Heart J
 
2016
;
178
:
126
34
.

530

Stiell
 
IG
,
Eagles
 
D
,
Nemnom
 
MJ
,
Brown
 
E
,
Taljaard
 
M
,
Archambault
 
PM
, et al.  
Adverse events associated with electrical cardioversion in patients with acute atrial fibrillation and atrial flutter
.
Can J Cardiol
 
2021
;
37
:
1775
82
.

531

Stiell
 
IG
,
Archambault
 
PM
,
Morris
 
J
,
Mercier
 
E
,
Eagles
 
D
,
Perry
 
JJ
, et al.  
RAFF-3 Trial: a stepped-wedge cluster randomised trial to improve care of acute atrial fibrillation and flutter in the emergency department
.
Can J Cardiol
 
2021
;
37
:
1569
77
.

532

Gurevitz
 
OT
,
Ammash
 
NM
,
Malouf
 
JF
,
Chandrasekaran
 
K
,
Rosales
 
AG
,
Ballman
 
KV
, et al.  
Comparative efficacy of monophasic and biphasic waveforms for transthoracic cardioversion of atrial fibrillation and atrial flutter
.
Am Heart J
 
2005
;
149
:
316
21
.

533

Mortensen
 
K
,
Risius
 
T
,
Schwemer
 
TF
,
Aydin
 
MA
,
Köster
 
R
,
Klemm
 
HU
, et al.  
Biphasic versus monophasic shock for external cardioversion of atrial flutter: a prospective, randomized trial
.
Cardiology
 
2008
;
111
:
57
62
.

534

Inácio
 
JF
,
da Rosa Mdos
 
S
,
Shah
 
J
,
Rosário
 
J
,
Vissoci
 
JR
,
Manica
 
AL
, et al.  
Monophasic and biphasic shock for transthoracic conversion of atrial fibrillation: systematic review and network meta-analysis
.
Resuscitation
 
2016
;
100
:
66
75
.

535

Eid
 
M
,
Abu Jazar
 
D
,
Medhekar
 
A
,
Khalife
 
W
,
Javaid
 
A
,
Ahsan
 
C
, et al.  
Anterior-posterior versus anterior-lateral electrodes position for electrical cardioversion of atrial fibrillation: a meta-analysis of randomized controlled trials
.
Int J Cardiol Heart Vasc
 
2022
;
43
:
101129
.

536

Squara
 
F
,
Elbaum
 
C
,
Garret
 
G
,
Liprandi
 
L
,
Scarlatti
 
D
,
Bun
 
SS
, et al.  
Active compression versus standard anterior-posterior defibrillation for external cardioversion of atrial fibrillation: a prospective randomized study
.
Heart Rhythm
 
2021
;
18
:
360
5
.

537

Schmidt
 
AS
,
Lauridsen
 
KG
,
Torp
 
P
,
Bach
 
LF
,
Rickers
 
H
,
Løfgren
 
B
.
Maximum-fixed energy shocks for cardioverting atrial fibrillation
.
Eur Heart J
 
2020
;
41
:
626
31
.

538

Müssigbrodt
 
A
,
John
 
S
,
Kosiuk
 
J
,
Richter
 
S
,
Hindricks
 
G
,
Bollmann
 
A
.
Vernakalant-facilitated electrical cardioversion: comparison of intravenous vernakalant and amiodarone for drug-enhanced electrical cardioversion of atrial fibrillation after failed electrical cardioversion
.
Europace
 
2016
;
18
:
51
6
.

539

Climent
 
VE
,
Marin
 
F
,
Mainar
 
L
,
Gomez-Aldaravi
 
R
,
Martinez
 
JG
,
Chorro
 
FJ
, et al.  
Effects of pretreatment with intravenous flecainide on efficacy of external cardioversion of persistent atrial fibrillation
.
Pacing Clin Electrophysiol
 
2004
;
27
:
368
72
.

540

Tieleman
 
RG
,
Van Gelder
 
IC
,
Bosker
 
HA
,
Kingma
 
T
,
Wilde
 
AA
,
Kirchhof
 
CJ
, et al.  
Does flecainide regain its antiarrhythmic activity after electrical cardioversion of persistent atrial fibrillation?
 
Heart Rhythm
 
2005
;
2
:
223
30
.

541

Oral
 
H
,
Souza
 
JJ
,
Michaud
 
GF
,
Knight
 
BP
,
Goyal
 
R
,
Strickberger
 
SA
, et al.  
Facilitating transthoracic cardioversion of atrial fibrillation with ibutilide pretreatment
.
N Engl J Med
 
1999
;
340
:
1849
54
.

542

Nair
 
M
,
George
 
LK
,
Koshy
 
SK
.
Safety and efficacy of ibutilide in cardioversion of atrial flutter and fibrillation
.
J Am Board Fam Med
 
2011
;
24
:
86
92
.

543

Bianconi
 
L
,
Mennuni
 
M
,
Lukic
 
V
,
Castro
 
A
,
Chieffi
 
M
,
Santini
 
M
.
Effects of oral propafenone administration before electrical cardioversion of chronic atrial fibrillation: a placebo-controlled study
.
J Am Coll Cardiol
 
1996
;
28
:
700
6
.

544

Channer
 
KS
,
Birchall
 
A
,
Steeds
 
RP
,
Walters
 
SJ
,
Yeo
 
WW
,
West
 
JN
, et al.  
A randomized placebo-controlled trial of pre-treatment and short- or long-term maintenance therapy with amiodarone supporting DC cardioversion for persistent atrial fibrillation
.
Eur Heart J
 
2004
;
25
:
144
50
.

545

Capucci
 
A
,
Villani
 
GQ
,
Aschieri
 
D
,
Rosi
 
A
,
Piepoli
 
MF
.
Oral amiodarone increases the efficacy of direct-current cardioversion in restoration of sinus rhythm in patients with chronic atrial fibrillation
.
Eur Heart J
 
2000
;
21
:
66
73
.

546

Um
 
KJ
,
McIntyre
 
WF
,
Healey
 
JS
,
Mendoza
 
PA
,
Koziarz
 
A
,
Amit
 
G
, et al.  
Pre- and post-treatment with amiodarone for elective electrical cardioversion of atrial fibrillation: a systematic review and meta-analysis
.
Europace
 
2019
;
21
:
856
63
.

547

Toso
 
E
,
Iannaccone
 
M
,
Caponi
 
D
,
Rotondi
 
F
,
Santoro
 
A
,
Gallo
 
C
, et al.  
Does antiarrhythmic drugs premedication improve electrical cardioversion success in persistent atrial fibrillation?
 
J Electrocardiol
 
2017
;
50
:
294
300
.

548

Ganapathy
 
AV
,
Monjazeb
 
S
,
Ganapathy
 
KS
,
Shanoon
 
F
,
Razavi
 
M
.
“Asymptomatic” persistent or permanent atrial fibrillation: a misnomer in selected patients
.
Int J Cardiol
 
2015
;
185
:
112
3
.

549

Voskoboinik
 
A
,
Kalman
 
E
,
Plunkett
 
G
,
Knott
 
J
,
Moskovitch
 
J
,
Sanders
 
P
, et al.  
A comparison of early versus delayed elective electrical cardioversion for recurrent episodes of persistent atrial fibrillation: a multi-center study
.
Int J Cardiol
 
2019
;
284
:
33
7
.

550

Airaksinen
 
KEJ
.
Early versus delayed cardioversion: why should we wait?
 
Expert Rev Cardiovasc Ther
 
2020
;
18
:
149
54
.

551

Boriani
 
G
,
Diemberger
 
I
,
Biffi
 
M
,
Martignani
 
C
,
Branzi
 
A
.
Pharmacological cardioversion of atrial fibrillation: current management and treatment options
.
Drugs
 
2004
;
64
:
2741
62
.

552

Dan
 
GA
,
Martinez-Rubio
 
A
,
Agewall
 
S
,
Boriani
 
G
,
Borggrefe
 
M
,
Gaita
 
F
, et al.  
Antiarrhythmic drugs–clinical use and clinical decision making: a consensus document from the European Heart Rhythm Association (EHRA) and European Society of Cardiology (ESC) Working Group on Cardiovascular Pharmacology, endorsed by the Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS) and International Society of Cardiovascular Pharmacotherapy (ISCP)
.
Europace
 
2018
;
20
:
731
732an
.

553

Gitt
 
AK
,
Smolka
 
W
,
Michailov
 
G
,
Bernhardt
 
A
,
Pittrow
 
D
,
Lewalter
 
T
.
Types and outcomes of cardioversion in patients admitted to hospital for atrial fibrillation: results of the German RHYTHM-AF study
.
Clin Res Cardiol
 
2013
;
102
:
713
23
.

554

Calkins
 
H
,
Hindricks
 
G
,
Cappato
 
R
,
Kim
 
YH
,
Saad
 
EB
,
Aguinaga
 
L
, et al.  
2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation: executive summary
.
Europace
 
2018
;
20
:
157
208
.

555

Danias
 
PG
,
Caulfield
 
TA
,
Weigner
 
MJ
,
Silverman
 
DI
,
Manning
 
WJ
.
Likelihood of spontaneous conversion of atrial fibrillation to sinus rhythm
.
J Am Coll Cardiol
 
1998
;
31
:
588
92
.

556

Tsiachris
 
D
,
Doundoulakis
 
I
,
Pagkalidou
 
E
,
Kordalis
 
A
,
Deftereos
 
S
,
Gatzoulis
 
KA
, et al.  
Pharmacologic cardioversion in patients with paroxysmal atrial fibrillation: a network meta-analysis
.
Cardiovasc Drugs Ther
 
2021
;
35
:
293
308
.

557

Grönberg
 
T
,
Nuotio
 
I
,
Nikkinen
 
M
,
Ylitalo
 
A
,
Vasankari
 
T
,
Hartikainen
 
JE
, et al.  
Arrhythmic complications after electrical cardioversion of acute atrial fibrillation: the FinCV study
.
Europace
 
2013
;
15
:
1432
5
.

558

Brandes
 
A
,
Crijns
 
H
,
Rienstra
 
M
,
Kirchhof
 
P
,
Grove
 
EL
,
Pedersen
 
KB
, et al.  
Cardioversion of atrial fibrillation and atrial flutter revisited: current evidence and practical guidance for a common procedure
.
Europace
 
2020
;
22
:
1149
61
.

559

Stiell
 
IG
,
Sivilotti
 
MLA
,
Taljaard
 
M
,
Birnie
 
D
,
Vadeboncoeur
 
A
,
Hohl
 
CM
, et al.  
Electrical versus pharmacological cardioversion for emergency department patients with acute atrial fibrillation (RAFF2): a partial factorial randomised trial
.
Lancet
 
2020
;
395
:
339
49
.

560

Alboni
 
P
,
Botto
 
GL
,
Baldi
 
N
,
Luzi
 
M
,
Russo
 
V
,
Gianfranchi
 
L
, et al.  
Outpatient treatment of recent-onset atrial fibrillation with the “pill-in-the-pocket” approach
.
N Engl J Med
 
2004
;
351
:
2384
91
.

561

Brembilla-Perrot
 
B
,
Houriez
 
P
,
Beurrier
 
D
,
Claudon
 
O
,
Terrier de la Chaise
 
A
,
Louis
 
P
.
Predictors of atrial flutter with 1:1 conduction in patients treated with class I antiarrhythmic drugs for atrial tachyarrhythmias
.
Int J Cardiol
 
2001
;
80
:
7
15
.

562

Conde
 
D
,
Costabel
 
JP
,
Caro
 
M
,
Ferro
 
A
,
Lambardi
 
F
,
Corrales Barboza
 
A
, et al.  
Flecainide versus vernakalant for conversion of recent-onset atrial fibrillation
.
Int J Cardiol
 
2013
;
168
:
2423
5
.

563

Markey
 
GC
,
Salter
 
N
,
Ryan
 
J
.
Intravenous flecainide for emergency department management of acute atrial fibrillation
.
J Emerg Med
 
2018
;
54
:
320
7
.

564

Martinez-Marcos
 
FJ
,
Garcia-Garmendia
 
JL
,
Ortega-Carpio
 
A
,
Fernandez-Gomez
 
JM
,
Santos
 
JM
,
Camacho
 
C
.
Comparison of intravenous flecainide, propafenone, and amiodarone for conversion of acute atrial fibrillation to sinus rhythm
.
Am J Cardiol
 
2000
;
86
:
950
3
.

565

Reisinger
 
J
,
Gatterer
 
E
,
Lang
 
W
,
Vanicek
 
T
,
Eisserer
 
G
,
Bachleitner
 
T
, et al.  
Flecainide versus ibutilide for immediate cardioversion of atrial fibrillation of recent onset
.
Eur Heart J
 
2004
;
25
:
1318
24
.

566

Zhang
 
N
,
Guo
 
JH
,
Zhang
 
H
,
Li
 
XB
,
Zhang
 
P
,
Xn
 
Y
.
Comparison of intravenous ibutilide vs. propafenone for rapid termination of recent onset atrial fibrillation
.
Int J Clin Pract
 
2005
;
59
:
1395
400
.

567

Camm
 
AJ
,
Capucci
 
A
,
Hohnloser
 
SH
,
Torp-Pedersen
 
C
,
Van Gelder
 
IC
,
Mangal
 
B
, et al.  
A randomized active-controlled study comparing the efficacy and safety of vernakalant to amiodarone in recent-onset atrial fibrillation
.
J Am Coll Cardiol
 
2011
;
57
:
313
21
.

568

Roy
 
D
,
Pratt
 
CM
,
Torp-Pedersen
 
C
,
Wyse
 
DG
,
Toft
 
E
,
Juul-Moller
 
S
, et al.  
Vernakalant hydrochloride for rapid conversion of atrial fibrillation: a phase 3, randomized, placebo-controlled trial
.
Circulation
 
2008
;
117
:
1518
25
.

569

Deedwania
 
PC
,
Singh
 
BN
,
Ellenbogen
 
K
,
Fisher
 
S
,
Fletcher
 
R
,
Singh
 
SN
.
Spontaneous conversion and maintenance of sinus rhythm by amiodarone in patients with heart failure and atrial fibrillation: observations from the veterans affairs congestive heart failure survival trial of antiarrhythmic therapy (CHF-STAT). The Department of Veterans Affairs CHF-STAT Investigators
.
Circulation
 
1998
;
98
:
2574
9
.

570

Hofmann
 
R
,
Steinwender
 
C
,
Kammler
 
J
,
Kypta
 
A
,
Wimmer
 
G
,
Leisch
 
F
.
Intravenous amiodarone bolus for treatment of atrial fibrillation in patients with advanced congestive heart failure or cardiogenic shock
.
Wien Klin Wochenschr
 
2004
;
116
:
744
9
.

571

Alboni
 
P
,
Botto
 
GL
,
Boriani
 
G
,
Russo
 
G
,
Pacchioni
 
F
,
Iori
 
M
, et al.  
Intravenous administration of flecainide or propafenone in patients with recent-onset atrial fibrillation does not predict adverse effects during ‘pill-in-the-pocket’ treatment
.
Heart
 
2010
;
96
:
546
9
.

572

Khan
 
IA
.
Single oral loading dose of propafenone for pharmacological cardioversion of recent-onset atrial fibrillation
.
J Am Coll Cardiol
 
2001
;
37
:
542
7
.

573

Khan
 
IA
.
Oral loading single dose flecainide for pharmacological cardioversion of recent-onset atrial fibrillation
.
Int J Cardiol
 
2003
;
87
:
121
8
.

574

Al-Khatib
 
SM
,
Allen LaPointe
 
NM
,
Chatterjee
 
R
,
Crowley
 
MJ
,
Dupre
 
ME
,
Kong
 
DF
, et al.  
Rate- and rhythm-control therapies in patients with atrial fibrillation: a systematic review
.
Ann Intern Med
 
2014
;
160
:
760
73
.

575

Andrade
 
JG
,
Aguilar
 
M
,
Atzema
 
C
,
Bell
 
A
,
Cairns
 
JA
,
Cheung
 
CC
, et al.  
The 2020 Canadian Cardiovascular Society/Canadian Heart Rhythm Society Comprehensive Guidelines for the Management of Atrial Fibrillation
.
Can J Cardiol
 
2020
;
36
:
1847
948
.

576

Lafuente-Lafuente
 
C
,
Valembois
 
L
,
Bergmann
 
JF
,
Belmin
 
J
.
Antiarrhythmics for maintaining sinus rhythm after cardioversion of atrial fibrillation
.
Cochrane Database Syst Rev
 
2015
;3:
CD005049
.

577

Valembois
 
L
,
Audureau
 
E
,
Takeda
 
A
,
Jarzebowski
 
W
,
Belmin
 
J
,
Lafuente-Lafuente
 
C
.
Antiarrhythmics for maintaining sinus rhythm after cardioversion of atrial fibrillation
.
Cochrane Database Syst Rev
 
2019
;
2019
:
Cd005049
.

578

Crijns
 
HJ
,
Van Gelder
 
IC
,
Van Gilst
 
WH
,
Hillege
 
H
,
Gosselink
 
AM
,
Lie
 
KI
.
Serial antiarrhythmic drug treatment to maintain sinus rhythm after electrical cardioversion for chronic atrial fibrillation or atrial flutter
.
Am J Cardiol
 
1991
;
68
:
335
41
.

579

Chatterjee
 
S
,
Sardar
 
P
,
Lichstein
 
E
,
Mukherjee
 
D
,
Aikat
 
S
.
Pharmacologic rate versus rhythm-control strategies in atrial fibrillation: an updated comprehensive review and meta-analysis
.
Pacing Clin Electrophysiol
 
2013
;
36
:
122
33
.

580

Kotecha
 
D
,
Kirchhof
 
P
.
Rate and rhythm control have comparable effects on mortality and stroke in atrial fibrillation but better data are needed
.
Evid Based Med
 
2014
;
19
:
222
3
.

581

Piccini
 
JP
,
Hasselblad
 
V
,
Peterson
 
ED
,
Washam
 
JB
,
Califf
 
RM
,
Kong
 
DF
.
Comparative efficacy of dronedarone and amiodarone for the maintenance of sinus rhythm in patients with atrial fibrillation
.
J Am Coll Cardiol
 
2009
;
54
:
1089
95
.

582

Eckardt
 
L
,
Sehner
 
S
,
Suling
 
A
,
Borof
 
K
,
Breithardt
 
G
,
Crijns
 
H
, et al.  
Attaining sinus rhythm mediates improved outcome with early rhythm control therapy of atrial fibrillation: the EAST-AFNET 4 trial
.
Eur Heart J
 
2022
;
43
:
4127
44
.

583

Freemantle
 
N
,
Lafuente-Lafuente
 
C
,
Mitchell
 
S
,
Eckert
 
L
,
Reynolds
 
M
.
Mixed treatment comparison of dronedarone, amiodarone, sotalol, flecainide, and propafenone, for the management of atrial fibrillation
.
Europace
 
2011
;
13
:
329
45
.

584

Frommeyer
 
G
,
Eckardt
 
L
.
Drug-induced proarrhythmia: risk factors and electrophysiological mechanisms
.
Nat Rev Cardiol
 
2016
;
13
:
36
47
.

585

Kochiadakis
 
GE
,
Marketou
 
ME
,
Igoumenidis
 
NE
,
Chrysostomakis
 
SI
,
Mavrakis
 
HE
,
Kaleboubas
 
MD
, et al.  
Amiodarone, sotalol, or propafenone in atrial fibrillation: which is preferred to maintain normal sinus rhythm?
 
Pacing Clin Electrophysiol
 
2000
;
23
:
1883
7
.

586

Roy
 
D
,
Talajic
 
M
,
Dorian
 
P
,
Connolly
 
S
,
Eisenberg
 
MJ
,
Green
 
M
, et al.  
Amiodarone to prevent recurrence of atrial fibrillation. Canadian Trial of Atrial Fibrillation Investigators
.
N Engl J Med
 
2000
;
342
:
913
20
.

587

Singh
 
BN
,
Singh
 
SN
,
Reda
 
DJ
,
Tang
 
XC
,
Lopez
 
B
,
Harris
 
CL
, et al.  
Amiodarone versus sotalol for atrial fibrillation
.
N Engl J Med
 
2005
;
352
:
1861
72
.

588

Ehrlich
 
JR
,
Look
 
C
,
Kostev
 
K
,
Israel
 
CW
,
Goette
 
A
.
Impact of dronedarone on the risk of myocardial infarction and stroke in atrial fibrillation patients followed in general practices in Germany
.
Int J Cardiol
 
2019
;
278
:
126
32
.

589

Singh
 
BN
,
Connolly
 
SJ
,
Crijns
 
HJ
,
Roy
 
D
,
Kowey
 
PR
,
Capucci
 
A
, et al.  
Dronedarone for maintenance of sinus rhythm in atrial fibrillation or flutter
.
N Engl J Med
 
2007
;
357
:
987
99
.

590

Stroobandt
 
R
,
Stiels
 
B
,
Hoebrechts
 
R
.
Propafenone for conversion and prophylaxis of atrial fibrillation. Propafenone Atrial Fibrillation Trial Investigators
.
Am J Cardiol
 
1997
;
79
:
418
23
.

591

Wazni
 
OM
,
Dandamudi
 
G
,
Sood
 
N
,
Hoyt
 
R
,
Tyler
 
J
,
Durrani
 
S
, et al.  
Cryoballoon ablation as initial therapy for atrial fibrillation
.
N Engl J Med
 
2021
;
384
:
316
24
.

592

Cosedis Nielsen
 
J
,
Johannessen
 
A
,
Raatikainen
 
P
,
Hindricks
 
G
,
Walfridsson
 
H
,
Kongstad
 
O
, et al.  
Radiofrequency ablation as initial therapy in paroxysmal atrial fibrillation
.
N Engl J Med
 
2012
;
367
:
1587
95
.

593

Morillo
 
CA
,
Verma
 
A
,
Connolly
 
SJ
,
Kuck
 
KH
,
Nair
 
GM
,
Champagne
 
J
, et al.  
Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of paroxysmal atrial fibrillation (RAAFT-2): a randomized trial
.
JAMA
 
2014
;
311
:
692
700
.

594

Kuniss
 
M
,
Pavlovic
 
N
,
Velagic
 
V
,
Hermida
 
JS
,
Healey
 
S
,
Arena
 
G
, et al.  
Cryoballoon ablation vs. antiarrhythmic drugs: first-line therapy for patients with paroxysmal atrial fibrillation
.
Europace
 
2021
;
23
:
1033
41
.

595

Hocini
 
M
,
Sanders
 
P
,
Deisenhofer
 
I
,
Jais
 
P
,
Hsu
 
LF
,
Scavee
 
C
, et al.  
Reverse remodeling of sinus node function after catheter ablation of atrial fibrillation in patients with prolonged sinus pauses
.
Circulation
 
2003
;
108
:
1172
5
.

596

Inada
 
K
,
Yamane
 
T
,
Tokutake
 
K
,
Yokoyama
 
K
,
Mishima
 
T
,
Hioki
 
M
, et al.  
The role of successful catheter ablation in patients with paroxysmal atrial fibrillation and prolonged sinus pauses: outcome during a 5-year follow-up
.
Europace
 
2014
;
16
:
208
13
.

597

Chen
 
YW
,
Bai
 
R
,
Lin
 
T
,
Salim
 
M
,
Sang
 
CH
,
Long
 
DY
, et al.  
Pacing or ablation: which is better for paroxysmal atrial fibrillation-related tachycardia-bradycardia syndrome?
 
Pacing Clin Electrophysiol
 
2014
;
37
:
403
11
.

598

Zhang
 
R
,
Wang
 
Y
,
Yang
 
M
,
Yang
 
Y
,
Wang
 
Z
,
Yin
 
X
, et al.  
Risk stratification for atrial fibrillation and outcomes in tachycardia-bradycardia syndrome: ablation vs. pacing
.
Front Cardiovasc Med
 
2021
;
8
:
674471
.

599

Oral
 
H
,
Pappone
 
C
,
Chugh
 
A
,
Good
 
E
,
Bogun
 
F
,
Pelosi
 
F
, Jr
, et al.  
Circumferential pulmonary-vein ablation for chronic atrial fibrillation
.
N Engl J Med
 
2006
;
354
:
934
41
.

600

Yan Huo
 
TG
,
Schönbauer
 
R
,
Wójcik
 
M
,
Fiedler
 
L
,
Roithinger
 
FX
,
Martinek
 
M
, et al.  
Low-voltage myocardium-guided ablation trial of persistent atrial fibrillation
.
NEJM Evid
 
2022
;
1
:EVIDoa2200141. .

601

Reddy
 
VY
,
Gerstenfeld
 
EP
,
Natale
 
A
,
Whang
 
W
,
Cuoco
 
FA
,
Patel
 
C
, et al.  
Pulsed field or conventional thermal ablation for paroxysmal atrial fibrillation
.
N Engl J Med
 
2023
;
389
:
1660
71
.

602

Kalman
 
JM
,
Al-Kaisey
 
AM
,
Parameswaran
 
R
,
Hawson
 
J
,
Anderson
 
RD
,
Lim
 
M
, et al.  
Impact of early vs. delayed atrial fibrillation catheter ablation on atrial arrhythmia recurrences
.
Eur Heart J
 
2023
;
44
:
2447
54
.

603

Kalman
 
JM
,
Sanders
 
P
,
Rosso
 
R
,
Calkins
 
H
.
Should we perform catheter ablation for asymptomatic atrial fibrillation?
 
Circulation
 
2017
;
136
:
490
9
.

604

Hunter
 
RJ
,
Berriman
 
TJ
,
Diab
 
I
,
Kamdar
 
R
,
Richmond
 
L
,
Baker
 
V
, et al.  
A randomized controlled trial of catheter ablation versus medical treatment of atrial fibrillation in heart failure (the CAMTAF trial)
.
Circ Arrhythm Electrophysiol
 
2014
;
7
:
31
8
.

605

Jones
 
DG
,
Haldar
 
SK
,
Hussain
 
W
,
Sharma
 
R
,
Francis
 
DP
,
Rahman-Haley
 
SL
, et al.  
A randomized trial to assess catheter ablation versus rate control in the management of persistent atrial fibrillation in heart failure
.
J Am Coll Cardiol
 
2013
;
61
:
1894
903
.

606

Kuck
 
KH
,
Merkely
 
B
,
Zahn
 
R
,
Arentz
 
T
,
Seidl
 
K
,
Schluter
 
M
, et al.  
Catheter ablation versus best medical therapy in patients with persistent atrial fibrillation and congestive heart failure: the randomized AMICA trial
.
Circ Arrhythm Electrophysiol
 
2019
;
12
:
e007731
.

607

MacDonald
 
MR
,
Connelly
 
DT
,
Hawkins
 
NM
,
Steedman
 
T
,
Payne
 
J
,
Shaw
 
M
, et al.  
Radiofrequency ablation for persistent atrial fibrillation in patients with advanced heart failure and severe left ventricular systolic dysfunction: a randomised controlled trial
.
Heart
 
2011
;
97
:
740
7
.

608

Parkash
 
R
,
Wells
 
GA
,
Rouleau
 
J
,
Talajic
 
M
,
Essebag
 
V
,
Skanes
 
A
, et al.  
Randomized ablation-based rhythm-control versus rate-control trial in patients with heart failure and atrial fibrillation: results from the RAFT-AF trial
.
Circulation
 
2022
;
145
:
1693
704
.

609

Romero
 
J
,
Gabr
 
M
,
Alviz
 
I
,
Briceno
 
D
,
Diaz
 
JC
,
Rodriguez
 
D
, et al.  
Improved survival in patients with atrial fibrillation and heart failure undergoing catheter ablation compared to medical treatment: a systematic review and meta-analysis of randomized controlled trials
.
J Cardiovasc Electrophysiol
 
2022
;
33
:
2356
66
.

610

Chen
 
S
,
Purerfellner
 
H
,
Meyer
 
C
,
Acou
 
WJ
,
Schratter
 
A
,
Ling
 
Z
, et al.  
Rhythm control for patients with atrial fibrillation complicated with heart failure in the contemporary era of catheter ablation: a stratified pooled analysis of randomized data
.
Eur Heart J
 
2020
;
41
:
2863
73
.

611

Prabhu
 
S
,
Taylor
 
AJ
,
Costello
 
BT
,
Kaye
 
DM
,
McLellan
 
AJA
,
Voskoboinik
 
A
, et al.  
Catheter ablation versus medical rate control in atrial fibrillation and systolic dysfunction: the CAMERA-MRI study
.
J Am Coll Cardiol
 
2017
;
70
:
1949
61
.

612

Packer
 
DL
,
Piccini
 
JP
,
Monahan
 
KH
,
Al-Khalidi
 
HR
,
Silverstein
 
AP
,
Noseworthy
 
PA
, et al.  
Ablation versus drug therapy for atrial fibrillation in heart failure: results from the CABANA trial
.
Circulation
 
2021
;
143
:
1377
90
.

613

Smit
 
MD
,
Moes
 
ML
,
Maass
 
AH
,
Achekar
 
ID
,
Van Geel
 
PP
,
Hillege
 
HL
, et al.  
The importance of whether atrial fibrillation or heart failure develops first
.
Eur J Heart Fail
 
2012
;
14
:
1030
40
.

614

Sohns
 
C
,
Zintl
 
K
,
Zhao
 
Y
,
Dagher
 
L
,
Andresen
 
D
,
Siebels
 
J
, et al.  
Impact of left ventricular function and heart failure symptoms on outcomes post ablation of atrial fibrillation in heart failure: CASTLE-AF trial
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e008461
.

615

Sugumar
 
H
,
Prabhu
 
S
,
Costello
 
B
,
Chieng
 
D
,
Azzopardi
 
S
,
Voskoboinik
 
A
, et al.  
Catheter ablation versus medication in atrial fibrillation and systolic dysfunction: late outcomes of CAMERA-MRI study
.
JACC Clin Electrophysiol
 
2020
;
6
:
1721
31
.

616

Kirstein
 
B
,
Neudeck
 
S
,
Gaspar
 
T
,
Piorkowski
 
J
,
Wechselberger
 
S
,
Kronborg
 
MB
, et al.  
Left atrial fibrosis predicts left ventricular ejection fraction response after atrial fibrillation ablation in heart failure patients: the fibrosis-HF study
.
Europace
 
2020
;
22
:
1812
21
.

617

Ishiguchi
 
H
,
Yoshiga
 
Y
,
Shimizu
 
A
,
Ueyama
 
T
,
Fukuda
 
M
,
Kato
 
T
, et al.  
Long-term events following catheter-ablation for atrial fibrillation in heart failure with preserved ejection fraction
.
ESC Heart Fail
 
2022
;
9
:
3505
18
.

618

Gu
 
G
,
Wu
 
J
,
Gao
 
X
,
Liu
 
M
,
Jin
 
C
,
Xu
 
Y
.
Catheter ablation of atrial fibrillation in patients with heart failure and preserved ejection fraction: a meta-analysis
.
Clin Cardiol
 
2022
;
45
:
786
93
.

619

Yamauchi
 
R
,
Morishima
 
I
,
Okumura
 
K
,
Kanzaki
 
Y
,
Morita
 
Y
,
Takagi
 
K
, et al.  
Catheter ablation for non-paroxysmal atrial fibrillation accompanied by heart failure with preserved ejection fraction: feasibility and benefits in functions and B-type natriuretic peptide
.
Europace
 
2021
;
23
:
1252
61
.

620

Rordorf
 
R
,
Scazzuso
 
F
,
Chun
 
KRJ
,
Khelae
 
SK
,
Kueffer
 
FJ
,
Braegelmann
 
KM
, et al.  
Cryoballoon ablation for the treatment of atrial fibrillation in patients with concomitant heart failure and either reduced or preserved left ventricular ejection fraction: results from the Cryo AF global registry
.
J Am Heart Assoc
 
2021
;
10
:
e021323
.

621

Cha
 
YM
,
Wokhlu
 
A
,
Asirvatham
 
SJ
,
Shen
 
WK
,
Friedman
 
PA
,
Munger
 
TM
, et al.  
Success of ablation for atrial fibrillation in isolated left ventricular diastolic dysfunction: a comparison to systolic dysfunction and normal ventricular function
.
Circ Arrhythm Electrophysiol
 
2011
;
4
:
724
32
.

622

Machino-Ohtsuka
 
T
,
Seo
 
Y
,
Ishizu
 
T
,
Sugano
 
A
,
Atsumi
 
A
,
Yamamoto
 
M
, et al.  
Efficacy, safety, and outcomes of catheter ablation of atrial fibrillation in patients with heart failure with preserved ejection fraction
.
J Am Coll Cardiol
 
2013
;
62
:
1857
65
.

623

Aldaas
 
OM
,
Lupercio
 
F
,
Darden
 
D
,
Mylavarapu
 
PS
,
Malladi
 
CL
,
Han
 
FT
, et al.  
Meta-analysis of the usefulness of catheter ablation of atrial fibrillation in patients with heart failure with preserved ejection fraction
.
Am J Cardiol
 
2021
;
142
:
66
73
.

624

Black-Maier
 
E
,
Ren
 
X
,
Steinberg
 
BA
,
Green
 
CL
,
Barnett
 
AS
,
Rosa
 
NS
, et al.  
Catheter ablation of atrial fibrillation in patients with heart failure and preserved ejection fraction
.
Heart Rhythm
 
2018
;
15
:
651
7
.

625

von Olshausen
 
G
,
Benson
 
L
,
Dahlström
 
U
,
Lund
 
LH
,
Savarese
 
G
,
Braunschweig
 
F
.
Catheter ablation for patients with atrial fibrillation and heart failure: insights from the Swedish Heart Failure Registry
.
Eur J Heart Fail
 
2022
;
24
:
1636
46
.

626

Shiraishi
 
Y
,
Kohsaka
 
S
,
Ikemura
 
N
,
Kimura
 
T
,
Katsumata
 
Y
,
Tanimoto
 
K
, et al.  
Catheter ablation for patients with atrial fibrillation and heart failure with reduced and preserved ejection fraction: insights from the KiCS-AF multicentre cohort study
.
Europace
 
2023
;
25
:
83
91
.

627

Wu
 
L
,
Narasimhan
 
B
,
Ho
 
KS
,
Zheng
 
Y
,
Shah
 
AN
,
Kantharia
 
BK
.
Safety and complications of catheter ablation for atrial fibrillation: predictors of complications from an updated analysis the national inpatient database
.
J Cardiovasc Electrophysiol
 
2021
;
32
:
1024
34
.

628

Tripathi
 
B
,
Arora
 
S
,
Kumar
 
V
,
Abdelrahman
 
M
,
Lahewala
 
S
,
Dave
 
M
, et al.  
Temporal trends of in-hospital complications associated with catheter ablation of atrial fibrillation in the United States: an update from nationwide inpatient sample database (2011–2014)
.
J Cardiovasc Electrophysiol
 
2018
;
29
:
715
24
.

629

Steinbeck
 
G
,
Sinner
 
MF
,
Lutz
 
M
,
Muller-Nurasyid
 
M
,
Kaab
 
S
,
Reinecke
 
H
.
Incidence of complications related to catheter ablation of atrial fibrillation and atrial flutter: a nationwide in-hospital analysis of administrative data for Germany in 2014
.
Eur Heart J
 
2018
;
39
:
4020
9
.

630

Deshmukh
 
A
,
Patel
 
NJ
,
Pant
 
S
,
Shah
 
N
,
Chothani
 
A
,
Mehta
 
K
, et al.  
In-hospital complications associated with catheter ablation of atrial fibrillation in the United States between 2000 and 2010: analysis of 93 801 procedures
.
Circulation
 
2013
;
128
:
2104
12
.

631

Cheng
 
EP
,
Liu
 
CF
,
Yeo
 
I
,
Markowitz
 
SM
,
Thomas
 
G
,
Ip
 
JE
, et al.  
Risk of mortality following catheter ablation of atrial fibrillation
.
J Am Coll Cardiol
 
2019
;
74
:
2254
64
.

632

Gawalko
 
M
,
Duncker
 
D
,
Manninger
 
M
,
van der Velden
 
RMJ
,
Hermans
 
ANL
,
Verhaert
 
DVM
, et al.  
The European TeleCheck-AF project on remote app-based management of atrial fibrillation during the COVID-19 pandemic: centre and patient experiences
.
Europace
 
2021
;
23
:
1003
15
.

633

Rizas
 
KD
,
Freyer
 
L
,
Sappler
 
N
,
von Stülpnagel
 
L
,
Spielbichler
 
P
,
Krasniqi
 
A
, et al.  
Smartphone-based screening for atrial fibrillation: a pragmatic randomized clinical trial
.
Nat Med
 
2022
;
28
:
1823
30
.

634

Andrade
 
JG
,
Champagne
 
J
,
Dubuc
 
M
,
Deyell
 
MW
,
Verma
 
A
,
Macle
 
L
, et al.  
Cryoballoon or radiofrequency ablation for atrial fibrillation assessed by continuous monitoring: a randomized clinical trial
.
Circulation
 
2019
;
140
:
1779
88
.

635

Duytschaever
 
M
,
Demolder
 
A
,
Phlips
 
T
,
Sarkozy
 
A
,
El Haddad
 
M
,
Taghji
 
P
, et al.  
PulmOnary vein isolation with vs. without continued antiarrhythmic Drug trEatment in subjects with Recurrent Atrial Fibrillation (POWDER AF): results from a multicentre randomized trial
.
Eur Heart J
 
2018
;
39
:
1429
37
.

636

Darkner
 
S
,
Chen
 
X
,
Hansen
 
J
,
Pehrson
 
S
,
Johannessen
 
A
,
Nielsen
 
JB
, et al.  
Recurrence of arrhythmia following short-term oral AMIOdarone after CATheter ablation for atrial fibrillation: a double-blind, randomized, placebo-controlled study (AMIO-CAT trial)
.
Eur Heart J
 
2014
;
35
:
3356
64
.

637

Leong-Sit
 
P
,
Roux
 
JF
,
Zado
 
E
,
Callans
 
DJ
,
Garcia
 
F
,
Lin
 
D
, et al.  
Antiarrhythmics after ablation of atrial fibrillation (5A study): six-month follow-up study
.
Circ Arrhythm Electrophysiol
 
2011
;
4
:
11
4
.

638

Kaitani
 
K
,
Inoue
 
K
,
Kobori
 
A
,
Nakazawa
 
Y
,
Ozawa
 
T
,
Kurotobi
 
T
, et al.  
Efficacy of antiarrhythmic drugs short-term use after catheter ablation for atrial fibrillation (EAST-AF) trial
.
Eur Heart J
 
2016
;
37
:
610
8
.

639

Noseworthy
 
PA
,
Van Houten
 
HK
,
Sangaralingham
 
LR
,
Deshmukh
 
AJ
,
Kapa
 
S
,
Mulpuru
 
SK
, et al.  
Effect of antiarrhythmic drug initiation on readmission after catheter ablation for atrial fibrillation
.
JACC Clin Electrophysiol
 
2015
;
1
:
238
44
.

640

Xu
 
X
,
Alida
 
CT
,
Yu
 
B
.
Administration of antiarrhythmic drugs to maintain sinus rhythm after catheter ablation for atrial fibrillation: a meta-analysis
.
Cardiovasc Ther
 
2015
;
33
:
242
6
.

641

Chen
 
W
,
Liu
 
H
,
Ling
 
Z
,
Xu
 
Y
,
Fan
 
J
,
Du
 
H
, et al.  
Efficacy of short-term antiarrhythmic drugs use after catheter ablation of atrial fibrillation—a systematic review with meta-analyses and trial sequential analyses of randomized controlled trials
.
PLoS One
 
2016
;
11
:
e0156121
.

642

Schleberger
 
R
,
Metzner
 
A
,
Kuck
 
KH
,
Andresen
 
D
,
Willems
 
S
,
Hoffmann
 
E
, et al.  
Antiarrhythmic drug therapy after catheter ablation for atrial fibrillation–insights from the German Ablation Registry
.
Pharmacol Res Perspect
 
2021
;
9
:
e00880
.

643

Zhang
 
XD
,
Gu
 
J
,
Jiang
 
WF
,
Zhao
 
L
,
Zhou
 
L
,
Wang
 
YL
, et al.  
Optimal rhythm-control strategy for recurrent atrial tachycardia after catheter ablation of persistent atrial fibrillation: a randomized clinical trial
.
Eur Heart J
 
2014
;
35
:
1327
34
.

644

Zhou
 
L
,
He
 
L
,
Wang
 
W
,
Li
 
C
,
Li
 
S
,
Tang
 
R
, et al.  
Effect of repeat catheter ablation vs. antiarrhythmic drug therapy among patients with recurrent atrial tachycardia/atrial fibrillation after atrial fibrillation catheter ablation: data from CHINA-AF registry
.
Europace
 
2023
;
25
:
382
9
.

645

Fink
 
T
,
Metzner
 
A
,
Willems
 
S
,
Eckardt
 
L
,
Ince
 
H
,
Brachmann
 
J
, et al.  
Procedural success, safety and patients satisfaction after second ablation of atrial fibrillation in the elderly: results from the German ablation registry
.
Clin Res Cardiol
 
2019
;
108
:
1354
63
.

646

Winkle
 
RA
,
Mead
 
RH
,
Engel
 
G
,
Kong
 
MH
,
Fleming
 
W
,
Salcedo
 
J
, et al.  
Impact of obesity on atrial fibrillation ablation: patient characteristics, long-term outcomes, and complications
.
Heart Rhythm
 
2017
;
14
:
819
27
.

647

Sticherling
 
C
,
Marin
 
F
,
Birnie
 
D
,
Boriani
 
G
,
Calkins
 
H
,
Dan
 
G-A
, et al.  
Antithrombotic management in patients undergoing electrophysiological procedures: a European Heart Rhythm Association (EHRA) position document endorsed by the ESC Working Group Thrombosis, Heart Rhythm Society (HRS), and Asia Pacific Heart Rhythm Society (APHRS)
.
EP Europace
 
2015
;
17
:
1197
214
.

648

Calkins
 
H
,
Kuck
 
KH
,
Cappato
 
R
,
Brugada
 
J
,
Camm
 
AJ
,
Chen
 
SA
, et al.  
2012 HRS/EHRA/ECAS expert consensus statement on catheter and surgical ablation of atrial fibrillation: recommendations for patient selection, procedural techniques, patient management and follow-up, definitions, endpoints, and research trial design
.
J Interv Card Electrophysiol
 
2012
;
33
:
171
257
.

649

Noubiap
 
JJ
,
Agbaedeng
 
TA
,
Ndoadoumgue
 
AL
,
Nyaga
 
UF
,
Kengne
 
AP
.
Atrial thrombus detection on transoesophageal echocardiography in patients with atrial fibrillation undergoing cardioversion or catheter ablation: a pooled analysis of rates and predictors
.
J Cardiovasc Electrophysiol
 
2021
;
32
:
2179
88
.

650

Lurie
 
A
,
Wang
 
J
,
Hinnegan
 
KJ
,
McIntyre
 
WF
,
Belley-Côté
 
EP
,
Amit
 
G
, et al.  
Prevalence of left atrial thrombus in anticoagulated patients with atrial fibrillation
.
J Am Coll Cardiol
 
2021
;
77
:
2875
86
.

651

Efremidis
 
M
,
Bazoukis
 
G
,
Vlachos
 
K
,
Prappa
 
E
,
Megarisiotou
 
A
,
Dragasis
 
S
, et al.  
Safety of catheter ablation of atrial fibrillation without pre- or peri-procedural imaging for the detection of left atrial thrombus in the era of uninterrupted anticoagulation
.
J Arrhythm
 
2021
;
37
:
28
32
.

652

Diab
 
M
,
Wazni
 
OM
,
Saliba
 
WI
,
Tarakji
 
KG
,
Ballout
 
JA
,
Hutt
 
E
, et al.  
Ablation of atrial fibrillation without left atrial appendage imaging in patients treated with direct oral anticoagulants
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e008301
.

653

Patel
 
K
,
Natale
 
A
,
Yang
 
R
,
Trivedi
 
C
,
Romero
 
J
,
Briceno
 
D
, et al.  
Is transesophageal echocardiography necessary in patients undergoing ablation of atrial fibrillation on an uninterrupted direct oral anticoagulant regimen? Results from a prospective multicenter registry
.
Heart Rhythm
 
2020
;
17
:
2093
9
.

654

Mao
 
YJ
,
Wang
 
H
,
Huang
 
PF
.
Meta-analysis of the safety and efficacy of using minimally interrupted novel oral anticoagulants in patients undergoing catheter ablation for atrial fibrillation
.
J Interv Card Electrophysiol
 
2021
;
60
:
407
17
.

655

van Vugt
 
SPG
,
Westra
 
SW
,
Volleberg
 
R
,
Hannink
 
G
,
Nakamura
 
R
,
de Asmundis
 
C
, et al.  
Meta-analysis of controlled studies on minimally interrupted vs. continuous use of non-vitamin K antagonist oral anticoagulants in catheter ablation for atrial fibrillation
.
Europace
 
2021
;
23
:
1961
9
.

656

Ge
 
Z
,
Faggioni
 
M
,
Baber
 
U
,
Sartori
 
S
,
Sorrentino
 
S
,
Farhan
 
S
, et al.  
Safety and efficacy of nonvitamin K antagonist oral anticoagulants during catheter ablation of atrial fibrillation: a systematic review and meta-analysis
.
Cardiovasc Ther
 
2018
;
36
:
e12457
.

657

Asad
 
ZUA
,
Akhtar
 
KH
,
Jafry
 
AH
,
Khan
 
MH
,
Khan
 
MS
,
Munir
 
MB
, et al.  
Uninterrupted versus interrupted direct oral anticoagulation for catheter ablation of atrial fibrillation: a systematic review and meta-analysis
.
J Cardiovasc Electrophysiol
 
2021
;
32
:
1995
2004
.

658

Mao
 
YJ
,
Wang
 
H
,
Huang
 
PF
.
Peri-procedural novel oral anticoagulants dosing strategy during atrial fibrillation ablation: a meta-analysis
.
Pacing Clin Electrophysiol
 
2020
;
43
:
1104
14
.

659

Basu-Ray
 
I
,
Khanra
 
D
,
Kupó
 
P
,
Bunch
 
J
,
Theus
 
SA
,
Mukherjee
 
A
, et al.  
Outcomes of uninterrupted vs interrupted periprocedural direct oral anticoagulants in atrial fibrillation ablation: a meta-analysis
.
J Arrhythm
 
2021
;
37
:
384
93
.

660

Romero
 
J
,
Cerrud-Rodriguez
 
RC
,
Diaz
 
JC
,
Rodriguez
 
D
,
Arshad
 
S
,
Alviz
 
I
, et al.  
Oral anticoagulation after catheter ablation of atrial fibrillation and the associated risk of thromboembolic events and intracranial hemorrhage: a systematic review and meta-analysis
.
J Cardiovasc Electrophysiol
 
2019
;
30
:
1250
7
.

661

Liu
 
XH
,
Xu
 
Q
,
Luo
 
T
,
Zhang
 
L
,
Liu
 
HJ
.
Discontinuation of oral anticoagulation therapy after successful atrial fibrillation ablation: a systematic review and meta-analysis of prospective studies
.
PLoS One
 
2021
;
16
:
e0253709
.

662

Proietti
 
R
,
AlTurki
 
A
,
Di Biase
 
L
,
China
 
P
,
Forleo
 
G
,
Corrado
 
A
, et al.  
Anticoagulation after catheter ablation of atrial fibrillation: an unnecessary evil? A systematic review and meta-analysis
.
J Cardiovasc Electrophysiol
 
2019
;
30
:
468
78
.

663

Maduray
 
K
,
Moneruzzaman
 
M
,
Changwe
 
GJ
,
Zhong
 
J
.
Benefits and risks associated with long-term oral anticoagulation after successful atrial fibrillation catheter ablation: systematic review and meta-analysis
.
Clin Appl Thromb Hemost
 
2022
;
28
:10760296221118480.

664

Brockmeyer
 
M
,
Lin
 
Y
,
Parco
 
C
,
Karathanos
 
A
,
Krieger
 
T
,
Schulze
 
V
, et al.  
Uninterrupted anticoagulation during catheter ablation for atrial fibrillation: no difference in major bleeding and stroke between direct oral anticoagulants and vitamin K antagonists in an updated meta-analysis of randomised controlled trials
.
Acta Cardiol
 
2021
;
76
:
288
95
.

665

Di Monaco
 
A
,
Guida
 
P
,
Vitulano
 
N
,
Quadrini
 
F
,
Troisi
 
F
,
Langialonga
 
T
, et al.  
Catheter ablation of atrial fibrillation with uninterrupted anticoagulation: a meta-analysis of six randomized controlled trials
.
J Cardiovasc Med (Hagerstown)
 
2020
;
21
:
483
90
.

666

Maesen
 
B
,
Luermans
 
J
,
Bidar
 
E
,
Chaldoupi
 
SM
,
Gelsomino
 
S
,
Maessen
 
JG
, et al.  
A hybrid approach to complex arrhythmias
.
Europace
 
2021
;
23
:
ii28
33
.

667

van der Heijden
 
CAJ
,
Vroomen
 
M
,
Luermans
 
JG
,
Vos
 
R
,
Crijns
 
H
,
Gelsomino
 
S
, et al.  
Hybrid versus catheter ablation in patients with persistent and longstanding persistent atrial fibrillation: a systematic review and meta-analysis
.
Eur J Cardiothorac Surg
 
2019
;
56
:
433
43
.

668

Boersma
 
LV
,
Castella
 
M
,
van Boven
 
W
,
Berruezo
 
A
,
Yilmaz
 
A
,
Nadal
 
M
, et al.  
Atrial fibrillation catheter ablation versus surgical ablation treatment (FAST): a 2-center randomized clinical trial
.
Circulation
 
2012
;
125
:
23
30
.

669

Castella
 
M
,
Kotecha
 
D
,
van Laar
 
C
,
Wintgens
 
L
,
Castillo
 
Y
,
Kelder
 
J
, et al.  
Thoracoscopic vs. catheter ablation for atrial fibrillation: long-term follow-up of the FAST randomized trial
.
Europace
 
2019
;
21
:
746
53
.

670

van der Heijden
 
CAJ
,
Weberndörfer
 
V
,
Vroomen
 
M
,
Luermans
 
JG
,
Chaldoupi
 
SM
,
Bidar
 
E
, et al.  
Hybrid ablation versus repeated catheter ablation in persistent atrial fibrillation: a randomized controlled trial
.
JACC Clin Electrophysiol
 
2023
;
9
:
1013
23
.

671

DeLurgio
 
DB
,
Crossen
 
KJ
,
Gill
 
J
,
Blauth
 
C
,
Oza
 
SR
,
Magnano
 
AR
, et al.  
Hybrid convergent procedure for the treatment of persistent and long-standing persistent atrial fibrillation: results of CONVERGE clinical trial
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e009288
.

672

Pokushalov
 
E
,
Romanov
 
A
,
Elesin
 
D
,
Bogachev-Prokophiev
 
A
,
Losik
 
D
,
Bairamova
 
S
, et al.  
Catheter versus surgical ablation of atrial fibrillation after a failed initial pulmonary vein isolation procedure: a randomized controlled trial
.
J Cardiovasc Electrophysiol
 
2013
;
24
:
1338
43
.

673

Haldar
 
S
,
Khan
 
HR
,
Boyalla
 
V
,
Kralj-Hans
 
I
,
Jones
 
S
,
Lord
 
J
, et al.  
Catheter ablation vs. thoracoscopic surgical ablation in long-standing persistent atrial fibrillation: CASA-AF randomized controlled trial
.
Eur Heart J
 
2020
;
41
:
4471
80
.

674

Doll
 
N
,
Weimar
 
T
,
Kosior
 
DA
,
Bulava
 
A
,
Mokracek
 
A
,
Mönnig
 
G
, et al.  
Efficacy and safety of hybrid epicardial and endocardial ablation versus endocardial ablation in patients with persistent and longstanding persistent atrial fibrillation: a randomised, controlled trial
.
EClinicalMedicine
 
2023
;
61
:
102052
.

675

Malaisrie
 
SC
,
McCarthy
 
PM
,
Kruse
 
J
,
Matsouaka
 
R
,
Andrei
 
AC
,
Grau-Sepulveda
 
MV
, et al.  
Burden of preoperative atrial fibrillation in patients undergoing coronary artery bypass grafting
.
J Thorac Cardiovasc Surg
 
2018
;
155
:
2358
2367 e1
.

676

Saxena
 
A
,
Dinh
 
DT
,
Reid
 
CM
,
Smith
 
JA
,
Shardey
 
GC
,
Newcomb
 
AE
.
Does preoperative atrial fibrillation portend a poorer prognosis in patients undergoing isolated aortic valve replacement? A multicentre Australian study
.
Can J Cardiol
 
2013
;
29
:
697
703
.

677

Quader
 
MA
,
McCarthy
 
PM
,
Gillinov
 
AM
,
Alster
 
JM
,
Cosgrove
 
DM
, 3rd
,
Lytle
 
BW
, et al.  
Does preoperative atrial fibrillation reduce survival after coronary artery bypass grafting?
 
Ann Thorac Surg
 
2004
;
77
:
1514
22
; discussion 1522–4.

678

Damiano
 
RJ
, Jr,
Schwartz
 
FH
,
Bailey
 
MS
,
Maniar
 
HS
,
Munfakh
 
NA
,
Moon
 
MR
, et al.  
The Cox Maze IV procedure: predictors of late recurrence
.
J Thorac Cardiovasc Surg
 
2011
;
141
:
113
21
.

679

Cox
 
JL
,
Schuessler
 
RB
,
Boineau
 
JP
.
The development of the Maze procedure for the treatment of atrial fibrillation
.
Semin Thorac Cardiovasc Surg
 
2000
;
12
:
2
14
.

680

Melby
 
SJ
,
Zierer
 
A
,
Bailey
 
MS
,
Cox
 
JL
,
Lawton
 
JS
,
Munfakh
 
N
, et al.  
A new era in the surgical treatment of atrial fibrillation: the impact of ablation technology and lesion set on procedural efficacy
.
Ann Surg
 
2006
;
244
:
583
92
.

681

Badhwar
 
V
,
Rankin
 
JS
,
Damiano
 
RJ
, Jr
,
Gillinov
 
AM
,
Bakaeen
 
FG
,
Edgerton
 
JR
, et al.  
The Society of Thoracic Surgeons 2017 clinical practice guidelines for the surgical treatment of atrial fibrillation
.
Ann Thorac Surg
 
2017
;
103
:
329
41
.

682

Ad
 
N
,
Henry
 
L
,
Hunt
 
S
,
Holmes
 
SD
.
Impact of clinical presentation and surgeon experience on the decision to perform surgical ablation
.
Ann Thorac Surg
 
2013
;
96
:
763
8
; discussion 768–9.

683

Cheng
 
DC
,
Ad
 
N
,
Martin
 
J
,
Berglin
 
EE
,
Chang
 
BC
,
Doukas
 
G
, et al.  
Surgical ablation for atrial fibrillation in cardiac surgery: a meta-analysis and systematic review
.
Innovations (Phila)
 
2010
;
5
:
84
96
.

684

McClure
 
GR
,
Belley-Cote
 
EP
,
Jaffer
 
IH
,
Dvirnik
 
N
,
An
 
KR
,
Fortin
 
G
, et al.  
Surgical ablation of atrial fibrillation: a systematic review and meta-analysis of randomized controlled trials
.
Europace
 
2018
;
20
:
1442
50
.

685

Phan
 
K
,
Xie
 
A
,
La Meir
 
M
,
Black
 
D
,
Yan
 
TD
.
Surgical ablation for treatment of atrial fibrillation in cardiac surgery: a cumulative meta-analysis of randomised controlled trials
.
Heart
 
2014
;
100
:
722
30
.

686

Barnett
 
SD
,
Ad
 
N
.
Surgical ablation as treatment for the elimination of atrial fibrillation: a meta-analysis
.
J Thorac Cardiovasc Surg
 
2006
;
131
:
1029
35
.

687

Gillinov
 
AM
,
Gelijns
 
AC
,
Parides
 
MK
,
DeRose
 
JJ
, Jr
,
Moskowitz
 
AJ
,
Voisine
 
P
, et al.  
Surgical ablation of atrial fibrillation during mitral-valve surgery
.
N Engl J Med
 
2015
;
372
:
1399
409
.

688

MacGregor
 
RM
,
Bakir
 
NH
,
Pedamallu
 
H
,
Sinn
 
LA
,
Maniar
 
HS
,
Melby
 
SJ
, et al.  
Late results after stand-alone surgical ablation for atrial fibrillation
.
J Thorac Cardiovasc Surg
 
2022
;
164
:
1515
1528.e8
.

689

Musharbash
 
FN
,
Schill
 
MR
,
Sinn
 
LA
,
Schuessler
 
RB
,
Maniar
 
HS
,
Moon
 
MR
, et al.  
Performance of the Cox-Maze IV procedure is associated with improved long-term survival in patients with atrial fibrillation undergoing cardiac surgery
.
J Thorac Cardiovasc Surg
 
2018
;
155
:
159
70
.

690

Rankin
 
JS
,
Lerner
 
DJ
,
Braid-Forbes
 
MJ
,
McCrea
 
MM
,
Badhwar
 
V
.
Surgical ablation of atrial fibrillation concomitant to coronary-artery bypass grafting provides cost-effective mortality reduction
.
J Thorac Cardiovasc Surg
 
2020
;
160
:
675
686 e13
.

691

Suwalski
 
P
,
Kowalewski
 
M
,
Jasinski
 
M
,
Staromlynski
 
J
,
Zembala
 
M
,
Widenka
 
K
, et al.  
Survival after surgical ablation for atrial fibrillation in mitral valve surgery: analysis from the Polish National Registry of Cardiac Surgery Procedures (KROK)
.
J Thorac Cardiovasc Surg
 
2019
;
157
:
1007
1018 e4
.

692

Suwalski
 
P
,
Kowalewski
 
M
,
Jasinski
 
M
,
Staromlynski
 
J
,
Zembala
 
M
,
Widenka
 
K
, et al.  
Surgical ablation for atrial fibrillation during isolated coronary artery bypass surgery
.
Eur J Cardiothorac Surg
 
2020
;
57
:
691
700
.

693

Wehbe
 
M
,
Albert
 
M
,
Lewalter
 
T
,
Ouarrak
 
T
,
Senges
 
J
,
Hanke
 
T
, et al.  
The German cardiosurgery atrial fibrillation registry: 1-year follow-up outcomes
.
Thorac Cardiovasc Surg
 
2023
;
71
:
255
63
.

694

Kim
 
HJ
,
Kim
 
YJ
,
Kim
 
M
,
Yoo
 
JS
,
Kim
 
DH
,
Park
 
DW
, et al.  
Surgical ablation for atrial fibrillation during aortic and mitral valve surgery: a nationwide population-based cohort study
.
J Thorac Cardiovasc Surg
 
2024
;
167
:
981
93
.

695

Ad
 
N
,
Henry
 
L
,
Hunt
 
S
,
Holmes
 
SD
.
Do we increase the operative risk by adding the Cox Maze III procedure to aortic valve replacement and coronary artery bypass surgery?
 
J Thorac Cardiovasc Surg
 
2012
;
143
:
936
44
.

696

Maesen
 
B
,
van der Heijden
 
CAJ
,
Bidar
 
E
,
Vos
 
R
,
Athanasiou
 
T
,
Maessen
 
JG
.
Patient-reported quality of life after stand-alone and concomitant arrhythmia surgery: a systematic review and meta-analysis
.
Interact Cardiovasc Thorac Surg
 
2022
;
34
:
339
48
.

697

Osmancik
 
P
,
Budera
 
P
,
Talavera
 
D
,
Hlavicka
 
J
,
Herman
 
D
,
Holy
 
J
, et al.  
Five-year outcomes in cardiac surgery patients with atrial fibrillation undergoing concomitant surgical ablation versus no ablation. The long-term follow-up of the PRAGUE-12 study
.
Heart Rhythm
 
2019
;
16
:
1334
40
.

698

Lee
 
R
,
Jivan
 
A
,
Kruse
 
J
,
McGee
 
EC
, Jr
,
Malaisrie
 
SC
,
Bernstein
 
R
, et al.  
Late neurologic events after surgery for atrial fibrillation: rare but relevant
.
Ann Thorac Surg
 
2013
;
95
:
126
31
; discussion 131–2.

699

Kowalewski
 
M
,
Pasierski
 
M
,
Kołodziejczak
 
M
,
Litwinowicz
 
R
,
Kowalówka
 
A
,
Wańha
 
W
, et al.  
Atrial fibrillation ablation improves late survival after concomitant cardiac surgery
.
J Thorac Cardiovasc Surg
 
2023
;
166
:
1656
1668.e8
.

700

Cox
 
JL
,
Ad
 
N
,
Palazzo
 
T
.
Impact of the maze procedure on the stroke rate in patients with atrial fibrillation
.
J Thorac Cardiovasc Surg
 
1999
;
118
:
833
40
.

701

Huffman
 
MD
,
Karmali
 
KN
,
Berendsen
 
MA
,
Andrei
 
AC
,
Kruse
 
J
,
McCarthy
 
PM
, et al.  
Concomitant atrial fibrillation surgery for people undergoing cardiac surgery
.
Cochrane Database Syst Rev
 
2016
;8:
CD011814
.

702

Kowalewski
 
M
,
Pasierski
 
M
,
Finke
 
J
,
Kolodziejczak
 
M
,
Staromlynski
 
J
,
Litwinowicz
 
R
, et al.  
Permanent pacemaker implantation after valve and arrhythmia surgery in patients with preoperative atrial fibrillation
.
Heart Rhythm
 
2022
;
19
:
1442
9
.

703

Pokushalov
 
E
,
Romanov
 
A
,
Corbucci
 
G
,
Cherniavsky
 
A
,
Karaskov
 
A
.
Benefit of ablation of first diagnosed paroxysmal atrial fibrillation during coronary artery bypass grafting: a pilot study
.
Eur J Cardiothorac Surg
 
2012
;
41
:
556
60
.

704

Yoo
 
JS
,
Kim
 
JB
,
Ro
 
SK
,
Jung
 
Y
,
Jung
 
SH
,
Choo
 
SJ
, et al.  
Impact of concomitant surgical atrial fibrillation ablation in patients undergoing aortic valve replacement
.
Circ J
 
2014
;
78
:
1364
71
.

705

Malaisrie
 
SC
,
Lee
 
R
,
Kruse
 
J
,
Lapin
 
B
,
Wang
 
EC
,
Bonow
 
RO
, et al.  
Atrial fibrillation ablation in patients undergoing aortic valve replacement
.
J Heart Valve Dis
 
2012
;
21
:
350
7
.

706

Rankin
 
JS
,
Lerner
 
DJ
,
Braid-Forbes
 
MJ
,
Ferguson
 
MA
,
Badhwar
 
V
.
One-year mortality and costs associated with surgical ablation for atrial fibrillation concomitant to coronary artery bypass grafting
.
Eur J Cardiothorac Surg
 
2017
;
52
:
471
7
.

707

Schill
 
MR
,
Musharbash
 
FN
,
Hansalia
 
V
,
Greenberg
 
JW
,
Melby
 
SJ
,
Maniar
 
HS
, et al.  
Late results of the Cox-Maze IV procedure in patients undergoing coronary artery bypass grafting
.
J Thorac Cardiovasc Surg
 
2017
;
153
:
1087
94
.

708

Gupta
 
D
,
Ding
 
WY
,
Calvert
 
P
,
Williams
 
E
,
Das
 
M
,
Tovmassian
 
L
, et al.  
Cryoballoon pulmonary vein isolation as first-line treatment for typical atrial flutter
.
Heart
 
2023
;
109
:
364
71
.

709

Steinberg
 
C
,
Champagne
 
J
,
Deyell
 
MW
,
Dubuc
 
M
,
Leong-Sit
 
P
,
Calkins
 
H
, et al.  
Prevalence and outcome of early recurrence of atrial tachyarrhythmias in the cryoballoon vs irrigated radiofrequency catheter ablation (CIRCA-DOSE) study
.
Heart Rhythm
 
2021
;
18
:
1463
70
.

710

Heijman
 
J
,
Linz
 
D
,
Schotten
 
U
.
Dynamics of atrial fibrillation mechanisms and comorbidities
.
Annu Rev Physiol
 
2021
;
83
:
83
106
.

711

Fabritz
 
L
,
Crijns
 
H
,
Guasch
 
E
,
Goette
 
A
,
Hausler
 
KG
,
Kotecha
 
D
, et al.  
Dynamic risk assessment to improve quality of care in patients with atrial fibrillation: the 7th AFNET/EHRA consensus conference
.
Europace
 
2021
;
23
:
329
44
.

712

Brandes
 
A
,
Smit
 
MD
,
Nguyen
 
BO
,
Rienstra
 
M
,
Van Gelder
 
IC
.
Risk factor management in atrial fibrillation
.
Arrhythm Electrophysiol Rev
 
2018
;
7
:
118
27
.

713

Pokorney
 
SD
,
Cocoros
 
N
,
Al-Khalidi
 
HR
,
Haynes
 
K
,
Li
 
S
,
Al-Khatib
 
SM
, et al.  
Effect of mailing educational material to patients with atrial fibrillation and their clinicians on use of oral anticoagulants: a randomized clinical trial
.
JAMA Netw Open
 
2022
;
5
:
e2214321
.

714

Ritchie
 
LA
,
Penson
 
PE
,
Akpan
 
A
,
Lip
 
GYH
,
Lane
 
DA
.
Integrated care for atrial fibrillation management: the role of the pharmacist
.
Am J Med
 
2022
;
135
:
1410
26
.

715

Guo
 
Y
,
Guo
 
J
,
Shi
 
X
,
Yao
 
Y
,
Sun
 
Y
,
Xia
 
Y
, et al.  
Mobile health technology-supported atrial fibrillation screening and integrated care: a report from the mAFA-II trial long-term extension cohort
.
Eur J Intern Med
 
2020
;
82
:
105
11
.

716

Yan
 
H
,
Du
 
YX
,
Wu
 
FQ
,
Lu
 
XY
,
Chen
 
RM
,
Zhang
 
Y
.
Effects of nurse-led multidisciplinary team management on cardiovascular hospitalization and quality of life in patients with atrial fibrillation: randomized controlled trial
.
Int J Nurs Stud
 
2022
;
127
:
104159
.

717

Stewart
 
S
,
Ball
 
J
,
Horowitz
 
JD
,
Marwick
 
TH
,
Mahadevan
 
G
,
Wong
 
C
, et al.  
Standard versus atrial fibrillation-specific management strategy (SAFETY) to reduce recurrent admission and prolong survival: pragmatic, multicentre, randomised controlled trial
.
Lancet
 
2015
;
385
:
775
84
.

718

Cox
 
JL
,
Parkash
 
R
,
Foster
 
GA
,
Xie
 
F
,
MacKillop
 
JH
,
Ciaccia
 
A
, et al.  
Integrated management program advancing community treatment of atrial fibrillation (IMPACT-AF): a cluster randomized trial of a computerized clinical decision support tool
.
Am Heart J
 
2020
;
224
:
35
46
.

719

Sposato
 
LA
,
Stirling
 
D
,
Saposnik
 
G
.
Therapeutic decisions in atrial fibrillation for stroke prevention: the role of aversion to ambiguity and physicians’ risk preferences
.
J Stroke Cerebrovasc Dis
 
2018
;
27
:
2088
95
.

720

Noseworthy
 
PA
,
Brito
 
JP
,
Kunneman
 
M
,
Hargraves
 
IG
,
Zeballos-Palacios
 
C
,
Montori
 
VM
, et al.  
Shared decision-making in atrial fibrillation: navigating complex issues in partnership with the patient
.
J Interv Card Electrophysiol
 
2019
;
56
:
159
63
.

721

Poorcheraghi
 
H
,
Negarandeh
 
R
,
Pashaeypoor
 
S
,
Jorian
 
J
.
Effect of using a mobile drug management application on medication adherence and hospital readmission among elderly patients with polypharmacy: a randomized controlled trial
.
BMC Health Serv Res
 
2023
;
23
:
1192
.

722

Kotecha
 
D
,
Chua
 
WWL
,
Fabritz
 
L
,
Hendriks
 
J
,
Casadei
 
B
,
Schotten
 
U
, et al.  
European Society of Cardiology (ESC) Atrial Fibrillation Guidelines Taskforce, the CATCH ME consortium, and the European Heart Rhythm Association (EHRA). European Society of Cardiology smartphone and tablet applications for patients with atrial fibrillation and their health care providers
.
Europace
 
2018
;
20
:
225
33
.

723

Bunting
 
KV
,
Gill
 
SK
,
Sitch
 
A
,
Mehta
 
S
,
O’Connor
 
K
,
Lip
 
GY
, et al.  
Improving the diagnosis of heart failure in patients with atrial fibrillation
.
Heart
 
2021
;
107
:
902
8
.

724

Donal
 
E
,
Lip
 
GY
,
Galderisi
 
M
,
Goette
 
A
,
Shah
 
D
,
Marwan
 
M
, et al.  
EACVI/EHRA expert consensus document on the role of multi-modality imaging for the evaluation of patients with atrial fibrillation
.
Eur Heart J Cardiovasc Imaging
 
2016
;
17
:
355
83
.

725

Bunting
 
KV
,
O’Connor
 
K
,
Steeds
 
RP
,
Kotecha
 
D
.
Cardiac imaging to assess left ventricular systolic function in atrial fibrillation
.
Am J Cardiol
 
2021
;
139
:
40
9
.

726

Timperley
 
J
,
Mitchell
 
AR
,
Becher
 
H
.
Contrast echocardiography for left ventricular opacification
.
Heart
 
2003
;
89
:
1394
7
.

727

Kotecha
 
D
,
Mohamed
 
M
,
Shantsila
 
E
,
Popescu
 
BA
,
Steeds
 
RP
.
Is echocardiography valid and reproducible in patients with atrial fibrillation? A systematic review
.
Europace
 
2017
;
19
:
1427
38
.

728

Quintana
 
RA
,
Dong
 
T
,
Vajapey
 
R
,
Reyaldeen
 
R
,
Kwon
 
DH
,
Harb
 
S
, et al.  
Preprocedural multimodality imaging in atrial fibrillation
.
Circ Cardiovasc Imaging
 
2022
;
15
:
e014386
.

729

Laubrock
 
K
,
von Loesch
 
T
,
Steinmetz
 
M
,
Lotz
 
J
,
Frahm
 
J
,
Uecker
 
M
, et al.  
Imaging of arrhythmia: real-time cardiac magnetic resonance imaging in atrial fibrillation
.
Eur J Radiol Open
 
2022
;
9
:
100404
.

730

Sciagrà
 
R
,
Sotgia
 
B
,
Boni
 
N
,
Pupi
 
A
.
Assessment of the influence of atrial fibrillation on gated SPECT perfusion data by comparison with simultaneously acquired nongated SPECT data
.
J Nucl Med
 
2008
;
49
:
1283
7
.

731

Clayton
 
B
,
Roobottom
 
C
,
Morgan-Hughes
 
G
.
CT coronary angiography in atrial fibrillation: a comparison of radiation dose and diagnostic confidence with retrospective gating vs prospective gating with systolic acquisition
.
Br J Radiol
 
2015
;
88
:
20150533
.

732

Thrall
 
G
,
Lane
 
D
,
Carroll
 
D
,
Lip
 
GY
.
Quality of life in patients with atrial fibrillation: a systematic review
.
Am J Med
 
2006
;
119
:
448.e1
19
.

733

Steinberg
 
BA
,
Dorian
 
P
,
Anstrom
 
KJ
,
Hess
 
R
,
Mark
 
DB
,
Noseworthy
 
PA
, et al.  
Patient-reported outcomes in atrial fibrillation research: results of a Clinicaltrials.gov analysis
.
JACC Clin Electrophysiol
 
2019
;
5
:
599
605
.

734

Potpara
 
TS
,
Mihajlovic
 
M
,
Zec
 
N
,
Marinkovic
 
M
,
Kovacevic
 
V
,
Simic
 
J
, et al.  
Self-reported treatment burden in patients with atrial fibrillation: quantification, major determinants, and implications for integrated holistic management of the arrhythmia
.
Europace
 
2020
;
22
:
1788
97
.

735

Moons
 
P
,
Norekvål
 
TM
,
Arbelo
 
E
,
Borregaard
 
B
,
Casadei
 
B
,
Cosyns
 
B
, et al.  
Placing patient-reported outcomes at the centre of cardiovascular clinical practice: implications for quality of care and management
.
Eur Heart J
 
2023
;
44
:
3405
22
.

736

Allan
 
KS
,
Aves
 
T
,
Henry
 
S
,
Banfield
 
L
,
Victor
 
JC
,
Dorian
 
P
, et al.  
Health-related quality of life in patients with atrial fibrillation treated with catheter ablation or antiarrhythmic drug therapy: a systematic review and meta-analysis
.
CJC Open
 
2020
;
2
:
286
95
.

737

Vanderhout
 
S
,
Fergusson
 
DA
,
Cook
 
JA
,
Taljaard
 
M
.
Patient-reported outcomes and target effect sizes in pragmatic randomized trials in ClinicalTrials.gov: a cross-sectional analysis
.
PLoS Med
 
2022
;
19
:
e1003896
.

738

Steinberg
 
BA
,
Piccini
 
JP
, Sr
.
Tackling patient-reported outcomes in atrial fibrillation and heart failure: identifying disease-specific symptoms?
 
Cardiol Clin
 
2019
;
37
:
139
46
.

739

Härdén
 
M
,
Nyström
 
B
,
Bengtson
 
A
,
Medin
 
J
,
Frison
 
L
,
Edvardsson
 
N
.
Responsiveness of AF6, a new, short, validated, atrial fibrillation-specific questionnaire—symptomatic benefit of direct current cardioversion
.
J Interv Card Electrophysiol
 
2010
;
28
:
185
91
.

740

Tailachidis
 
P
,
Tsimtsiou
 
Z
,
Galanis
 
P
,
Theodorou
 
M
,
Kouvelas
 
D
,
Athanasakis
 
K
.
The atrial fibrillation effect on QualiTy-of-Life (AFEQT) questionnaire: cultural adaptation and validation of the Greek version
.
Hippokratia
 
2016
;
20
:
264
7
.

741

Moreira
 
RS
,
Bassolli
 
L
,
Coutinho
 
E
,
Ferrer
 
P
,
Bragança
 
ÉO
,
Carvalho
 
AC
, et al.  
Reproducibility and reliability of the quality of life questionnaire in patients with atrial fibrillation
.
Arq Bras Cardiol
 
2016
;
106
:
171
81
.

742

Arribas
 
F
,
Ormaetxe
 
JM
,
Peinado
 
R
,
Perulero
 
N
,
Ramírez
 
P
,
Badia
 
X
.
Validation of the AF-QoL, a disease-specific quality of life questionnaire for patients with atrial fibrillation
.
Europace
 
2010
;
12
:
364
70
.

743

Braganca
 
EO
,
Filho
 
BL
,
Maria
 
VH
,
Levy
 
D
,
de Paola
 
AA
.
Validating a new quality of life questionnaire for atrial fibrillation patients
.
Int J Cardiol
 
2010
;
143
:
391
8
.

744

International Consortium for Health Outcomes Measurement
. Atrial fibrillation data collection reference guide Version 5.0.1. https://connect.ichom.org/wp-content/uploads/2023/02/03-Atrial-Fibrillation-Reference-Guide-2023.5.0.1.pdf.

745

Dan
 
GA
,
Dan
 
AR
,
Ivanescu
 
A
,
Buzea
 
AC
.
Acute rate control in atrial fibrillation: an urgent need for the clinician
.
Eur Heart J Suppl
 
2022
;
24
:
D3
D10
.

746

Shima
 
N
,
Miyamoto
 
K
,
Kato
 
S
,
Yoshida
 
T
,
Uchino
 
S
;
AFTER-ICU study group
.
Primary success of electrical cardioversion for new-onset atrial fibrillation and its association with clinical course in non-cardiac critically ill patients: sub-analysis of a multicenter observational study
.
J Intensive Care
 
2021
;
9
:
46
.

747

Betthauser
 
KD
,
Gibson
 
GA
,
Piche
 
SL
,
Pope
 
HE
.
Evaluation of amiodarone use for new-onset atrial fibrillation in critically ill patients with septic shock
.
Hosp Pharm
 
2021
;
56
:
116
23
.

748

Drikite
 
L
,
Bedford
 
JP
,
O’Bryan
 
L
,
Petrinic
 
T
,
Rajappan
 
K
,
Doidge
 
J
, et al.  
Treatment strategies for new onset atrial fibrillation in patients treated on an intensive care unit: a systematic scoping review
.
Crit Care
 
2021
;
25
:
257
.

749

Bedford
 
JP
,
Johnson
 
A
,
Redfern
 
O
,
Gerry
 
S
,
Doidge
 
J
,
Harrison
 
D
, et al.  
Comparative effectiveness of common treatments for new-onset atrial fibrillation within the ICU: accounting for physiological status
.
J Crit Care
 
2022
;
67
:
149
56
.

750

Iwahashi
 
N
,
Takahashi
 
H
,
Abe
 
T
,
Okada
 
K
,
Akiyama
 
E
,
Matsuzawa
 
Y
, et al.  
Urgent control of rapid atrial fibrillation by landiolol in patients with acute decompensated heart failure with severely reduced ejection fraction
.
Circ Rep
 
2019
;
1
:
422
30
.

751

Unger
 
M
,
Morelli
 
A
,
Singer
 
M
,
Radermacher
 
P
,
Rehberg
 
S
,
Trimmel
 
H
, et al.  
Landiolol in patients with septic shock resident in an intensive care unit (LANDI-SEP): study protocol for a randomized controlled trial
.
Trials
 
2018
;
19
:
637
.

752

Gonzalez-Pacheco
 
H
,
Marquez
 
MF
,
Arias-Mendoza
 
A
,
Alvarez-Sangabriel
 
A
,
Eid-Lidt
 
G
,
Gonzalez-Hermosillo
 
A
, et al.  
Clinical features and in-hospital mortality associated with different types of atrial fibrillation in patients with acute coronary syndrome with and without ST elevation
.
J Cardiol
 
2015
;
66
:
148
54
.

753

Krijthe
 
BP
,
Leening
 
MJ
,
Heeringa
 
J
,
Kors
 
JA
,
Hofman
 
A
,
Franco
 
OH
, et al.  
Unrecognized myocardial infarction and risk of atrial fibrillation: the Rotterdam Study
.
Int J Cardiol
 
2013
;
168
:
1453
7
.

754

Soliman
 
EZ
,
Safford
 
MM
,
Muntner
 
P
,
Khodneva
 
Y
,
Dawood
 
FZ
,
Zakai
 
NA
, et al.  
Atrial fibrillation and the risk of myocardial infarction
.
JAMA Intern Med
 
2014
;
174
:
107
14
.

755

Kralev
 
S
,
Schneider
 
K
,
Lang
 
S
,
Suselbeck
 
T
,
Borggrefe
 
M
.
Incidence and severity of coronary artery disease in patients with atrial fibrillation undergoing first-time coronary angiography
.
PLoS One
 
2011
;
6
:
e24964
.

756

Coscia
 
T
,
Nestelberger
 
T
,
Boeddinghaus
 
J
,
Lopez-Ayala
 
P
,
Koechlin
 
L
,
Miró
 
Ò
, et al.  
Characteristics and outcomes of type 2 myocardial infarction
.
JAMA Cardiol
 
2022
;
7
:
427
34
.

757

Guimaraes
 
PO
,
Zakroysky
 
P
,
Goyal
 
A
,
Lopes
 
RD
,
Kaltenbach
 
LA
,
Wang
 
TY
.
Usefulness of antithrombotic therapy in patients with atrial fibrillation and acute myocardial infarction
.
Am J Cardiol
 
2019
;
123
:
12
8
.

758

Erez
 
A
,
Goldenberg
 
I
,
Sabbag
 
A
,
Nof
 
E
,
Zahger
 
D
,
Atar
 
S
, et al.  
Temporal trends and outcomes associated with atrial fibrillation observed during acute coronary syndrome: real-world data from the Acute Coronary Syndrome Israeli Survey (ACSIS), 2000–2013
.
Clin Cardiol
 
2017
;
40
:
275
80
.

759

Vrints C, Andreotti F, Koskinas K, Rossell X, Adamo M, Ainslie J, et al. 2024 ESC Guidelines for the management of chronic coronary syndromes. Eur Heart J  

2024
;
45
:
3415
537
.

760

Byrne
 
RA
,
Rossello
 
X
,
Coughlan
 
JJ
,
Barbato
 
E
,
Berry
 
C
,
Chieffo
 
A
, et al.  
2023 ESC Guidelines for the management of acute coronary syndromes
.
Eur Heart J
 
2023
;
44
:
3720
826
.

761

Park
 
DY
,
Wang
 
P
,
An
 
S
,
Grimshaw
 
AA
,
Frampton
 
J
,
Ohman
 
EM
, et al.  
Shortening the duration of dual antiplatelet therapy after percutaneous coronary intervention for acute coronary syndrome: a systematic review and meta-analysis
.
Am Heart J
 
2022
;
251
:
101
14
.

762

Gargiulo
 
G
,
Goette
 
A
,
Tijssen
 
J
,
Eckardt
 
L
,
Lewalter
 
T
,
Vranckx
 
P
, et al.  
Safety and efficacy outcomes of double vs. triple antithrombotic therapy in patients with atrial fibrillation following percutaneous coronary intervention: a systematic review and meta-analysis of non-vitamin K antagonist oral anticoagulant-based randomized clinical trials
.
Eur Heart J
 
2019
;
40
:
3757
67
.

763

Dewilde
 
WJ
,
Oirbans
 
T
,
Verheugt
 
FW
,
Kelder
 
JC
,
De Smet
 
BJ
,
Herrman
 
JP
, et al.  
Use of clopidogrel with or without aspirin in patients taking oral anticoagulant therapy and undergoing percutaneous coronary intervention: an open-label, randomised, controlled trial
.
Lancet
 
2013
;
381
:
1107
15
.

764

Lopes
 
RD
,
Heizer
 
G
,
Aronson
 
R
,
Vora
 
AN
,
Massaro
 
T
,
Mehran
 
R
, et al.  
Antithrombotic therapy after acute coronary syndrome or PCI in atrial fibrillation
.
N Engl J Med
 
2019
;
380
:
1509
24
.

765

Gibson
 
CM
,
Mehran
 
R
,
Bode
 
C
,
Halperin
 
J
,
Verheugt
 
FW
,
Wildgoose
 
P
, et al.  
Prevention of bleeding in patients with atrial fibrillation undergoing PCI
.
N Engl J Med
 
2016
;
375
:
2423
34
.

766

Cannon
 
CP
,
Bhatt
 
DL
,
Oldgren
 
J
,
Lip
 
GYH
,
Ellis
 
SG
,
Kimura
 
T
, et al.  
Dual antithrombotic therapy with dabigatran after PCI in atrial fibrillation
.
N Engl J Med
 
2017
;
377
:
1513
24
.

767

Vranckx
 
P
,
Valgimigli
 
M
,
Eckardt
 
L
,
Tijssen
 
J
,
Lewalter
 
T
,
Gargiulo
 
G
, et al.  
Edoxaban-based versus vitamin K antagonist-based antithrombotic regimen after successful coronary stenting in patients with atrial fibrillation (ENTRUST-AF PCI): a randomised, open-label, phase 3b trial
.
Lancet
 
2019
;
394
:
1335
43
.

768

Lopes
 
RD
,
Hong
 
H
,
Harskamp
 
RE
,
Bhatt
 
DL
,
Mehran
 
R
,
Cannon
 
CP
, et al.  
Safety and efficacy of antithrombotic strategies in patients with atrial fibrillation undergoing percutaneous coronary intervention: a network meta-analysis of randomized controlled trials
.
JAMA Cardiol
 
2019
;
4
:
747
55
.

769

Oldgren
 
J
,
Steg
 
PG
,
Hohnloser
 
SH
,
Lip
 
GYH
,
Kimura
 
T
,
Nordaby
 
M
, et al.  
Dabigatran dual therapy with ticagrelor or clopidogrel after percutaneous coronary intervention in atrial fibrillation patients with or without acute coronary syndrome: a subgroup analysis from the RE-DUAL PCI trial
.
Eur Heart J
 
2019
;
40
:
1553
62
.

770

Potpara
 
TS
,
Mujovic
 
N
,
Proietti
 
M
,
Dagres
 
N
,
Hindricks
 
G
,
Collet
 
JP
, et al.  
Revisiting the effects of omitting aspirin in combined antithrombotic therapies for atrial fibrillation and acute coronary syndromes or percutaneous coronary interventions: meta-analysis of pooled data from the PIONEER AF-PCI, RE-DUAL PCI, and AUGUSTUS trials
.
Europace
 
2020
;
22
:
33
46
.

771

Haller
 
PM
,
Sulzgruber
 
P
,
Kaufmann
 
C
,
Geelhoed
 
B
,
Tamargo
 
J
,
Wassmann
 
S
, et al.  
Bleeding and ischaemic outcomes in patients treated with dual or triple antithrombotic therapy: systematic review and meta-analysis
.
Eur Heart J Cardiovasc Pharmacother
 
2019
;
5
:
226
36
.

772

Lopes
 
RD
,
Hong
 
H
,
Harskamp
 
RE
,
Bhatt
 
DL
,
Mehran
 
R
,
Cannon
 
CP
, et al.  
Optimal antithrombotic regimens for patients with atrial fibrillation undergoing percutaneous coronary intervention: an updated network meta-analysis
.
JAMA Cardiol
 
2020
;
5
:
582
9
.

773

Lopes
 
RD
,
Vora
 
AN
,
Liaw
 
D
,
Granger
 
CB
,
Darius
 
H
,
Goodman
 
SG
, et al.  
An open-label, 2 × 2 factorial, randomized controlled trial to evaluate the safety of apixaban vs. vitamin K antagonist and aspirin vs. placebo in patients with atrial fibrillation and acute coronary syndrome and/or percutaneous coronary intervention: rationale and design of the AUGUSTUS trial
.
Am Heart J
 
2018
;
200
:
17
23
.

774

Windecker
 
S
,
Lopes
 
RD
,
Massaro
 
T
,
Jones-Burton
 
C
,
Granger
 
CB
,
Aronson
 
R
, et al.  
Antithrombotic therapy in patients with atrial fibrillation and acute coronary syndrome treated medically or with percutaneous coronary intervention or undergoing elective percutaneous coronary intervention: insights from the AUGUSTUS trial
.
Circulation
 
2019
;
140
:
1921
32
.

775

Harskamp
 
RE
,
Fanaroff
 
AC
,
Lopes
 
RD
,
Wojdyla
 
DM
,
Goodman
 
SG
,
Thomas
 
LE
, et al.  
Antithrombotic therapy in patients with atrial fibrillation after acute coronary syndromes or percutaneous intervention
.
J Am Coll Cardiol
 
2022
;
79
:
417
27
.

776

Alexander
 
JH
,
Wojdyla
 
D
,
Vora
 
AN
,
Thomas
 
L
,
Granger
 
CB
,
Goodman
 
SG
, et al.  
Risk/benefit tradeoff of antithrombotic therapy in patients with atrial fibrillation early and late after an acute coronary syndrome or percutaneous coronary intervention: insights from AUGUSTUS
.
Circulation
 
2020
;
141
:
1618
27
.

777

Moayyedi
 
P
,
Eikelboom
 
JW
,
Bosch
 
J
,
Connolly
 
SJ
,
Dyal
 
L
,
Shestakovska
 
O
, et al.  
Safety of proton pump inhibitors based on a large, multi-year, randomized trial of patients receiving rivaroxaban or aspirin
.
Gastroenterology
 
2019
;
157
:
682
691.e2
.

778

Jeridi
 
D
,
Pellat
 
A
,
Ginestet
 
C
,
Assaf
 
A
,
Hallit
 
R
,
Corre
 
F
, et al.  
The safety of long-term proton pump inhibitor use on cardiovascular health: a meta-analysis
.
J Clin Med
 
2022
;
11
:
4096
.

779

Scally
 
B
,
Emberson
 
JR
,
Spata
 
E
,
Reith
 
C
,
Davies
 
K
,
Halls
 
H
, et al.  
Effects of gastroprotectant drugs for the prevention and treatment of peptic ulcer disease and its complications: a meta-analysis of randomised trials
.
Lancet Gastroenterol Hepatol
 
2018
;
3
:
231
41
.

780

Fiedler
 
KA
,
Maeng
 
M
,
Mehilli
 
J
,
Schulz-Schupke
 
S
,
Byrne
 
RA
,
Sibbing
 
D
, et al.  
Duration of triple therapy in patients requiring oral anticoagulation after drug-eluting stent implantation: the ISAR-TRIPLE trial
.
J Am Coll Cardiol
 
2015
;
65
:
1619
29
.

781

Matsumura-Nakano
 
Y
,
Shizuta
 
S
,
Komasa
 
A
,
Morimoto
 
T
,
Masuda
 
H
,
Shiomi
 
H
, et al.  
Open-label randomized trial comparing oral anticoagulation with and without single antiplatelet therapy in patients with atrial fibrillation and stable coronary artery disease beyond 1 year after coronary stent implantation
.
Circulation
 
2019
;
139
:
604
16
.

782

Jensen
 
T
,
Thrane
 
PG
,
Olesen
 
KKW
,
Würtz
 
M
,
Mortensen
 
MB
,
Gyldenkerne
 
C
, et al.  
Antithrombotic treatment beyond 1 year after percutaneous coronary intervention in patients with atrial fibrillation
.
Eur Heart J Cardiovasc Pharmacother
 
2023
;
9
:
208
19
.

783

Vitalis
 
A
,
Shantsila
 
A
,
Proietti
 
M
,
Vohra
 
RK
,
Kay
 
M
,
Olshansky
 
B
, et al.  
Peripheral arterial disease in patients with atrial fibrillation: the AFFIRM study
.
Am J Med
 
2021
;
134
:
514
8
.

784

Wasmer
 
K
,
Unrath
 
M
,
Köbe
 
J
,
Malyar
 
NM
,
Freisinger
 
E
,
Meyborg
 
M
, et al.  
Atrial fibrillation is a risk marker for worse in-hospital and long-term outcome in patients with peripheral artery disease
.
Int J Cardiol
 
2015
;
199
:
223
8
.

785

Griffin
 
WF
,
Salahuddin
 
T
,
O’Neal
 
WT
,
Soliman
 
EZ
.
Peripheral arterial disease is associated with an increased risk of atrial fibrillation in the elderly
.
Europace
 
2016
;
18
:
794
8
.

786

Lin
 
LY
,
Lee
 
CH
,
Yu
 
CC
,
Tsai
 
CT
,
Lai
 
LP
,
Hwang
 
JJ
, et al.  
Risk factors and incidence of ischemic stroke in Taiwanese with nonvalvular atrial fibrillation—a nation wide database analysis
.
Atherosclerosis
 
2011
;
217
:
292
5
.

787

Su
 
MI
,
Cheng
 
YC
,
Huang
 
YC
,
Liu
 
CW
.
The impact of atrial fibrillation on one-year mortality in patients with severe lower extremity arterial disease
.
J Clin Med
 
2022
;
11
:
1936
.

788

Winkel
 
TA
,
Hoeks
 
SE
,
Schouten
 
O
,
Zeymer
 
U
,
Limbourg
 
T
,
Baumgartner
 
I
, et al.  
Prognosis of atrial fibrillation in patients with symptomatic peripheral arterial disease: data from the REduction of Atherothrombosis for Continued Health (REACH) Registry
.
Eur J Vasc Endovasc Surg
 
2010
;
40
:
9
16
.

789

Nejim
 
B
,
Mathlouthi
 
A
,
Weaver
 
L
,
Faateh
 
M
,
Arhuidese
 
I
,
Malas
 
MB
.
Safety of carotid artery revascularization procedures in patients with atrial fibrillation
.
J Vasc Surg
 
2020
;
72
:
2069
2078.e4
.

790

Jones
 
WS
,
Hellkamp
 
AS
,
Halperin
 
J
,
Piccini
 
JP
,
Breithardt
 
G
,
Singer
 
DE
, et al.  
Efficacy and safety of rivaroxaban compared with warfarin in patients with peripheral artery disease and non-valvular atrial fibrillation: insights from ROCKET AF
.
Eur Heart J
 
2014
;
35
:
242
9
.

791

Aboyans
 
V
,
Ricco
 
JB
,
Bartelink
 
MEL
,
Björck
 
M
,
Brodmann
 
M
,
Cohnert
 
T
, et al.  
2017 ESC Guidelines on the diagnosis and treatment of peripheral arterial diseases, in collaboration with the European Society for Vascular Surgery (ESVS): document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteries. Endorsed by: the European Stroke Organization (ESO) The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiology (ESC) and of the European Society for Vascular Surgery (ESVS)
.
Eur Heart J
 
2018
;
39
:
763
816
.

792

Schirmer
 
CM
,
Bulsara
 
KR
,
Al-Mufti
 
F
,
Haranhalli
 
N
,
Thibault
 
L
,
Hetts
 
SW
.
Antiplatelets and antithrombotics in neurointerventional procedures: guideline update
.
J Neurointerv Surg
 
2023
;
15
:
1155
62
.

793

Berge
 
E
,
Whiteley
 
W
,
Audebert
 
H
,
De Marchis
 
GM
,
Fonseca
 
AC
,
Padiglioni
 
C
, et al.  
European Stroke Organisation (ESO) guidelines on intravenous thrombolysis for acute ischaemic stroke
.
Eur Stroke J
 
2021
;
6
:
I
LXII
.

794

Caso
 
V
,
Masuhr
 
F
.
A narrative review of nonvitamin K antagonist oral anticoagulant use in secondary stroke prevention
.
J Stroke Cerebrovasc Dis
 
2019
;
28
:
2363
75
.

795

Fischer
 
U
,
Koga
 
M
,
Strbian
 
D
,
Branca
 
M
,
Abend
 
S
,
Trelle
 
S
, et al.  
Early versus later anticoagulation for stroke with atrial fibrillation
.
N Engl J Med
 
2023
;
388
:
2411
21
.

796

Oldgren
 
J
,
Åsberg
 
S
,
Hijazi
 
Z
,
Wester
 
P
,
Bertilsson
 
M
,
Norrving
 
B
.
Early versus delayed non-vitamin K antagonist oral anticoagulant therapy after acute ischemic stroke in atrial fibrillation (TIMING): a registry-based randomized controlled noninferiority study
.
Circulation
 
2022
;
146
:
1056
66
.

797

Schreuder
 
F
,
van Nieuwenhuizen
 
KM
,
Hofmeijer
 
J
,
Vermeer
 
SE
,
Kerkhoff
 
H
,
Zock
 
E
, et al.  
Apixaban versus no anticoagulation after anticoagulation-associated intracerebral haemorrhage in patients with atrial fibrillation in The Netherlands (APACHE-AF): a randomised, open-label, phase 2 trial
.
Lancet Neurol
 
2021
;
20
:
907
16
.

798

SoSTART Collaboration
.
Effects of oral anticoagulation for atrial fibrillation after spontaneous intracranial haemorrhage in the UK: a randomised, open-label, assessor-masked, pilot-phase, non-inferiority trial
.
Lancet Neurol
 
2021
;
20
:
842
53
.

799

Klein Klouwenberg
 
PM
,
Frencken
 
JF
,
Kuipers
 
S
,
Ong
 
DS
,
Peelen
 
LM
,
van Vught
 
LA
, et al.  
Incidence, predictors, and outcomes of new-onset atrial fibrillation in critically ill patients with sepsis. A cohort study
.
Am J Respir Crit Care Med
 
2017
;
195
:
205
11
.

800

Gundlund
 
A
,
Olesen
 
JB
,
Butt
 
JH
,
Christensen
 
MA
,
Gislason
 
GH
,
Torp-Pedersen
 
C
, et al.  
One-year outcomes in atrial fibrillation presenting during infections: a nationwide registry-based study
.
Eur Heart J
 
2020
;
41
:
1112
9
.

801

Induruwa
 
I
,
Hennebry
 
E
,
Hennebry
 
J
,
Thakur
 
M
,
Warburton
 
EA
,
Khadjooi
 
K
.
Sepsis-driven atrial fibrillation and ischaemic stroke. Is there enough evidence to recommend anticoagulation?
 
Eur J Intern Med
 
2022
;
98
:
32
6
.

802

Ko
 
D
,
Saleeba
 
C
,
Sadiq
 
H
,
Crawford
 
S
,
Paul
 
T
,
Shi
 
Q
, et al.  
Secondary precipitants of atrial fibrillation and anticoagulation therapy
.
J Am Heart Assoc
 
2021
;
10
:
e021746
.

803

Li
 
YG
,
Borgi
 
M
,
Lip
 
GY
.
Atrial fibrillation occurring initially during acute medical illness: the heterogeneous nature of disease, outcomes and management strategies
.
Eur Heart J Acute Cardiovasc Care
 
2018
;
10
:2048872618801763.

804

Lowres
 
N
,
Hillis
 
GS
,
Gladman
 
MA
,
Kol
 
M
,
Rogers
 
J
,
Chow
 
V
, et al.  
Self-monitoring for recurrence of secondary atrial fibrillation following non-cardiac surgery or acute illness: a pilot study
.
Int J Cardiol Heart Vasc
 
2020
;
29
:
100566
.

805

Søgaard
 
M
,
Skjøth
 
F
,
Nielsen
 
PB
,
Smit
 
J
,
Dalager-Pedersen
 
M
,
Larsen
 
TB
, et al.  
Thromboembolic risk in patients with pneumonia and new-onset atrial fibrillation not receiving anticoagulation therapy
.
JAMA Netw Open
 
2022
;
5
:
e2213945
.

806

Walkey
 
AJ
,
Hammill
 
BG
,
Curtis
 
LH
,
Benjamin
 
EJ
.
Long-term outcomes following development of new-onset atrial fibrillation during sepsis
.
Chest
 
2014
;
146
:
1187
95
.

807

McIntyre
 
WF
,
Um
 
KJ
,
Cheung
 
CC
,
Belley-Côté
 
EP
,
Dingwall
 
O
,
Devereaux
 
PJ
, et al.  
Atrial fibrillation detected initially during acute medical illness: a systematic review
.
Eur Heart J Acute Cardiovasc Care
 
2019
;
8
:
130
41
.

808

Marcus
 
GM
,
Vittinghoff
 
E
,
Whitman
 
IR
,
Joyce
 
S
,
Yang
 
V
,
Nah
 
G
, et al.  
Acute consumption of alcohol and discrete atrial fibrillation events
.
Ann Intern Med
 
2021
;
174
:
1503
9
.

809

Marcus
 
GM
,
Modrow
 
MF
,
Schmid
 
CH
,
Sigona
 
K
,
Nah
 
G
,
Yang
 
J
, et al.  
Individualized studies of triggers of paroxysmal atrial fibrillation: the I-STOP-AFib randomized clinical trial
.
JAMA Cardiol
 
2022
;
7
:
167
74
.

810

Lin
 
AL
,
Nah
 
G
,
Tang
 
JJ
,
Vittinghoff
 
E
,
Dewland
 
TA
,
Marcus
 
GM
.
Cannabis, cocaine, methamphetamine, and opiates increase the risk of incident atrial fibrillation
.
Eur Heart J
 
2022
;
43
:
4933
42
.

811

Butt
 
JH
,
Olesen
 
JB
,
Havers-Borgersen
 
E
,
Gundlund
 
A
,
Andersson
 
C
,
Gislason
 
GH
, et al.  
Risk of thromboembolism associated with atrial fibrillation following noncardiac surgery
.
J Am Coll Cardiol
 
2018
;
72
:
2027
36
.

812

Gundlund
 
A
,
Kümler
 
T
,
Bonde
 
AN
,
Butt
 
JH
,
Gislason
 
GH
,
Torp-Pedersen
 
C
, et al.  
Comparative thromboembolic risk in atrial fibrillation with and without a secondary precipitant–Danish nationwide cohort study
.
BMJ Open
 
2019
;
9
:
e028468
.

813

Walkey
 
AJ
,
Quinn
 
EK
,
Winter
 
MR
,
McManus
 
DD
,
Benjamin
 
EJ
.
Practice patterns and outcomes associated with use of anticoagulation among patients with atrial fibrillation during sepsis
.
JAMA Cardiol
 
2016
;
1
:
682
90
.

814

Darwish
 
OS
,
Strube
 
S
,
Nguyen
 
HM
,
Tanios
 
MA
.
Challenges of anticoagulation for atrial fibrillation in patients with severe sepsis
.
Ann Pharmacother
 
2013
;
47
:
1266
71
.

815

Quon
 
MJ
,
Behlouli
 
H
,
Pilote
 
L
.
Anticoagulant use and risk of ischemic stroke and bleeding in patients with secondary atrial fibrillation associated with acute coronary syndromes, acute pulmonary disease, or sepsis
.
JACC Clin Electrophysiol
 
2018
;
4
:
386
93
.

816

Echahidi
 
N
,
Pibarot
 
P
,
O’Hara
 
G
,
Mathieu
 
P
.
Mechanisms, prevention, and treatment of atrial fibrillation after cardiac surgery
.
J Am Coll Cardiol
 
2008
;
51
:
793
801
.

817

Gillinov
 
AM
,
Bagiella
 
E
,
Moskowitz
 
AJ
,
Raiten
 
JM
,
Groh
 
MA
,
Bowdish
 
ME
, et al.  
Rate control versus rhythm control for atrial fibrillation after cardiac surgery
.
N Engl J Med
 
2016
;
374
:
1911
21
.

818

Gaudino
 
M
,
Di Franco
 
A
,
Rong
 
LQ
,
Piccini
 
J
,
Mack
 
M
.
Postoperative atrial fibrillation: from mechanisms to treatment
.
Eur Heart J
 
2023
;
44
:
1020
39
.

819

Kotecha
 
D
,
Castella
 
M
.
Is it time to treat post-operative atrial fibrillation just like regular atrial fibrillation?
 
Eur Heart J
 
2020
;
41
:
652
654a
.

820

Konstantino
 
Y
,
Zelnik Yovel
 
D
,
Friger
 
MD
,
Sahar
 
G
,
Knyazer
 
B
,
Amit
 
G
.
Postoperative atrial fibrillation following coronary artery bypass graft surgery predicts long-term atrial fibrillation and stroke
.
Isr Med Assoc J
 
2016
;
18
:
744
8
.

821

Lee
 
SH
,
Kang
 
DR
,
Uhm
 
JS
,
Shim
 
J
,
Sung
 
JH
,
Kim
 
JY
, et al.  
New-onset atrial fibrillation predicts long-term newly developed atrial fibrillation after coronary artery bypass graft
.
Am Heart J
 
2014
;
167
:
593
600.e1
.

822

Lin
 
MH
,
Kamel
 
H
,
Singer
 
DE
,
Wu
 
YL
,
Lee
 
M
,
Ovbiagele
 
B
.
Perioperative/postoperative atrial fibrillation and risk of subsequent stroke and/or mortality
.
Stroke
 
2019
;
50
:
1364
71
.

823

AlTurki
 
A
,
Marafi
 
M
,
Proietti
 
R
,
Cardinale
 
D
,
Blackwell
 
R
,
Dorian
 
P
, et al.  
Major adverse cardiovascular events associated with postoperative atrial fibrillation after noncardiac surgery: a systematic review and meta-analysis
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e007437
.

824

Goyal
 
P
,
Kim
 
M
,
Krishnan
 
U
,
McCullough
 
SA
,
Cheung
 
JW
,
Kim
 
LK
, et al.  
Post-operative atrial fibrillation and risk of heart failure hospitalization
.
Eur Heart J
 
2022
;
43
:
2971
80
.

825

Eikelboom
 
R
,
Sanjanwala
 
R
,
Le
 
ML
,
Yamashita
 
MH
,
Arora
 
RC
.
Postoperative atrial fibrillation after cardiac surgery: a systematic review and meta-analysis
.
Ann Thorac Surg
 
2021
;
111
:
544
54
.

826

Benedetto
 
U
,
Gaudino
 
MF
,
Dimagli
 
A
,
Gerry
 
S
,
Gray
 
A
,
Lees
 
B
, et al.  
Postoperative atrial fibrillation and long-term risk of stroke after isolated coronary artery bypass graft surgery
.
Circulation
 
2020
;
142
:
1320
9
.

827

Taha
 
A
,
Nielsen
 
SJ
,
Bergfeldt
 
L
,
Ahlsson
 
A
,
Friberg
 
L
,
Björck
 
S
, et al.  
New-onset atrial fibrillation after coronary artery bypass grafting and long-term outcome: a population-based nationwide study from the SWEDEHEART registry
.
J Am Heart Assoc
 
2021
;
10
:
e017966
.

828

Dobrev
 
D
,
Aguilar
 
M
,
Heijman
 
J
,
Guichard
 
JB
,
Nattel
 
S
.
Postoperative atrial fibrillation: mechanisms, manifestations and management
.
Nat Rev Cardiol
 
2019
;
16
:
417
36
.

829

Mathew
 
JP
,
Fontes
 
ML
,
Tudor
 
IC
,
Ramsay
 
J
,
Duke
 
P
,
Mazer
 
CD
, et al.  
A multicenter risk index for atrial fibrillation after cardiac surgery
.
JAMA
 
2004
;
291
:
1720
9
.

830

Villareal
 
RP
,
Hariharan
 
R
,
Liu
 
BC
,
Kar
 
B
,
Lee
 
VV
,
Elayda
 
M
, et al.  
Postoperative atrial fibrillation and mortality after coronary artery bypass surgery
.
J Am Coll Cardiol
 
2004
;
43
:
742
8
.

831

Cardinale
 
D
,
Sandri
 
MT
,
Colombo
 
A
,
Salvatici
 
M
,
Tedeschi
 
I
,
Bacchiani
 
G
, et al.  
Prevention of atrial fibrillation in high-risk patients undergoing lung cancer surgery: the PRESAGE trial
.
Ann Surg
 
2016
;
264
:
244
51
.

832

Ojima
 
T
,
Nakamori
 
M
,
Nakamura
 
M
,
Katsuda
 
M
,
Hayata
 
K
,
Kato
 
T
, et al.  
Randomized clinical trial of landiolol hydrochloride for the prevention of atrial fibrillation and postoperative complications after oesophagectomy for cancer
.
Br J Surg
 
2017
;
104
:
1003
9
.

833

Arsenault
 
KA
,
Yusuf
 
AM
,
Crystal
 
E
,
Healey
 
JS
,
Morillo
 
CA
,
Nair
 
GM
, et al.  
Interventions for preventing post-operative atrial fibrillation in patients undergoing heart surgery
.
Cochrane Database Syst Rev
 
2013
;1:
CD003611
.

834

Ozaydin
 
M
,
Icli
 
A
,
Yucel
 
H
,
Akcay
 
S
,
Peker
 
O
,
Erdogan
 
D
, et al.  
Metoprolol vs. carvedilol or carvedilol plus N-acetyl cysteine on post-operative atrial fibrillation: a randomized, double-blind, placebo-controlled study
.
Eur Heart J
 
2013
;
34
:
597
604
.

835

O’Neal
 
JB
,
Billings
 
F
,
Liu
 
X
,
Shotwell
 
MS
,
Liang
 
Y
,
Shah
 
AS
, et al.  
Effect of preoperative beta-blocker use on outcomes following cardiac surgery
.
Am J Cardiol
 
2017
;
120
:
1293
7
.

836

Ziff
 
OJ
,
Samra
 
M
,
Howard
 
JP
,
Bromage
 
DI
,
Ruschitzka
 
F
,
Francis
 
DP
, et al.  
Beta-blocker efficacy across different cardiovascular indications: an umbrella review and meta-analytic assessment
.
BMC Med
 
2020
;
18
:
103
.

837

Tisdale
 
JE
,
Wroblewski
 
HA
,
Wall
 
DS
,
Rieger
 
KM
,
Hammoud
 
ZT
,
Young
 
JV
, et al.  
A randomized trial evaluating amiodarone for prevention of atrial fibrillation after pulmonary resection
.
Ann Thorac Surg
 
2009
;
88
:
886
93
; discussion 894–5.

838

Auer
 
J
,
Weber
 
T
,
Berent
 
R
,
Puschmann
 
R
,
Hartl
 
P
,
Ng
 
CK
, et al.  
A comparison between oral antiarrhythmic drugs in the prevention of atrial fibrillation after cardiac surgery: the pilot Study of Prevention of Postoperative Atrial Fibrillation (SPPAF), a randomized, placebo-controlled trial
.
Am Heart J
 
2004
;
147
:
636
43
.

839

Buckley
 
MS
,
Nolan
 
PE
, Jr
,
Slack
 
MK
,
Tisdale
 
JE
,
Hilleman
 
DE
,
Copeland
 
JG
.
Amiodarone prophylaxis for atrial fibrillation after cardiac surgery: meta-analysis of dose response and timing of initiation
.
Pharmacotherapy
 
2007
;
27
:
360
8
.

840

Riber
 
LP
,
Christensen
 
TD
,
Jensen
 
HK
,
Hoejsgaard
 
A
,
Pilegaard
 
HK
.
Amiodarone significantly decreases atrial fibrillation in patients undergoing surgery for lung cancer
.
Ann Thorac Surg
 
2012
;
94
:
339
44
; discussion 345–6.

841

Couffignal
 
C
,
Amour
 
J
,
Ait-Hamou
 
N
,
Cholley
 
B
,
Fellahi
 
JL
,
Duval
 
X
, et al.  
Timing of β-blocker reintroduction and the occurrence of postoperative atrial fibrillation after cardiac surgery: a prospective cohort study
.
Anesthesiology
 
2020
;
132
:
267
79
.

842

Piccini
 
JP
,
Ahlsson
 
A
,
Dorian
 
P
,
Gillinov
 
MA
,
Kowey
 
PR
,
Mack
 
MJ
, et al.  
Design and rationale of a phase 2 study of NeurOtoxin (Botulinum Toxin Type A) for the PreVention of post-operative atrial fibrillation—the NOVA study
.
Am Heart J
 
2022
;
245
:
51
9
.

843

O’Brien
 
B
,
Burrage
 
PS
,
Ngai
 
JY
,
Prutkin
 
JM
,
Huang
 
CC
,
Xu
 
X
, et al.  
Society of Cardiovascular Anesthesiologists/European Association of Cardiothoracic Anaesthetists Practice Advisory for the management of perioperative atrial fibrillation in patients undergoing cardiac surgery
.
J Cardiothorac Vasc Anesth
 
2019
;
33
:
12
26
.

844

Gaudino
 
M
,
Sanna
 
T
,
Ballman
 
KV
,
Robinson
 
NB
,
Hameed
 
I
,
Audisio
 
K
, et al.  
Posterior left pericardiotomy for the prevention of atrial fibrillation after cardiac surgery: an adaptive, single-centre, single-blind, randomised, controlled trial
.
Lancet
 
2021
;
398
:
2075
83
.

845

Abdelaziz
 
A
,
Hafez
 
AH
,
Elaraby
 
A
,
Roshdy
 
MR
,
Abdelaziz
 
M
,
Eltobgy
 
MA
, et al.  
Posterior pericardiotomy for the prevention of atrial fibrillation after cardiac surgery: a systematic review and meta-analysis of 25 randomised controlled trials
.
EuroIntervention
 
2023
;
19
:
e305
17
.

846

Soletti
 
GJ
,
Perezgrovas-Olaria
 
R
,
Harik
 
L
,
Rahouma
 
M
,
Dimagli
 
A
,
Alzghari
 
T
, et al.  
Effect of posterior pericardiotomy in cardiac surgery: a systematic review and meta-analysis of randomized controlled trials
.
Front Cardiovasc Med
 
2022
;
9
:
1090102
.

847

Conen
 
D
,
Ke Wang
 
M
,
Popova
 
E
,
Chan
 
MTV
,
Landoni
 
G
,
Cata
 
JP
, et al.  
Effect of colchicine on perioperative atrial fibrillation and myocardial injury after non-cardiac surgery in patients undergoing major thoracic surgery (COP-AF): an international randomised trial
.
Lancet
 
2023
;
402
:
1627
35
.

848

Fragão-Marques
 
M
,
Teixeira
 
F
,
Mancio
 
J
,
Seixas
 
N
,
Rocha-Neves
 
J
,
Falcão-Pires
 
I
, et al.  
Impact of oral anticoagulation therapy on postoperative atrial fibrillation outcomes: a systematic review and meta-analysis
.
Thromb J
 
2021
;
19
:
89
.

849

Neves
 
IA
,
Magalhães
 
A
,
Lima da Silva
 
G
,
Almeida
 
AG
,
Borges
 
M
,
Costa
 
J
, et al.  
Anticoagulation therapy in patients with post-operative atrial fibrillation: systematic review with meta-analysis
.
Vascul Pharmacol
 
2022
;
142
:
106929
.

850

Daoud
 
EG
,
Strickberger
 
SA
,
Man
 
KC
,
Goyal
 
R
,
Deeb
 
GM
,
Bolling
 
SF
, et al.  
Preoperative amiodarone as prophylaxis against atrial fibrillation after heart surgery
.
N Engl J Med
 
1997
;
337
:
1785
91
.

851

Yagdi
 
T
,
Nalbantgil
 
S
,
Ayik
 
F
,
Apaydin
 
A
,
Islamoglu
 
F
,
Posacioglu
 
H
, et al.  
Amiodarone reduces the incidence of atrial fibrillation after coronary artery bypass grafting
.
J Thorac Cardiovasc Surg
 
2003
;
125
:
1420
5
.

852

Butt
 
JH
,
Xian
 
Y
,
Peterson
 
ED
,
Olsen
 
PS
,
Rorth
 
R
,
Gundlund
 
A
, et al.  
Long-term thromboembolic risk in patients with postoperative atrial fibrillation after coronary artery bypass graft surgery and patients with nonvalvular atrial fibrillation
.
JAMA Cardiol
 
2018
;
3
:
417
24
.

853

Gialdini
 
G
,
Nearing
 
K
,
Bhave
 
PD
,
Bonuccelli
 
U
,
Iadecola
 
C
,
Healey
 
JS
, et al.  
Perioperative atrial fibrillation and the long-term risk of ischemic stroke
.
JAMA
 
2014
;
312
:
616
22
.

854

Horwich
 
P
,
Buth
 
KJ
,
Legare
 
JF
.
New onset postoperative atrial fibrillation is associated with a long-term risk for stroke and death following cardiac surgery
.
J Card Surg
 
2013
;
28
:
8
13
.

855

Devereaux
 
PJ
,
Yang
 
H
,
Yusuf
 
S
,
Guyatt
 
G
,
Leslie
 
K
, et al.  
Effects of extended-release metoprolol succinate in patients undergoing non-cardiac surgery (POISE trial): a randomised controlled trial
.
Lancet
 
2008
;
371
:
1839
47
.

856

Hart
 
RG
,
Diener
 
HC
,
Coutts
 
SB
,
Easton
 
JD
,
Granger
 
CB
,
O’Donnell
 
MJ
, et al.  
Embolic strokes of undetermined source: the case for a new clinical construct
.
Lancet Neurol
 
2014
;
13
:
429
38
.

857

Sanna
 
T
,
Diener
 
HC
,
Passman
 
RS
,
Di Lazzaro
 
V
,
Bernstein
 
RA
,
Morillo
 
CA
, et al.  
Cryptogenic stroke and underlying atrial fibrillation
.
N Engl J Med
 
2014
;
370
:
2478
86
.

858

Rubiera
 
M
,
Aires
 
A
,
Antonenko
 
K
,
Lémeret
 
S
,
Nolte
 
CH
,
Putaala
 
J
, et al.  
European Stroke Organisation (ESO) guideline on screening for subclinical atrial fibrillation after stroke or transient ischaemic attack of undetermined origin
.
Eur Stroke J
 
2022
;
7
:
VI
.

859

von Falkenhausen
 
AS
,
Feil
 
K
,
Sinner
 
MF
,
Schönecker
 
S
,
Müller
 
J
,
Wischmann
 
J
, et al.  
Atrial fibrillation risk assessment after embolic stroke of undetermined source
.
Ann Neurol
 
2023
;
93
:
479
88
.

860

Gladstone
 
DJ
,
Spring
 
M
,
Dorian
 
P
,
Panzov
 
V
,
Thorpe
 
KE
,
Hall
 
J
, et al.  
Atrial fibrillation in patients with cryptogenic stroke
.
N Engl J Med
 
2014
;
370
:
2467
77
.

861

Wachter
 
R
,
Gröschel
 
K
,
Gelbrich
 
G
,
Hamann
 
GF
,
Kermer
 
P
,
Liman
 
J
, et al.  
Holter-electrocardiogram-monitoring in patients with acute ischaemic stroke (Find-AF(RANDOMISED)): an open-label randomised controlled trial
.
Lancet Neurol
 
2017
;
16
:
282
90
.

862

Buck
 
BH
,
Hill
 
MD
,
Quinn
 
FR
,
Butcher
 
KS
,
Menon
 
BK
,
Gulamhusein
 
S
, et al.  
Effect of implantable vs prolonged external electrocardiographic monitoring on atrial fibrillation detection in patients with ischemic stroke: the PER DIEM randomized clinical trial
.
JAMA
 
2021
;
325
:
2160
8
.

863

Bernstein
 
RA
,
Kamel
 
H
,
Granger
 
CB
,
Piccini
 
JP
,
Sethi
 
PP
,
Katz
 
JM
, et al.  
Effect of long-term continuous cardiac monitoring vs usual care on detection of atrial fibrillation in patients with stroke attributed to large- or small-vessel disease: the STROKE-AF randomized clinical trial
.
JAMA
 
2021
;
325
:
2169
77
.

864

Tsivgoulis
 
G
,
Katsanos
 
AH
,
Köhrmann
 
M
,
Caso
 
V
,
Perren
 
F
,
Palaiodimou
 
L
, et al.  
Duration of implantable cardiac monitoring and detection of atrial fibrillation in ischemic stroke patients: a systematic review and meta-analysis
.
J Stroke
 
2019
;
21
:
302
11
.

865

Sagris
 
D
,
Harrison
 
SL
,
Buckley
 
BJR
,
Ntaios
 
G
,
Lip
 
GYH
.
Long-term cardiac monitoring after embolic stroke of undetermined source: search longer, look harder
.
Am J Med
 
2022
;
135
:
e311
7
.

866

Liantinioti
 
C
,
Palaiodimou
 
L
,
Tympas
 
K
,
Parissis
 
J
,
Theodorou
 
A
,
Ikonomidis
 
I
, et al.  
Potential utility of neurosonology in paroxysmal atrial fibrillation detection in patients with cryptogenic stroke
.
J Clin Med
 
2019
;
8
:
2002
.

867

Tsivgoulis
 
G
,
Katsanos
 
AH
,
Grory
 
BM
,
Köhrmann
 
M
,
Ricci
 
BA
,
Tsioufis
 
K
, et al.  
Prolonged cardiac rhythm monitoring and secondary stroke prevention in patients with cryptogenic cerebral ischemia
.
Stroke
 
2019
;
50
:
2175
80
.

868

Favilla
 
CG
,
Ingala
 
E
,
Jara
 
J
,
Fessler
 
E
,
Cucchiara
 
B
,
Messé
 
SR
, et al.  
Predictors of finding occult atrial fibrillation after cryptogenic stroke
.
Stroke
 
2015
;
46
:
1210
5
.

869

Lip
 
GY
,
Nielsen
 
PB
.
Should patients with atrial fibrillation and 1 stroke risk factor (CHA2DS2-VASc score 1 in men, 2 in women) be anticoagulated? Yes: even 1 stroke risk factor confers a real risk of stroke
.
Circulation
 
2016
;
133
:
1498
503
; discussion 1503.

870

Ricci
 
B
,
Chang
 
AD
,
Hemendinger
 
M
,
Dakay
 
K
,
Cutting
 
S
,
Burton
 
T
, et al.  
A simple score that predicts paroxysmal atrial fibrillation on outpatient cardiac monitoring after embolic stroke of unknown source
.
J Stroke Cerebrovasc Dis
 
2018
;
27
:
1692
6
.

871

Kwong
 
C
,
Ling
 
AY
,
Crawford
 
MH
,
Zhao
 
SX
,
Shah
 
NH
.
A clinical score for predicting atrial fibrillation in patients with cryptogenic stroke or transient ischemic attack
.
Cardiology
 
2017
;
138
:
133
40
.

872

Li
 
YG
,
Bisson
 
A
,
Bodin
 
A
,
Herbert
 
J
,
Grammatico-Guillon
 
L
,
Joung
 
B
, et al.  
C(2) HEST score and prediction of incident atrial fibrillation in poststroke patients: a French nationwide study
.
J Am Heart Assoc
 
2019
;
8
:
e012546
.

873

Haeusler
 
KG
,
Gröschel
 
K
,
Köhrmann
 
M
,
Anker
 
SD
,
Brachmann
 
J
,
Böhm
 
M
, et al.  
Expert opinion paper on atrial fibrillation detection after ischemic stroke
.
Clin Res Cardiol
 
2018
;
107
:
871
80
.

874

Dilaveris
 
PE
,
Antoniou
 
CK
,
Caiani
 
EG
,
Casado-Arroyo
 
R
,
Climent
 
A
,
Cluitmans
 
M
, et al.  
ESC working group on e-cardiology position paper: accuracy and reliability of electrocardiogram monitoring in the detection of atrial fibrillation in cryptogenic stroke patients: in collaboration with the Council on Stroke, the European Heart Rhythm Association, and the Digital Health Committee
.
Eur Heart J Digit Health
 
2022
;
3
:
341
58
.

875

Diener
 
HC
,
Sacco
 
RL
,
Easton
 
JD
,
Granger
 
CB
,
Bernstein
 
RA
,
Uchiyama
 
S
, et al.  
Dabigatran for prevention of stroke after embolic stroke of undetermined source
.
N Engl J Med
 
2019
;
380
:
1906
17
.

876

Hart
 
RG
,
Sharma
 
M
,
Mundl
 
H
,
Kasner
 
SE
,
Bangdiwala
 
SI
,
Berkowitz
 
SD
, et al.  
Rivaroxaban for stroke prevention after embolic stroke of undetermined source
.
N Engl J Med
 
2018
;
378
:
2191
201
.

877

Poli
 
S
,
Meissner
 
C
,
Baezner
 
HJ
,
Kraft
 
A
,
Hillenbrand
 
F
,
Hobohm
 
C
, et al.  
Apixaban for treatment of embolic stroke of undetermined source (ATTICUS) randomized trial—update of patient characteristics and study timeline after interim analysis
.
Eur Heart J
 
2021
;
42
:
ehab724.2070
.

878

Vaidya
 
VR
,
Arora
 
S
,
Patel
 
N
,
Badheka
 
AO
,
Patel
 
N
,
Agnihotri
 
K
, et al.  
Burden of arrhythmia in pregnancy
.
Circulation
 
2017
;
135
:
619
21
.

879

Lee
 
MS
,
Chen
 
W
,
Zhang
 
Z
,
Duan
 
L
,
Ng
 
A
,
Spencer
 
HT
, et al.  
Atrial fibrillation and atrial flutter in pregnant women—a population-based study
.
J Am Heart Assoc
 
2016
;
5
:
e003182
.

880

Tamirisa
 
KP
,
Elkayam
 
U
,
Briller
 
JE
,
Mason
 
PK
,
Pillarisetti
 
J
,
Merchant
 
FM
, et al.  
Arrhythmias in pregnancy
.
JACC Clin Electrophysiol
 
2022
;
8
:
120
35
.

881

Salam
 
AM
,
Ertekin
 
E
,
van Hagen
 
IM
,
Al Suwaidi
 
J
,
Ruys
 
TPE
,
Johnson
 
MR
, et al.  
Atrial fibrillation or flutter during pregnancy in patients with structural heart disease: data from the ROPAC (Registry on Pregnancy and Cardiac Disease)
.
JACC Clin Electrophysiol
 
2015
;
1
:
284
92
.

882

Chokesuwattanaskul
 
R
,
Thongprayoon
 
C
,
Bathini
 
T
,
O’Corragain
 
OA
,
Sharma
 
K
,
Prechawat
 
S
, et al.  
Incidence of atrial fibrillation in pregnancy and clinical significance: a meta-analysis
.
Adv Med Sci
 
2019
;
64
:
415
22
.

883

Tamirisa
 
KP
,
Dye
 
C
,
Bond
 
RM
,
Hollier
 
LM
,
Marinescu
 
K
,
Vaseghi
 
M
, et al.  
Arrhythmias and heart failure in pregnancy: a dialogue on multidisciplinary collaboration
.
J Cardiovasc Dev Dis
 
2022
;
9
:
199
.

884

Al Bahhawi
 
T
,
Aqeeli
 
A
,
Harrison
 
SL
,
Lane
 
DA
,
Skjøth
 
F
,
Buchan
 
I
, et al.  
Pregnancy-related complications and incidence of atrial fibrillation: a systematic review
.
J Clin Med
 
2023
;
12
:
1316
.

885

Regitz-Zagrosek
 
V
,
Roos-Hesselink
 
JW
,
Bauersachs
 
J
,
Blomstrom-Lundqvist
 
C
,
Cifkova
 
R
,
De Bonis
 
M
, et al.  
2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy
.
Eur Heart J
 
2018
;
39
:
3165
241
.

886

Areia
 
AL
,
Mota-Pinto
 
A
.
Experience with direct oral anticoagulants in pregnancy—a systematic review
.
J Perinat Med
 
2022
;
50
:
457
61
.

887

Ueberham
 
L
,
Hindricks
 
G
.
Anticoagulation in special patient populations with atrial fibrillation
.
Herz
 
2021
;
46
:
323
8
.

888

Bateman
 
BT
,
Heide-Jørgensen
 
U
,
Einarsdóttir
 
K
,
Engeland
 
A
,
Furu
 
K
,
Gissler
 
M
, et al.  
β-Blocker use in pregnancy and the risk for congenital malformations: an international cohort study
.
Ann Intern Med
 
2018
;
169
:
665
73
.

889

Butters
 
L
,
Kennedy
 
S
,
Rubin
 
PC
.
Atenolol in essential hypertension during pregnancy
.
BMJ
 
1990
;
301
:
587
9
.

890

Ramlakhan
 
KP
,
Kauling
 
RM
,
Schenkelaars
 
N
,
Segers
 
D
,
Yap
 
SC
,
Post
 
MC
, et al.  
Supraventricular arrhythmia in pregnancy
.
Heart
 
2022
;
108
:
1674
81
.

891

Katritsis
 
DG
,
Boriani
 
G
,
Cosio
 
FG
,
Hindricks
 
G
,
Jais
 
P
,
Josephson
 
ME
, et al.  
European Heart Rhythm Association (EHRA) consensus document on the management of supraventricular arrhythmias, endorsed by Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS), and Sociedad Latinoamericana de Estimulacion Cardiaca y Electrofisiologia (SOLAECE)
.
Eur Heart J
 
2017
;
19
:
465
511
.

892

Moore
 
JS
,
Teefey
 
P
,
Rao
 
K
,
Berlowitz
 
MS
,
Chae
 
SH
,
Yankowitz
 
J
.
Maternal arrhythmia: a case report and review of the literature
.
Obstet Gynecol Surv
 
2012
;
67
:
298
312
.

893

Wang
 
YC
,
Chen
 
CH
,
Su
 
HY
,
Yu
 
MH
.
The impact of maternal cardioversion on fetal haemodynamics
.
Eur J Obstet Gynecol Reprod Biol
 
2006
;
126
:
268
9
.

894

European Heart Rhythm Association
;
European Association for Cardio-Thoracic Surgery
;
Camm
 
AJ
,
Kirchhof
 
P
,
Lip
 
GY
,
Schotten
 
U
, et al.  
Guidelines for the management of atrial fibrillation: the Task Force for the Management of Atrial Fibrillation of the European Society of Cardiology (ESC)
.
Eur Heart J
 
2010
;
31
:
2369
429
.

895

Kockova
 
R
,
Kocka
 
V
,
Kiernan
 
T
,
Fahy
 
GJ
.
Ibutilide-induced cardioversion of atrial fibrillation during pregnancy
.
J Cardiovasc Electrophysiol
 
2007
;
18
:
545
7
.

896

Georgiopoulos
 
G
,
Tsiachris
 
D
,
Kordalis
 
A
,
Kontogiannis
 
C
,
Spartalis
 
M
,
Pietri
 
P
, et al.  
Pharmacotherapeutic strategies for atrial fibrillation in pregnancy
.
Expert Opin Pharmacother
 
2019
;
20
:
1625
36
.

897

Jensen
 
AS
,
Idorn
 
L
,
Nørager
 
B
,
Vejlstrup
 
N
,
Sondergaard
 
L
.
Anticoagulation in adults with congenital heart disease: the who, the when and the how?
 
Heart
 
2015
;
101
:
424
9
.

898

Pujol
 
C
,
Niesert
 
AC
,
Engelhardt
 
A
,
Schoen
 
P
,
Kusmenkov
 
E
,
Pittrow
 
D
, et al.  
Usefulness of direct oral anticoagulants in adult congenital heart disease
.
Am J Cardiol
 
2016
;
117
:
450
5
.

899

Yang
 
H
,
Bouma
 
BJ
,
Dimopoulos
 
K
,
Khairy
 
P
,
Ladouceur
 
M
,
Niwa
 
K
, et al.  
Non-vitamin K antagonist oral anticoagulants (NOACs) for thromboembolic prevention, are they safe in congenital heart disease? Results of a worldwide study
.
Int J Cardiol
 
2020
;
299
:
123
30
.

900

Renda
 
G
,
Ricci
 
F
,
Giugliano
 
RP
,
De Caterina
 
R
.
Non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation and valvular heart disease
.
J Am Coll Cardiol
 
2017
;
69
:
1363
71
.

901

Caldeira
 
D
,
David
 
C
,
Costa
 
J
,
Ferreira
 
JJ
,
Pinto
 
FJ
.
Non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation and valvular heart disease: systematic review and meta-analysis
.
Eur Heart J Cardiovasc Pharmacother
 
2018
;
4
:
111
8
.

902

Ammash
 
NM
,
Phillips
 
SD
,
Hodge
 
DO
,
Connolly
 
HM
,
Grogan
 
MA
,
Friedman
 
PA
, et al.  
Outcome of direct current cardioversion for atrial arrhythmias in adults with congenital heart disease
.
Int J Cardiol
 
2012
;
154
:
270
4
.

903

Feltes
 
TF
,
Friedman
 
RA
.
Transesophageal echocardiographic detection of atrial thrombi in patients with nonfibrillation atrial tachyarrhythmias and congenital heart disease
.
J Am Coll Cardiol
 
1994
;
24
:
1365
70
.

904

Roos-Hesselink
 
JW
,
Meijboom
 
FJ
,
Spitaels
 
SE
,
van Domburg
 
R
,
van Rijen
 
EH
,
Utens
 
EM
, et al.  
Excellent survival and low incidence of arrhythmias, stroke and heart failure long-term after surgical ASD closure at young age. A prospective follow-up study of 21–33 years
.
Eur Heart J
 
2003
;
24
:
190
7
.

905

Mas
 
JL
,
Derumeaux
 
G
,
Guillon
 
B
,
Massardier
 
E
,
Hosseini
 
H
,
Mechtouff
 
L
, et al.  
Patent foramen ovale closure or anticoagulation vs. antiplatelets after stroke
.
N Engl J Med
 
2017
;
377
:
1011
21
.

906

Gutierrez
 
SD
,
Earing
 
MG
,
Singh
 
AK
,
Tweddell
 
JS
,
Bartz
 
PJ
.
Atrial tachyarrhythmias and the Cox-Maze procedure in congenital heart disease
.
Congenit Heart Dis
 
2013
;
8
:
434
9
.

907

Kobayashi
 
J
,
Yamamoto
 
F
,
Nakano
 
K
,
Sasako
 
Y
,
Kitamura
 
S
,
Kosakai
 
Y
.
Maze procedure for atrial fibrillation associated with atrial septal defect
.
Circulation
 
1998
;
98
:
II399
402
.

908

Shim
 
H
,
Yang
 
JH
,
Park
 
PW
,
Jeong
 
DS
,
Jun
 
TG
.
Efficacy of the Maze procedure for atrial fibrillation associated with atrial septal defect
.
Korean J Thorac Cardiovasc Surg
 
2013
;
46
:
98
103
.

909

Sherwin
 
ED
,
Triedman
 
JK
,
Walsh
 
EP
.
Update on interventional electrophysiology in congenital heart disease: evolving solutions for complex hearts
.
Circ Arrhythm Electrophysiol
 
2013
;
6
:
1032
40
.

910

Chiha
 
M
,
Samarasinghe
 
S
,
Kabaker
 
AS
.
Thyroid storm: an updated review
.
J Intensive Care Med
 
2015
;
30
:
131
40
.

911

Y-Hassan
 
S
,
Falhammar
 
H
.
Cardiovascular manifestations and complications of pheochromocytomas and paragangliomas
.
J Clin Med
 
2020
;
9
:
2435
.

912

Baumgartner
 
C
,
da Costa
 
BR
,
Collet
 
TH
,
Feller
 
M
,
Floriani
 
C
,
Bauer
 
DC
, et al.  
Thyroid function within the normal range, subclinical hypothyroidism, and the risk of atrial fibrillation
.
Circulation
 
2017
;
136
:
2100
16
.

913

Huang
 
M
,
Yang
 
S
,
Ge
 
G
,
Zhi
 
H
,
Wang
 
L
.
Effects of thyroid dysfunction and the thyroid-stimulating hormone levels on the risk of atrial fibrillation: a systematic review and dose-response meta-analysis from cohort studies
.
Endocr Pract
 
2022
;
28
:
822
31
.

914

Shin
 
DG
,
Kang
 
MK
,
Han
 
D
,
Choi
 
S
,
Cho
 
JR
,
Lee
 
N
.
Enlarged left atrium and decreased left atrial strain are associated with atrial fibrillation in patients with hyperthyroidism irrespective of conventional risk factors
.
Int J Cardiovasc Imaging
 
2022
;
38
:
613
20
.

915

Sawin
 
CT
,
Geller
 
A
,
Wolf
 
PA
,
Belanger
 
AJ
,
Baker
 
E
,
Bacharach
 
P
, et al.  
Low serum thyrotropin concentrations as a risk factor for atrial fibrillation in older persons
.
N Engl J Med
 
1994
;
331
:
1249
52
.

916

Lyon
 
AR
,
López-Fernández
 
T
,
Couch
 
LS
,
Asteggiano
 
R
,
Aznar
 
MC
,
Bergler-Klein
 
J
, et al.  
2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS)
.
Eur Heart J
 
2022
;
43
:
4229
361
.

917

El Sabbagh
 
R
,
Azar
 
NS
,
Eid
 
AA
,
Azar
 
ST
.
Thyroid dysfunctions due to immune checkpoint inhibitors: a review
.
Int J Gen Med
 
2020
;
13
:
1003
9
.

918

Gammage
 
MD
,
Parle
 
JV
,
Holder
 
RL
,
Roberts
 
LM
,
Hobbs
 
FD
,
Wilson
 
S
, et al.  
Association between serum free thyroxine concentration and atrial fibrillation
.
Arch Intern Med
 
2007
;
167
:
928
34
.

919

Selmer
 
C
,
Olesen
 
JB
,
Hansen
 
ML
,
Lindhardsen
 
J
,
Olsen
 
AM
,
Madsen
 
JC
, et al.  
The spectrum of thyroid disease and risk of new onset atrial fibrillation: a large population cohort study
.
BMJ
 
2012
;
345
:
e7895
.

920

Kim
 
K
,
Yang
 
PS
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
,
Uhm
 
JS
, et al.  
Increased risk of ischemic stroke and systemic embolism in hyperthyroidism-related atrial fibrillation: a nationwide cohort study
.
Am Heart J
 
2021
;
242
:
123
31
.

921

Zhang
 
J
,
Bisson
 
A
,
Fauchier
 
G
,
Bodin
 
A
,
Herbert
 
J
,
Ducluzeau
 
PH
, et al.  
Yearly incidence of stroke and bleeding in atrial fibrillation with concomitant hyperthyroidism: a national discharge database study
.
J Clin Med
 
2022
;
11
:
1342
.

922

Bartalena
 
L
,
Bogazzi
 
F
,
Chiovato
 
L
,
Hubalewska-Dydejczyk
 
A
,
Links
 
TP
,
Vanderpump
 
M
.
2018 European Thyroid Association (ETA) guidelines for the management of amiodarone-associated thyroid dysfunction
.
Eur Thyroid J
 
2018
;
7
:
55
66
.

923

Cappellani
 
D
,
Papini
 
P
,
Di Certo
 
AM
,
Morganti
 
R
,
Urbani
 
C
,
Manetti
 
L
, et al.  
Duration of exposure to thyrotoxicosis increases mortality of compromised AIT patients: the role of early thyroidectomy
.
J Clin Endocrinol Metab
 
2020
;
105
:
dgaa464
.

924

Trevisan
 
C
,
Piovesan
 
F
,
Lucato
 
P
,
Zanforlini
 
BM
,
De Rui
 
M
,
Maggi
 
S
, et al.  
Parathormone, vitamin D and the risk of atrial fibrillation in older adults: a prospective study
.
Nutr Metab Cardiovasc Dis
 
2019
;
29
:
939
45
.

925

Pepe
 
J
,
Cipriani
 
C
,
Curione
 
M
,
Biamonte
 
F
,
Colangelo
 
L
,
Danese
 
V
, et al.  
Reduction of arrhythmias in primary hyperparathyroidism, by parathyroidectomy, evaluated with 24-h ECG monitoring
.
Eur J Endocrinol
 
2018
;
179
:
117
24
.

926

Pepe
 
J
,
Cipriani
 
C
,
Sonato
 
C
,
Raimo
 
O
,
Biamonte
 
F
,
Minisola
 
S
.
Cardiovascular manifestations of primary hyperparathyroidism: a narrative review
.
Eur J Endocrinol
 
2017
;
177
:
R297
308
.

927

Monticone
 
S
,
D’Ascenzo
 
F
,
Moretti
 
C
,
Williams
 
TA
,
Veglio
 
F
,
Gaita
 
F
, et al.  
Cardiovascular events and target organ damage in primary aldosteronism compared with essential hypertension: a systematic review and meta-analysis
.
Lancet Diabetes Endocrinol
 
2018
;
6
:
41
50
.

928

Bollati
 
M
,
Lopez
 
C
,
Bioletto
 
F
,
Ponzetto
 
F
,
Ghigo
 
E
,
Maccario
 
M
, et al.  
Atrial fibrillation and aortic ectasia as complications of primary aldosteronism: focus on pathophysiological aspects
.
Int J Mol Sci
 
2022
;
23
:
2111
.

929

Kim
 
KJ
,
Hong
 
N
,
Yu
 
MH
,
Lee
 
H
,
Lee
 
S
,
Lim
 
JS
, et al.  
Time-dependent risk of atrial fibrillation in patients with primary aldosteronism after medical or surgical treatment initiation
.
Hypertension
 
2021
;
77
:
1964
73
.

930

Larsson
 
SC
,
Lee
 
WH
,
Burgess
 
S
,
Allara
 
E
.
Plasma cortisol and risk of atrial fibrillation: a Mendelian randomization study
.
J Clin Endocrinol Metab
 
2021
;
106
:
e2521
6
.

931

Di Dalmazi
 
G
,
Vicennati
 
V
,
Pizzi
 
C
,
Mosconi
 
C
,
Tucci
 
L
,
Balacchi
 
C
, et al.  
Prevalence and incidence of atrial fibrillation in a large cohort of adrenal incidentalomas: a long-term study
.
J Clin Endocrinol Metab
 
2020
;
105
:dgaa270.

932

Hong
 
S
,
Kim
 
KS
,
Han
 
K
,
Park
 
CY
.
Acromegaly and cardiovascular outcomes: a cohort study
.
Eur Heart J
 
2022
;
43
:
1491
9
.

933

Polina
 
I
,
Jansen
 
HJ
,
Li
 
T
,
Moghtadaei
 
M
,
Bohne
 
LJ
,
Liu
 
Y
, et al.  
Loss of insulin signaling may contribute to atrial fibrillation and atrial electrical remodeling in type 1 diabetes
.
Proc Natl Acad Sci USA
 
2020
;
117
:
7990
8000
.

934

Lee
 
YB
,
Han
 
K
,
Kim
 
B
,
Lee
 
SE
,
Jun
 
JE
,
Ahn
 
J
, et al.  
Risk of early mortality and cardiovascular disease in type 1 diabetes: a comparison with type 2 diabetes, a nationwide study
.
Cardiovasc Diabetol
 
2019
;
18
:
157
.

935

Bisson
 
A
,
Bodin
 
A
,
Fauchier
 
G
,
Herbert
 
J
,
Angoulvant
 
D
,
Ducluzeau
 
PH
, et al.  
Sex, age, type of diabetes and incidence of atrial fibrillation in patients with diabetes mellitus: a nationwide analysis
.
Cardiovasc Diabetol
 
2021
;
20
:
24
.

936

Dahlqvist
 
S
,
Rosengren
 
A
,
Gudbjörnsdottir
 
S
,
Pivodic
 
A
,
Wedel
 
H
,
Kosiborod
 
M
, et al.  
Risk of atrial fibrillation in people with type 1 diabetes compared with matched controls from the general population: a prospective case-control study
.
Lancet Diabetes Endocrinol
 
2017
;
5
:
799
807
.

937

Cai
 
X
,
Li
 
J
,
Cai
 
W
,
Chen
 
C
,
Ma
 
J
,
Xie
 
Z
, et al.  
Meta-analysis of type 1 diabetes mellitus and risk of cardiovascular disease
.
J Diabetes Complications
 
2021
;
35
:
107833
.

938

Zellerhoff
 
S
,
Pistulli
 
R
,
Monnig
 
G
,
Hinterseer
 
M
,
Beckmann
 
BM
,
Kobe
 
J
, et al.  
Atrial arrhythmias in long-QT syndrome under daily life conditions: a nested case control study
.
J Cardiovasc Electrophysiol
 
2009
;
20
:
401
7
.

939

Johnson
 
JN
,
Tester
 
DJ
,
Perry
 
J
,
Salisbury
 
BA
,
Reed
 
CR
,
Ackerman
 
MJ
.
Prevalence of early-onset atrial fibrillation in congenital long QT syndrome
.
Heart Rhythm
 
2008
;
5
:
704
9
.

940

Gaita
 
F
,
Giustetto
 
C
,
Bianchi
 
F
,
Wolpert
 
C
,
Schimpf
 
R
,
Riccardi
 
R
, et al.  
Short QT syndrome: a familial cause of sudden death
.
Circulation
 
2003
;
108
:
965
70
.

941

Borggrefe
 
M
,
Wolpert
 
C
,
Antzelevitch
 
C
,
Veltmann
 
C
,
Giustetto
 
C
,
Gaita
 
F
, et al.  
Short QT syndrome genotype-phenotype correlations
.
J Electrocardiol
 
2005
;
38
:
75
80
.

942

Giustetto
 
C
,
Di Monte
 
F
,
Wolpert
 
C
,
Borggrefe
 
M
,
Schimpf
 
R
,
Sbragia
 
P
, et al.  
Short QT syndrome: clinical findings and diagnostic-therapeutic implications
.
Eur Heart J
 
2006
;
27
:
2440
7
.

943

Bordachar
 
P
,
Reuter
 
S
,
Garrigue
 
S
,
Cai
 
X
,
Hocini
 
M
,
Jais
 
P
, et al.  
Incidence, clinical implications and prognosis of atrial arrhythmias in Brugada syndrome
.
Eur Heart J
 
2004
;
25
:
879
84
.

944

Francis
 
J
,
Antzelevitch
 
C
.
Atrial fibrillation and Brugada syndrome
.
J Am Coll Cardiol
 
2008
;
51
:
1149
53
.

945

Giustetto
 
C
,
Schimpf
 
R
,
Mazzanti
 
A
,
Scrocco
 
C
,
Maury
 
P
,
Anttonen
 
O
, et al.  
Long-term follow-up of patients with short QT syndrome
.
J Am Coll Cardiol
 
2011
;
58
:
587
95
.

946

Gollob
 
MH
,
Redpath
 
CJ
,
Roberts
 
JD
.
The short QT syndrome: proposed diagnostic criteria
.
J Am Coll Cardiol
 
2011
;
57
:
802
12
.

947

Kusano
 
KF
,
Taniyama
 
M
,
Nakamura
 
K
,
Miura
 
D
,
Banba
 
K
,
Nagase
 
S
, et al.  
Atrial fibrillation in patients with Brugada syndrome relationships of gene mutation, electrophysiology, and clinical backgrounds
.
J Am Coll Cardiol
 
2008
;
51
:
1169
75
.

948

Rodriguez-Manero
 
M
,
Namdar
 
M
,
Sarkozy
 
A
,
Casado-Arroyo
 
R
,
Ricciardi
 
D
,
de Asmundis
 
C
, et al.  
Prevalence, clinical characteristics and management of atrial fibrillation in patients with Brugada syndrome
.
Am J Cardiol
 
2013
;
111
:
362
7
.

949

Choi
 
YJ
,
Choi
 
EK
,
Han
 
KD
,
Jung
 
JH
,
Park
 
J
,
Lee
 
E
, et al.  
Temporal trends of the prevalence and incidence of atrial fibrillation and stroke among Asian patients with hypertrophic cardiomyopathy: a nationwide population-based study
.
Int J Cardiol
 
2018
;
273
:
130
5
.

950

Hernandez-Ojeda
 
J
,
Arbelo
 
E
,
Borras
 
R
,
Berne
 
P
,
Tolosana
 
JM
,
Gomez-Juanatey
 
A
, et al.  
Patients with Brugada syndrome and implanted cardioverter-defibrillators: long-term follow-up
.
J Am Coll Cardiol
 
2017
;
70
:
1991
2002
.

951

Klopotowski
 
M
,
Kwapiszewska
 
A
,
Kukula
 
K
,
Jamiolkowski
 
J
,
Dabrowski
 
M
,
Derejko
 
P
, et al.  
Clinical and echocardiographic parameters as risk factors for atrial fibrillation in patients with hypertrophic cardiomyopathy
.
Clin Cardiol
 
2018
;
41
:
1336
40
.

952

Rowin
 
EJ
,
Orfanos
 
A
,
Estes
 
NAM
,
Wang
 
W
,
Link
 
MS
,
Maron
 
MS
, et al.  
Occurrence and natural history of clinically silent episodes of atrial fibrillation in hypertrophic cardiomyopathy
.
Am J Cardiol
 
2017
;
119
:
1862
5
.

953

Sacher
 
F
,
Probst
 
V
,
Maury
 
P
,
Babuty
 
D
,
Mansourati
 
J
,
Komatsu
 
Y
, et al.  
Outcome after implantation of a cardioverter-defibrillator in patients with Brugada syndrome: a multicenter study-part 2
.
Circulation
 
2013
;
128
:
1739
47
.

954

Siontis
 
KC
,
Geske
 
JB
,
Ong
 
K
,
Nishimura
 
RA
,
Ommen
 
SR
,
Gersh
 
BJ
.
Atrial fibrillation in hypertrophic cardiomyopathy: prevalence, clinical correlations, and mortality in a large high-risk population
.
J Am Heart Assoc
 
2014
;
3
:
e001002
.

955

Sumitomo
 
N
,
Sakurada
 
H
,
Taniguchi
 
K
,
Matsumura
 
M
,
Abe
 
O
,
Miyashita
 
M
, et al.  
Association of atrial arrhythmia and sinus node dysfunction in patients with catecholaminergic polymorphic ventricular tachycardia
.
Circ J
 
2007
;
71
:
1606
9
.

956

Sy
 
RW
,
Gollob
 
MH
,
Klein
 
GJ
,
Yee
 
R
,
Skanes
 
AC
,
Gula
 
LJ
, et al.  
Arrhythmia characterization and long-term outcomes in catecholaminergic polymorphic ventricular tachycardia
.
Heart Rhythm
 
2011
;
8
:
864
71
.

957

van Velzen
 
HG
,
Theuns
 
DA
,
Yap
 
SC
,
Michels
 
M
,
Schinkel
 
AF
.
Incidence of device-detected atrial fibrillation and long-term outcomes in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2017
;
119
:
100
5
.

958

Bourfiss
 
M
,
Te Riele
 
AS
,
Mast
 
TP
,
Cramer
 
MJ
,
Van Der Heijden
 
J
,
Van Veen
 
TAB
, et al.  
Influence of genotype on structural atrial abnormalities and atrial fibrillation or flutter in arrhythmogenic right ventricular dysplasia/cardiomyopathy
.
J Cardiovasc Electrophysiol
 
2016
;
27
:
1420
8
.

959

Camm
 
CF
,
James
 
CA
,
Tichnell
 
C
,
Murray
 
B
,
Bhonsale
 
A
,
te Riele
 
AS
, et al.  
Prevalence of atrial arrhythmias in arrhythmogenic right ventricular dysplasia/cardiomyopathy
.
Heart Rhythm
 
2013
;
10
:
1661
8
.

960

Chu
 
AF
,
Zado
 
E
,
Marchlinski
 
FE
.
Atrial arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia and ventricular tachycardia
.
Am J Cardiol
 
2010
;
106
:
720
2
.

961

Hasselberg
 
NE
,
Haland
 
TF
,
Saberniak
 
J
,
Brekke
 
PH
,
Berge
 
KE
,
Leren
 
TP
, et al.  
Lamin A/C cardiomyopathy: young onset, high penetrance, and frequent need for heart transplantation
.
Eur Heart J
 
2018
;
39
:
853
60
.

962

Kumar
 
S
,
Baldinger
 
SH
,
Gandjbakhch
 
E
,
Maury
 
P
,
Sellal
 
JM
,
Androulakis
 
AF
, et al.  
Long-term arrhythmic and nonarrhythmic outcomes of lamin A/C mutation carriers
.
J Am Coll Cardiol
 
2016
;
68
:
2299
307
.

963

Mussigbrodt
 
A
,
Knopp
 
H
,
Efimova
 
E
,
Weber
 
A
,
Bertagnolli
 
L
,
Hilbert
 
S
, et al.  
Supraventricular arrhythmias in patients with arrhythmogenic right ventricular dysplasia/cardiomyopathy associate with long-term outcome after catheter ablation of ventricular tachycardias
.
Europace
 
2018
;
20
:
1182
7
.

964

Pasotti
 
M
,
Klersy
 
C
,
Pilotto
 
A
,
Marziliano
 
N
,
Rapezzi
 
C
,
Serio
 
A
, et al.  
Long-term outcome and risk stratification in dilated cardiolaminopathies
.
J Am Coll Cardiol
 
2008
;
52
:
1250
60
.

965

Saguner
 
AM
,
Ganahl
 
S
,
Kraus
 
A
,
Baldinger
 
SH
,
Medeiros-Domingo
 
A
,
Saguner
 
AR
, et al.  
Clinical role of atrial arrhythmias in patients with arrhythmogenic right ventricular dysplasia
.
Circ J
 
2014
;
78
:
2854
61
.

966

Tonet
 
JL
,
Castro-Miranda
 
R
,
Iwa
 
T
,
Poulain
 
F
,
Frank
 
R
,
Fontaine
 
GH
.
Frequency of supraventricular tachyarrhythmias in arrhythmogenic right ventricular dysplasia
.
Am J Cardiol
 
1991
;
67
:
1153
.

967

van Rijsingen
 
IA
,
Nannenberg
 
EA
,
Arbustini
 
E
,
Elliott
 
PM
,
Mogensen
 
J
,
Hermans-van Ast
 
JF
, et al.  
Gender-specific differences in major cardiac events and mortality in lamin A/C mutation carriers
.
Eur J Heart Fail
 
2013
;
15
:
376
84
.

968

Aras
 
D
,
Tufekcioglu
 
O
,
Ergun
 
K
,
Ozeke
 
O
,
Yildiz
 
A
,
Topaloglu
 
S
, et al.  
Clinical features of isolated ventricular noncompaction in adults long-term clinical course, echocardiographic properties, and predictors of left ventricular failure
.
J Card Fail
 
2006
;
12
:
726
33
.

969

Li
 
S
,
Zhang
 
C
,
Liu
 
N
,
Bai
 
H
,
Hou
 
C
,
Wang
 
J
, et al.  
Genotype-positive status is associated with poor prognoses in patients with left ventricular noncompaction cardiomyopathy
.
J Am Heart Assoc
 
2018
;
7
:
e009910
.

970

Stollberger
 
C
,
Blazek
 
G
,
Winkler-Dworak
 
M
,
Finsterer
 
J
.
Atrial fibrillation in left ventricular noncompaction with and without neuromuscular disorders is associated with a poor prognosis
.
Int J Cardiol
 
2009
;
133
:
41
5
.

971

Fatkin
 
D
,
MacRae
 
C
,
Sasaki
 
T
,
Wolff
 
MR
,
Porcu
 
M
,
Frenneaux
 
M
, et al.  
Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease
.
N Engl J Med
 
1999
;
341
:
1715
24
.

972

Hong
 
K
,
Bjerregaard
 
P
,
Gussak
 
I
,
Brugada
 
R
.
Short QT syndrome and atrial fibrillation caused by mutation in KCNH2
.
J Cardiovasc Electrophysiol
 
2005
;
16
:
394
6
.

973

Olesen
 
MS
,
Yuan
 
L
,
Liang
 
B
,
Holst
 
AG
,
Nielsen
 
N
,
Nielsen
 
JB
, et al.  
High prevalence of long QT syndrome-associated SCN5A variants in patients with early-onset lone atrial fibrillation
.
Circ Cardiovasc Genet
 
2012
;
5
:
450
9
.

974

Pappone
 
C
,
Radinovic
 
A
,
Manguso
 
F
,
Vicedomini
 
G
,
Sala
 
S
,
Sacco
 
FM
, et al.  
New-onset atrial fibrillation as first clinical manifestation of latent Brugada syndrome: prevalence and clinical significance
.
Eur Heart J
 
2009
;
30
:
2985
92
.

975

Peters
 
S
.
Atrial arrhythmias in arrhythmogenic cardiomyopathy: at the beginning or at the end of the disease story?
 
Circ J
 
2015
;
79
:
446
.

976

Rowin
 
EJ
,
Hausvater
 
A
,
Link
 
MS
,
Abt
 
P
,
Gionfriddo
 
W
,
Wang
 
W
, et al.  
Clinical profile and consequences of atrial fibrillation in hypertrophic cardiomyopathy
.
Circulation
 
2017
;
136
:
2420
36
.

977

Mazzanti
 
A
,
Ng
 
K
,
Faragli
 
A
,
Maragna
 
R
,
Chiodaroli
 
E
,
Orphanou
 
N
, et al.  
Arrhythmogenic right ventricular cardiomyopathy: clinical course and predictors of arrhythmic risk
.
J Am Coll Cardiol
 
2016
;
68
:
2540
50
.

978

Giustetto
 
C
,
Cerrato
 
N
,
Gribaudo
 
E
,
Scrocco
 
C
,
Castagno
 
D
,
Richiardi
 
E
, et al.  
Atrial fibrillation in a large population with Brugada electrocardiographic pattern: prevalence, management, and correlation with prognosis
.
Heart Rhythm
 
2014
;
11
:
259
65
.

979

Beckmann
 
BM
,
Holinski-Feder
 
E
,
Walter
 
MC
,
Haserück
 
N
,
Reithmann
 
C
,
Hinterseer
 
M
, et al.  
Laminopathy presenting as familial atrial fibrillation
.
Int J Cardiol
 
2010
;
145
:
394
6
.

980

Pizzale
 
S
,
Gollob
 
MH
,
Gow
 
R
,
Birnie
 
DH
.
Sudden death in a young man with catecholaminergic polymorphic ventricular tachycardia and paroxysmal atrial fibrillation
.
J Cardiovasc Electrophysiol
 
2008
;
19
:
1319
21
.

981

Sugiyasu
 
A
,
Oginosawa
 
Y
,
Nogami
 
A
,
Hata
 
Y
.
A case with catecholaminergic polymorphic ventricular tachycardia unmasked after successful ablation of atrial tachycardias from pulmonary veins
.
Pacing Clin Electrophysiol
 
2009
;
32
:
e21
4
.

982

Veltmann
 
C
,
Kuschyk
 
J
,
Schimpf
 
R
,
Streitner
 
F
,
Schoene
 
N
,
Borggrefe
 
M
, et al.  
Prevention of inappropriate ICD shocks in patients with Brugada syndrome
.
Clin Res Cardiol
 
2010
;
99
:
37
44
.

983

Brugada
 
J
,
Katritsis
 
DG
,
Arbelo
 
E
,
Arribas
 
F
,
Bax
 
JJ
,
Blomstrom-Lundqvist
 
C
, et al.  
2019 ESC Guidelines for the management of patients with supraventricular tachycardia. The Task Force for the management of patients with supraventricular tachycardia of the European Society of Cardiology (ESC)
.
Eur Heart J
 
2020
;
41
:
655
720
.

984

Klein
 
GJ
,
Bashore
 
TM
,
Sellers
 
TD
,
Pritchett
 
EL
,
Smith
 
WM
,
Gallagher
 
JJ
.
Ventricular fibrillation in the Wolff–Parkinson–White syndrome
.
N Engl J Med
 
1979
;
301
:
1080
5
.

985

Morady
 
F
,
DiCarlo
 
LA
, Jr
,
Baerman
 
JM
,
De Buitleir
 
M
.
Effect of propranolol on ventricular rate during atrial fibrillation in the Wolff–Parkinson–White syndrome
.
Pacing Clin Electrophysiol
 
1987
;
10
:
492
6
.

986

Sellers
 
TD
, Jr,
Bashore
 
TM
,
Gallagher
 
JJ
.
Digitalis in the pre-excitation syndrome. Analysis during atrial fibrillation
.
Circulation
 
1977
;
56
:
260
7
.

987

Glatter
 
KA
,
Dorostkar
 
PC
,
Yang
 
Y
,
Lee
 
RJ
,
Van Hare
 
GF
,
Keung
 
E
, et al.  
Electrophysiological effects of ibutilide in patients with accessory pathways
.
Circulation
 
2001
;
104
:
1933
9
.

988

Ludmer
 
PL
,
McGowan
 
NE
,
Antman
 
EM
,
Friedman
 
PL
.
Efficacy of propafenone in Wolff–Parkinson–White syndrome: electrophysiologic findings and long-term follow-up
.
J Am Coll Cardiol
 
1987
;
9
:
1357
63
.

989

Simonian
 
SM
,
Lotfipour
 
S
,
Wall
 
C
,
Langdorf
 
MI
.
Challenging the superiority of amiodarone for rate control in Wolff–Parkinson–White and atrial fibrillation
.
Intern Emerg Med
 
2010
;
5
:
421
6
.

990

Arbelo
 
E
,
Protonotarios
 
A
,
Gimeno
 
JR
,
Arbustini
 
E
,
Barriales-Villa
 
R
,
Basso
 
C
, et al.  
2023 ESC Guidelines for the management of cardiomyopathies
.
Eur Heart J
 
2023
;
44
:
3503
626
.

991

Hu
 
YF
,
Liu
 
CJ
,
Chang
 
PM
,
Tsao
 
HM
,
Lin
 
YJ
,
Chang
 
SL
, et al.  
Incident thromboembolism and heart failure associated with new-onset atrial fibrillation in cancer patients
.
Int J Cardiol
 
2013
;
165
:
355
7
.

992

Mosarla
 
RC
,
Vaduganathan
 
M
,
Qamar
 
A
,
Moslehi
 
J
,
Piazza
 
G
,
Giugliano
 
RP
.
Anticoagulation strategies in patients with cancer: JACC review topic of the week
.
J Am Coll Cardiol
 
2019
;
73
:
1336
49
.

993

Malavasi
 
VL
,
Fantecchi
 
E
,
Gianolio
 
L
,
Pesce
 
F
,
Longo
 
G
,
Marietta
 
M
, et al.  
Atrial fibrillation in patients with active malignancy and use of anticoagulants: under-prescription but no adverse impact on all-cause mortality
.
Eur J Intern Med
 
2019
;
59
:
27
33
.

994

Farmakis
 
D
,
Parissis
 
J
,
Filippatos
 
G
.
Insights into onco-cardiology: atrial fibrillation in cancer
.
J Am Coll Cardiol
 
2014
;
63
:
945
53
.

995

Yun
 
JP
,
Choi
 
EK
,
Han
 
KD
,
Park
 
SH
,
Jung
 
JH
,
Ahn
 
HJ
, et al.  
Risk of atrial fibrillation according to cancer type: a nationwide population-based study
.
JACC CardioOncol
 
2021
;
3
:
221
32
.

996

Alexandre
 
J
,
Salem
 
JE
,
Moslehi
 
J
,
Sassier
 
M
,
Ropert
 
C
,
Cautela
 
J
, et al.  
Identification of anticancer drugs associated with atrial fibrillation: analysis of the WHO pharmacovigilance database
.
Eur Heart J Cardiovasc Pharmacother
 
2021
;
7
:
312
20
.

997

Guha
 
A
,
Fradley
 
MG
,
Dent
 
SF
,
Weintraub
 
NL
,
Lustberg
 
MB
,
Alonso
 
A
, et al.  
Incidence, risk factors, and mortality of atrial fibrillation in breast cancer: a SEER-medicare analysis
.
Eur Heart J
 
2022
;
43
:
300
12
.

998

Pastori
 
D
,
Marang
 
A
,
Bisson
 
A
,
Menichelli
 
D
,
Herbert
 
J
,
Lip
 
GYH
, et al.  
Thromboembolism, mortality, and bleeding in 2,435,541 atrial fibrillation patients with and without cancer: a nationwide cohort study
.
Cancer
 
2021
;
127
:
2122
9
.

999

Aspberg
 
S
,
Yu
 
L
,
Gigante
 
B
,
Smedby
 
KE
,
Singer
 
DE
.
Risk of ischemic stroke and major bleeding in patients with atrial fibrillation and cancer
.
J Stroke Cerebrovasc Dis
 
2020
;
29
:
104560
.

1000

D’Souza
 
M
,
Carlson
 
N
,
Fosbøl
 
E
,
Lamberts
 
M
,
Smedegaard
 
L
,
Nielsen
 
D
, et al.  
CHA(2)DS(2)-VASc score and risk of thromboembolism and bleeding in patients with atrial fibrillation and recent cancer
.
Eur J Prev Cardiol
 
2018
;
25
:
651
8
.

1001

Chen
 
ST
,
Hellkamp
 
AS
,
Becker
 
RC
,
Berkowitz
 
SD
,
Breithardt
 
G
,
Fox
 
KAA
, et al.  
Efficacy and safety of rivaroxaban vs. warfarin in patients with non-valvular atrial fibrillation and a history of cancer: observations from ROCKET AF
.
Eur Heart J Qual Care Clin Outcomes
 
2019
;
5
:
145
52
.

1002

Melloni
 
C
,
Dunning
 
A
,
Granger
 
CB
,
Thomas
 
L
,
Khouri
 
MG
,
Garcia
 
DA
, et al.  
Efficacy and safety of apixaban versus warfarin in patients with atrial fibrillation and a history of cancer: insights from the ARISTOTLE trial
.
Am J Med
 
2017
;
130
:
1440
8.e1
.

1003

Fanola
 
CL
,
Ruff
 
CT
,
Murphy
 
SA
,
Jin
 
J
,
Duggal
 
A
,
Babilonia
 
NA
, et al.  
Efficacy and safety of edoxaban in patients with active malignancy and atrial fibrillation: analysis of the ENGAGE AF-TIMI 48 trial
.
J Am Heart Assoc
 
2018
;
7
:
e008987
.

1004

Sawant
 
AC
,
Kumar
 
A
,
McCray
 
W
,
Tetewsky
 
S
,
Parone
 
L
,
Sridhara
 
S
, et al.  
Superior safety of direct oral anticoagulants compared to warfarin in patients with atrial fibrillation and underlying cancer: a national veterans affairs database study
.
J Geriatr Cardiol
 
2019
;
16
:
706
9
.

1005

Shah
 
S
,
Norby
 
FL
,
Datta
 
YH
,
Lutsey
 
PL
,
MacLehose
 
RF
,
Chen
 
LY
, et al.  
Comparative effectiveness of direct oral anticoagulants and warfarin in patients with cancer and atrial fibrillation
.
Blood Adv
 
2018
;
2
:
200
9
.

1006

Mariani
 
MV
,
Magnocavallo
 
M
,
Straito
 
M
,
Piro
 
A
,
Severino
 
P
,
Iannucci
 
G
, et al.  
Direct oral anticoagulants versus vitamin K antagonists in patients with atrial fibrillation and cancer a meta-analysis
.
J Thromb Thrombolysis
 
2021
;
51
:
419
29
.

1007

Deitelzweig
 
S
,
Keshishian
 
AV
,
Zhang
 
Y
,
Kang
 
A
,
Dhamane
 
AD
,
Luo
 
X
, et al.  
Effectiveness and safety of oral anticoagulants among nonvalvular atrial fibrillation patients with active cancer
.
JACC CardioOncol
 
2021
;
3
:
411
24
.

1008

Lin
 
YS
,
Kuan
 
FC
,
Chao
 
TF
,
Wu
 
M
,
Chen
 
SW
,
Chen
 
MC
, et al.  
Mortality associated with the use of non-vitamin K antagonist oral anticoagulants in cancer patients: dabigatran versus rivaroxaban
.
Cancer Med
 
2021
;
10
:
7079
88
.

1009

Atterman
 
A
,
Asplund
 
K
,
Friberg
 
L
,
Engdahl
 
J
.
Use of oral anticoagulants after ischaemic stroke in patients with atrial fibrillation and cancer
.
J Intern Med
 
2020
;
288
:
457
68
.

1010

Atterman
 
A
,
Friberg
 
L
,
Asplund
 
K
,
Engdahl
 
J
.
Net benefit of oral anticoagulants in patients with atrial fibrillation and active cancer: a nationwide cohort study
.
Europace
 
2020
;
22
:
58
65
.

1011

Falanga
 
A
,
Leader
 
A
,
Ambaglio
 
C
,
Bagoly
 
Z
,
Castaman
 
G
,
Elalamy
 
I
, et al.  
EHA guidelines on management of antithrombotic treatments in thrombocytopenic patients with cancer
.
Hemasphere
 
2022
;
6
:
e750
.

1012

Lancellotti
 
P
,
Suter
 
TM
,
López-Fernández
 
T
,
Galderisi
 
M
,
Lyon
 
AR
,
Van der Meer
 
P
, et al.  
Cardio-oncology services: rationale, organization, and implementation
.
Eur Heart J
 
2019
;
40
:
1756
63
.

1013

Richter
 
D
,
Guasti
 
L
,
Walker
 
D
,
Lambrinou
 
E
,
Lionis
 
C
,
Abreu
 
A
, et al.  
Frailty in cardiology: definition, assessment and clinical implications for general cardiology. A consensus document of the Council for Cardiology Practice (CCP), Association for Acute Cardio Vascular Care (ACVC), Association of Cardiovascular Nursing and Allied Professions (ACNAP), European Association of Preventive Cardiology (EAPC), European Heart Rhythm Association (EHRA), Council on Valvular Heart Diseases (VHD), Council on Hypertension (CHT), Council of Cardio-Oncology (CCO), Working Group (WG) aorta and peripheral vascular diseases, WG e-cardiology, WG thrombosis, of the European Society of Cardiology, European Primary Care Cardiology Society (EPCCS)
.
Eur J Prev Cardiol
 
2022
;
29
:
216
27
.

1014

Proietti
 
M
,
Vitolo
 
M
,
Harrison
 
SL
,
Lane
 
DA
,
Fauchier
 
L
,
Marin
 
F
, et al.  
Impact of clinical phenotypes on management and outcomes in European atrial fibrillation patients: a report from the ESC-EHRA EURObservational Research Programme in AF (EORP-AF) general long-term registry
.
BMC Med
 
2021
;
19
:
256
.

1015

Proietti
 
M
,
Romiti
 
GF
,
Vitolo
 
M
,
Harrison
 
SL
,
Lane
 
DA
,
Fauchier
 
L
, et al.  
Epidemiology and impact of frailty in patients with atrial fibrillation in Europe
.
Age Ageing
 
2022
;
51
:
afac192
.

1016

Savelieva
 
I
,
Fumagalli
 
S
,
Kenny
 
RA
,
Anker
 
S
,
Benetos
 
A
,
Boriani
 
G
, et al.  
EHRA expert consensus document on the management of arrhythmias in frailty syndrome, endorsed by the Heart Rhythm Society (HRS), Asia Pacific Heart Rhythm Society (APHRS), Latin America Heart Rhythm Society (LAHRS), and Cardiac Arrhythmia Society of Southern Africa (CASSA)
.
Europace
 
2023
;
25
:
1249
76
.

1017

Clegg
 
A
,
Young
 
J
,
Iliffe
 
S
,
Rikkert
 
MO
,
Rockwood
 
K
.
Frailty in elderly people
.
Lancet
 
2013
;
381
:
752
62
.

1018

Villani
 
ER
,
Tummolo
 
AM
,
Palmer
 
K
,
Gravina
 
EM
,
Vetrano
 
DL
,
Bernabei
 
R
, et al.  
Frailty and atrial fibrillation: a systematic review
.
Eur J Intern Med
 
2018
;
56
:
33
8
.

1019

Hang
 
F
,
Chen
 
J
,
Wang
 
Z
,
Yan
 
J
,
Wu
 
Y
.
Association between the frailty and new-onset atrial fibrillation/flutter among elderly hypertensive patients
.
Front Cardiovasc Med
 
2022
;
9
:
881946
.

1020

Steinberg
 
BA
,
Holmes
 
DN
,
Ezekowitz
 
MD
,
Fonarow
 
GC
,
Kowey
 
PR
,
Mahaffey
 
KW
, et al.  
Rate versus rhythm control for management of atrial fibrillation in clinical practice: results from the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation (ORBIT-AF) registry
.
Am Heart J
 
2013
;
165
:
622
9
.

1021

Ko
 
D
,
Lin
 
KJ
,
Bessette
 
LG
,
Lee
 
SB
,
Walkey
 
AJ
,
Cheng
 
S
, et al.  
Trends in use of oral anticoagulants in older adults with newly diagnosed atrial fibrillation, 2010–2020
.
JAMA Netw Open
 
2022
;
5
:
e2242964
.

1022

Bul
 
M
,
Shaikh
 
F
,
McDonagh
 
J
,
Ferguson
 
C
.
Frailty and oral anticoagulant prescription in adults with atrial fibrillation: a systematic review
.
Aging Med (Milton)
 
2023
;
6
:
195
206
.

1023

Hu
 
J
,
Zhou
 
Y
,
Cai
 
Z
.
Outcome of novel oral anticoagulant versus warfarin in frail elderly patients with atrial fibrillation: a systematic review and meta-analysis of retrospective studies
.
Acta Clin Belg
 
2023
;
78
:
367
77
.

1024

Zeng
 
S
,
Zheng
 
Y
,
Jiang
 
J
,
Ma
 
J
,
Zhu
 
W
,
Cai
 
X
.
Effectiveness and safety of DOACs vs. warfarin in patients with atrial fibrillation and frailty: a systematic review and meta-analysis
.
Front Cardiovasc Med
 
2022
;
9
:
907197
.

1025

Grymonprez
 
M
,
Petrovic
 
M
,
De Backer
 
TL
,
Steurbaut
 
S
,
Lahousse
 
L
.
Impact of frailty on the effectiveness and safety of non-vitamin K antagonist oral anticoagulants (NOACs) in patients with atrial fibrillation: a nationwide cohort study
.
Eur Heart J Qual Care Clin Outcomes
 
2024
;
10
:
55
65
.

1026

Kim
 
D
,
Yang
 
PS
,
Sung
 
JH
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
, et al.  
Effectiveness and safety of anticoagulation therapy in frail patients with atrial fibrillation
.
Stroke
 
2022
;
53
:
1873
82
.

1027

Chao
 
TF
,
Liu
 
CJ
,
Lin
 
YJ
,
Chang
 
SL
,
Lo
 
LW
,
Hu
 
YF
, et al.  
Oral anticoagulation in very elderly patients with atrial fibrillation: a nationwide cohort study
.
Circulation
 
2018
;
138
:
37
47
.

1028

Da Costa
 
A
,
Thévenin
 
J
,
Roche
 
F
,
Romeyer-Bouchard
 
C
,
Abdellaoui
 
L
,
Messier
 
M
, et al.  
Results from the Loire-Ardèche-Drôme-Isère-Puy-de-Dôme (LADIP) trial on atrial flutter, a multicentric prospective randomized study comparing amiodarone and radiofrequency ablation after the first episode of symptomatic atrial flutter
.
Circulation
 
2006
;
114
:
1676
81
.

1029

Natale
 
A
,
Newby
 
KH
,
Pisano
 
E
,
Leonelli
 
F
,
Fanelli
 
R
,
Potenza
 
D
, et al.  
Prospective randomized comparison of antiarrhythmic therapy versus first-line radiofrequency ablation in patients with atrial flutter
.
J Am Coll Cardiol
 
2000
;
35
:
1898
904
.

1030

Chinitz
 
JS
,
Gerstenfeld
 
EP
,
Marchlinski
 
FE
,
Callans
 
DJ
.
Atrial fibrillation is common after ablation of isolated atrial flutter during long-term follow-up
.
Heart Rhythm
 
2007
;
4
:
1029
33
.

1031

De Bortoli
 
A
,
Shi
 
LB
,
Ohm
 
OJ
,
Hoff
 
PI
,
Schuster
 
P
,
Solheim
 
E
, et al.  
Incidence and clinical predictors of subsequent atrial fibrillation requiring additional ablation after cavotricuspid isthmus ablation for typical atrial flutter
.
Scand Cardiovasc J
 
2017
;
51
:
123
8
.

1032

Rahman
 
F
,
Wang
 
N
,
Yin
 
X
,
Ellinor
 
PT
,
Lubitz
 
SA
,
LeLorier
 
PA
, et al.  
Atrial flutter: clinical risk factors and adverse outcomes in the Framingham Heart Study
.
Heart Rhythm
 
2016
;
13
:
233
40
.

1033

Roth
 
GA
,
Mensah
 
GA
,
Johnson
 
CO
,
Addolorato
 
G
,
Ammirati
 
E
,
Baddour
 
LM
, et al.  
Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study
.
J Am Coll Cardiol
 
2020
;
76
:
2982
3021
.

1034

Lippi
 
G
,
Sanchis-Gomar
 
F
,
Cervellin
 
G
.
Global epidemiology of atrial fibrillation: an increasing epidemic and public health challenge
.
Int J Stroke
 
2021
;
16
:
217
21
.

1035

Alonso
 
A
,
Alam
 
AB
,
Kamel
 
H
,
Subbian
 
V
,
Qian
 
J
,
Boerwinkle
 
E
, et al.  
Epidemiology of atrial fibrillation in the all of US research program
.
PLoS One
 
2022
;
17
:
e0265498
.

1036

Ghelani
 
KP
,
Chen
 
LY
,
Norby
 
FL
,
Soliman
 
EZ
,
Koton
 
S
,
Alonso
 
A
.
Thirty-year trends in the incidence of atrial fibrillation: the ARIC study
.
J Am Heart Assoc
 
2022
;
11
:
e023583
.

1037

Williams
 
BA
,
Chamberlain
 
AM
,
Blankenship
 
JC
,
Hylek
 
EM
,
Voyce
 
S
.
Trends in atrial fibrillation incidence rates within an integrated health care delivery system, 2006 to 2018
.
JAMA Netw Open
 
2020
;
3
:
e2014874
.

1038

Magnussen
 
C
,
Niiranen
 
TJ
,
Ojeda
 
FM
,
Gianfagna
 
F
,
Blankenberg
 
S
,
Njølstad
 
I
, et al.  
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
97
.

1039

Rodriguez
 
CJ
,
Soliman
 
EZ
,
Alonso
 
A
,
Swett
 
K
,
Okin
 
PM
,
Goff
 
DC
, Jr
, et al.  
Atrial fibrillation incidence and risk factors in relation to race-ethnicity and the population attributable fraction of atrial fibrillation risk factors: the multi-ethnic study of atherosclerosis
.
Ann Epidemiol
 
2015
;
25
:
71
6
, 76.e1.

1040

Ugowe
 
FE
,
Jackson
 
LR
, 2nd
,
Thomas
 
KL
.
Racial and ethnic differences in the prevalence, management, and outcomes in patients with atrial fibrillation: a systematic review
.
Heart Rhythm
 
2018
;
15
:
1337
45
.

1041

Volgman
 
AS
,
Bairey Merz
 
CN
,
Benjamin
 
EJ
,
Curtis
 
AB
,
Fang
 
MC
,
Lindley
 
KJ
, et al.  
Sex and race/ethnicity differences in atrial fibrillation
.
J Am Coll Cardiol
 
2019
;
74
:
2812
5
.

1042

Chung
 
SC
,
Sofat
 
R
,
Acosta-Mena
 
D
,
Taylor
 
JA
,
Lambiase
 
PD
,
Casas
 
JP
, et al.  
Atrial fibrillation epidemiology, disparity and healthcare contacts: a population-wide study of 5.6 million individuals
.
Lancet Reg Health Eur
 
2021
;
7
:
100157
.

1043

Svennberg
 
E
,
Tjong
 
F
,
Goette
 
A
,
Akoum
 
N
,
Di Biase
 
L
,
Bordachar
 
P
, et al.  
How to use digital devices to detect and manage arrhythmias: an EHRA practical guide
.
Europace
 
2022
;
24
:
979
1005
.

1044

Spatz
 
ES
,
Ginsburg
 
GS
,
Rumsfeld
 
JS
,
Turakhia
 
MP
.
Wearable digital health technologies for monitoring in cardiovascular medicine
.
N Engl J Med
 
2024
;
390
:
346
56
.

1045

Cooke
 
G
,
Doust
 
J
,
Sanders
 
S
.
Is pulse palpation helpful in detecting atrial fibrillation? A systematic review
.
J Fam Pract
 
2006
;
55
:
130
4
.

1046

Attia
 
ZI
,
Noseworthy
 
PA
,
Lopez-Jimenez
 
F
,
Asirvatham
 
SJ
,
Deshmukh
 
AJ
,
Gersh
 
BJ
, et al.  
An artificial intelligence-enabled ECG algorithm for the identification of patients with atrial fibrillation during sinus rhythm: a retrospective analysis of outcome prediction
.
Lancet
 
2019
;
394
:
861
7
.

1047

Hobbs
 
FD
,
Fitzmaurice
 
DA
,
Mant
 
J
,
Murray
 
E
,
Jowett
 
S
,
Bryan
 
S
, et al.  
A randomised controlled trial and cost-effectiveness study of systematic screening (targeted and total population screening) versus routine practice for the detection of atrial fibrillation in people aged 65 and over. The SAFE study
.
Health Technol Assess
 
2005
;
9
:
iii
iv
, ix-x, 1–74.

1048

Grond
 
M
,
Jauss
 
M
,
Hamann
 
G
,
Stark
 
E
,
Veltkamp
 
R
,
Nabavi
 
D
, et al.  
Improved detection of silent atrial fibrillation using 72-hour Holter ECG in patients with ischemic stroke: a prospective multicenter cohort study
.
Stroke
 
2013
;
44
:
3357
64
.

1049

Rizos
 
T
,
Guntner
 
J
,
Jenetzky
 
E
,
Marquardt
 
L
,
Reichardt
 
C
,
Becker
 
R
, et al.  
Continuous stroke unit electrocardiographic monitoring versus 24-hour Holter electrocardiography for detection of paroxysmal atrial fibrillation after stroke
.
Stroke
 
2012
;
43
:
2689
94
.

1050

Doliwa
 
PS
,
Frykman
 
V
,
Rosenqvist
 
M
.
Short-term ECG for out of hospital detection of silent atrial fibrillation episodes
.
Scand Cardiovasc J
 
2009
;
43
:
163
8
.

1051

Tieleman
 
RG
,
Plantinga
 
Y
,
Rinkes
 
D
,
Bartels
 
GL
,
Posma
 
JL
,
Cator
 
R
, et al.  
Validation and clinical use of a novel diagnostic device for screening of atrial fibrillation
.
Europace
 
2014
;
16
:
1291
5
.

1052

Kearley
 
K
,
Selwood
 
M
,
Van den Bruel
 
A
,
Thompson
 
M
,
Mant
 
D
,
Hobbs
 
FR
, et al.  
Triage tests for identifying atrial fibrillation in primary care: a diagnostic accuracy study comparing single-lead ECG and modified BP monitors
.
BMJ Open
 
2014
;
4
:
e004565
.

1053

Barrett
 
PM
,
Komatireddy
 
R
,
Haaser
 
S
,
Topol
 
S
,
Sheard
 
J
,
Encinas
 
J
, et al.  
Comparison of 24-hour Holter monitoring with 14-day novel adhesive patch electrocardiographic monitoring
.
Am J Med
 
2014
;
127
:
95.e11
7
.

1054

Turakhia
 
MP
,
Hoang
 
DD
,
Zimetbaum
 
P
,
Miller
 
JD
,
Froelicher
 
VF
,
Kumar
 
UN
, et al.  
Diagnostic utility of a novel leadless arrhythmia monitoring device
.
Am J Cardiol
 
2013
;
112
:
520
4
.

1055

Rosenberg
 
MA
,
Samuel
 
M
,
Thosani
 
A
,
Zimetbaum
 
PJ
.
Use of a noninvasive continuous monitoring device in the management of atrial fibrillation: a pilot study
.
Pacing Clin Electrophysiol
 
2013
;
36
:
328
33
.

1056

Turakhia
 
MP
,
Ullal
 
AJ
,
Hoang
 
DD
,
Than
 
CT
,
Miller
 
JD
,
Friday
 
KJ
, et al.  
Feasibility of extended ambulatory electrocardiogram monitoring to identify silent atrial fibrillation in high-risk patients: the Screening Study for Undiagnosed Atrial Fibrillation (STUDY-AF)
.
Clin Cardiol
 
2015
;
38
:
285
92
.

1057

Rooney
 
MR
,
Soliman
 
EZ
,
Lutsey
 
PL
,
Norby
 
FL
,
Loehr
 
LR
,
Mosley
 
TH
, et al.  
Prevalence and characteristics of subclinical atrial fibrillation in a community-dwelling elderly population: the ARIC study
.
Circ Arrhythm Electrophysiol
 
2019
;
12
:
e007390
.

1058

Stehlik
 
J
,
Schmalfuss
 
C
,
Bozkurt
 
B
,
Nativi-Nicolau
 
J
,
Wohlfahrt
 
P
,
Wegerich
 
S
, et al.  
Continuous wearable monitoring analytics predict heart failure hospitalization: the LINK-HF multicenter study
.
Circ Heart Fail
 
2020
;
13
:
e006513
.

1059

Ganne
 
C
,
Talkad
 
SN
,
Srinivas
 
D
,
Somanna
 
S
.
Ruptured blebs and racing hearts: autonomic cardiac changes in neurosurgeons during microsurgical clipping of aneurysms
.
Br J Neurosurg
 
2016
;
30
:
450
2
.

1060

Smith
 
WM
,
Riddell
 
F
,
Madon
 
M
,
Gleva
 
MJ
.
Comparison of diagnostic value using a small, single channel, P-wave centric sternal ECG monitoring patch with a standard 3-lead Holter system over 24 hours
.
Am Heart J
 
2017
;
185
:
67
73
.

1061

Olson
 
JA
,
Fouts
 
AM
,
Padanilam
 
BJ
,
Prystowsky
 
EN
.
Utility of mobile cardiac outpatient telemetry for the diagnosis of palpitations, presyncope, syncope, and the assessment of therapy efficacy
.
J Cardiovasc Electrophysio