Table of contents

  • 1. Preamble 3509

  • 2. Introduction 3511

  • 3. Phenotypic approach to cardiomyopathies 3511

  •  3.1. Definitions 3514

  •  3.2. Cardiomyopathy phenotypes 3514

  •   3.2.1. Hypertrophic cardiomyopathy 3514

  •   3.2.2. Dilated cardiomyopathy 3514

  •   3.2.3. Non-dilated left ventricular cardiomyopathy 3514

  •   3.2.4. Arrhythmogenic right ventricular cardiomyopathy 3516

  •   3.2.5. Restrictive cardiomyopathy 3517

  •  3.3. Other traits and syndromes associated with cardiomyopathy phenotypes 3517

  •   3.3.1. Left ventricular hypertrabeculation (left ventricular noncompaction) 3517

  •   3.3.2. Takotsubo syndrome 3517

  • 4. Epidemiology 3517

  •  4.1. Special populations 3518

  • 5. Integrated patient management 3518

  •  5.1. Multidisciplinary cardiomyopathy teams 3518

  •  5.2. Co-ordination between different levels of care 3518

  • 6. The patient pathway 3519

  •  6.1. Clinical presentation 3520

  •  6.2. Initial work-up 3520

  •  6.3. Systematic approach to diagnosis of cardiomyopathy 3520

  •  6.4. History and physical examination 3520

  •  6.5. Resting and ambulatory electrocardiography 3521

  •  6.6. Laboratory tests 3524

  •  6.7. Multimodality imaging 3524

  •   6.7.1. General considerations 3524

  •   6.7.2. Echocardiography 3524

  •   6.7.3. Cardiac magnetic resonance 3525

  •    6.7.3.1. Special considerations 3525

  •   6.7.4. Computed tomography and nuclear medicine techniques 3528

  •   6.7.5. Endomyocardial biopsy 3528

  •  6.8. Genetic testing and counselling 3529

  •   6.8.1. Genetic architecture 3529

  •   6.8.2. Genetic testing 3529

  •    6.8.2.1. Non-Mendelian cardiomyopathies and implications for genetic testing 3534

  •    6.8.2.2. Genetic test reports and variant interpretation 3534

  •   6.8.3. Genetic counselling 3534

  •    6.8.3.1. Genetic counselling in children 3534

  •    6.8.3.2. Pre- and post-test genetic counselling (proband) 3535

  •    6.8.3.3. Genetic counselling for cascade testing 3535

  •    6.8.3.4. Pre-natal or pre-implantation genetic diagnosis 3536

  •  6.9. Diagnostic approach to paediatric patients 3537

  •   6.9.1. Infantile and early childhood-onset cardiomyopathy 3538

  •  6.10. General principles in the management of patients with cardiomyopathy 3539

  •   6.10.1. Assessment of symptoms 3539

  •   6.10.2. Heart failure management 3539

  •    6.10.2.1. Preventive heart failure medical therapy of asymptomatic carriers/early disease expression 3540

  •    6.10.2.2. Cardiac transplantation 3540

  •    6.10.2.3. Left ventricular assist devices 3540

  •   6.10.3. Management of atrial arrhythmias 3541

  •    6.10.3.1. Anticoagulation 3541

  •    6.10.3.2. Rate control 3541

  •    6.10.3.3. Rhythm control 3543

  •    6.10.3.4. Comorbidities and risk factor management 3543

  •   6.10.4. Management of ventricular arrhythmias 3544

  •   6.10.5. Device therapy: implantable cardioverter defibrillator 3544

  •   6.10.6. Routine follow-up of patients with cardiomyopathy 3546

  •  6.11. Family screening and follow-up evaluation of relatives 3546

  •   6.11.1. Special considerations in family screening 3547

  •  6.12. Psychological support in cardiomyopathy patients and family members 3548

  •  6.13. The patient pathway 3549

  • 7. Specific cardiomyopathy phenotypes 3549

  •  7.1. Hypertrophic cardiomyopathy 3549

  •   7.1.1. Diagnosis 3549

  •    7.1.1.1. Diagnostic criteria 3549

  •    7.1.1.2. Diagnostic work-up 3549

  •    7.1.1.3. Echocardiography 3549

  •    7.1.1.4. Cardiac magnetic resonance 3550

  •    7.1.1.5. Nuclear imaging 3551

  •   7.1.2. Genetic testing and family screening 3551

  •   7.1.3. Assessment of symptoms 3552

  •   7.1.4. Management of symptoms and complications 3552

  •    7.1.4.1. Management of left ventricular outflow tract obstruction 3553

  •     7.1.4.1.1. General measures 3553

  •     7.1.4.1.2. Drug therapy 3553

  •     7.1.4.1.3. Invasive treatment of left ventricular outflow tract (septal reduction therapy) 3555

  •    7.1.4.2. Management of symptoms in patients without left ventricular outflow tract obstruction 3557

  •     7.1.4.2.1. Heart failure and chest pain 3557

  •     7.1.4.2.2. Cardiac resynchronization therapy 3557

  •   7.1.5. Sudden cardiac death prevention in hypertrophic cardiomyopathy 3558

  •    7.1.5.1. Left ventricular apical aneurysms 3559

  •    7.1.5.2. Left ventricular systolic dysfunction 3559

  •    7.1.5.3. Late gadolinium enhancement on cardiac magnetic resonance imaging 3559

  •    7.1.5.4. Abnormal exercise blood pressure response 3559

  •    7.1.5.5. Sarcomeric variants 3560

  •    7.1.5.6. Prevention of sudden cardiac death 3560

  •  7.2. Dilated cardiomyopathy 3562

  •   7.2.1. Diagnosis 3562

  •    7.2.1.1. Index case 3562

  •    7.2.1.2. Relatives 3562

  •    7.2.1.3. Diagnostic work-up 3562

  •    7.2.1.4. Echocardiography 3562

  •    7.2.1.5. Cardiac magnetic resonance 3563

  •    7.2.1.6. Nuclear medicine 3563

  •   7.2.2. Genetic testing and family screening 3563

  •    7.2.2.1. Genetic testing 3563

  •   7.2.3. Assessment of symptoms 3564

  •   7.2.4. Management 3564

  •   7.2.5. Sudden cardiac death prevention in dilated cardiomyopathy 3564

  •    7.2.5.1. Secondary prevention of sudden cardiac death 3564

  •    7.2.5.2. Primary prevention of sudden cardiac death 3564

  •  7.3. Non-dilated left ventricular cardiomyopathy 3566

  •   7.3.1. Diagnosis 3566

  •    7.3.1.1. Index case 3566

  •    7.3.1.2. Relatives 3566

  •    7.3.1.3. Diagnostic work-up 3566

  •    7.3.1.4. Electrocardiographic features 3567

  •    7.3.1.5. Echocardiography 3567

  •    7.3.1.6. Cardiac magnetic resonance 3567

  •    7.3.1.7. Nuclear medicine 3567

  •    7.3.1.8. Endomyocardial biopsy 3567

  •   7.3.2. Genetic testing 3567

  •   7.3.3. Assessment of symptoms 3567

  •   7.3.4. Management 3567

  •   7.3.5. Sudden cardiac death prevention in non-dilated left ventricular cardiomyopathy 3568

  •    7.3.5.1. Secondary prevention of sudden cardiac death 3568

  •    7.3.5.2. Primary prevention of sudden cardiac death 3568

  •  7.4. Arrhythmogenic right ventricular cardiomyopathy 3569

  •   7.4.1. Diagnosis 3569

  •    7.4.1.1. Index case 3569

  •    7.4.1.2. Relatives 3569

  •    7.4.1.3. Diagnostic work-up 3569

  •    7.4.1.4. Electrocardiography and Holter monitoring 3569

  •    7.4.1.5. Echocardiography and cardiac magnetic resonance 3569

  •    7.4.1.6. Endomyocardial biopsy 3569

  •    7.4.1.7. Nuclear medicine 3569

  •    7.4.1.8. Arrhythmogenic right ventricular cardiomyopathy phenocopies 3570

  •   7.4.2. Genetic testing and family screening 3570

  •   7.4.3. Assessment of symptoms 3570

  •   7.4.4. Management 3570

  •    7.4.4.1. Antiarrhythmic therapy 3570

  •   7.4.5. Sudden cardiac death prevention in arrhythmogenic right ventricular cardiomyopathy 3570

  •    7.4.5.1. Secondary prevention of sudden cardiac death 3571

  •    7.4.5.2. Primary prevention of sudden cardiac death 3571

  •  7.5. Restrictive cardiomyopathy 3572

  •   7.5.1. Diagnosis 3572

  •   7.5.2. Genetic testing 3572

  •   7.5.3. Assessment of symptoms 3573

  •   7.5.4. Management 3573

  •  7.6. Syndromic and metabolic cardiomyopathies 3574

  •   7.6.1. Anderson–Fabry disease 3574

  •    7.6.1.1. Definition 3574

  •    7.6.1.2. Diagnosis, clinical work-up, and differential diagnosis 3574

  •    7.6.1.3. Clinical course, outcome, and risk stratification 3574

  •    7.6.1.4. Management 3577

  •   7.6.2. RASopathies 3577

  •    7.6.2.1. Definition 3577

  •    7.6.2.2. Diagnosis, clinical work-up, and differential diagnosis 3577

  •    7.6.2.3. Clinical course, management, and sudden death risk stratification 3577

  •    7.6.2.4. Management 3577

  •   7.6.3. Friedreich ataxia 3578

  •    7.6.3.1. Definition 3578

  •    7.6.3.2. Diagnosis, clinical work-up, and differential diagnosis 3578

  •    7.6.3.3. Clinical course, management, and risk stratification 3579

  •    7.6.3.4. Management 3579

  •   7.6.4. Glycogen storage disorders 3579

  •    7.6.4.1. Definition 3579

  •    7.6.4.2. Diagnosis, clinical work-up, and differential diagnosis 3579

  •    7.6.4.3. Clinical course, management, and risk stratification 3579

  •    7.6.4.4. Management 3579

  •  7.7. Amyloidosis 3579

  •   7.7.1. Definition 3579

  •   7.7.2. Diagnosis, clinical work-up, and differential diagnosis 3579

  •   7.7.3. Clinical course and risk stratification 3580

  •   7.7.4. Management 3580

  •    7.7.4.1. Specific therapies 3581

  • 8. Other recommendations 3581

  •  8.1. Sports 3581

  •   8.1.1. Cardiovascular benefits of exercise 3581

  •   8.1.2. Exercise-related sudden cardiac death and historical exercise recommendations for patients with cardiomyopathy 3582

  •   8.1.3. Exercise recommendations in hypertrophic cardiomyopathy 3582

  •   8.1.4. Exercise recommendations in arrhythmogenic right ventricular cardiomyopathy 3582

  •   8.1.5. Exercise recommendations in dilated cardiomyopathy and non-dilated left ventricular cardiomyopathy 3582

  •  8.2. Reproductive issues 3583

  •   8.2.1. Contraception, in vitro fertilization, and hormonal treatment 3583

  •   8.2.2. Pregnancy management 3583

  •    8.2.2.1. Pre-pregnancy 3583

  •    8.2.2.2. Pregnancy 3583

  •    8.2.2.3. Timing and mode of delivery 3584

  •    8.2.2.4. Post-partum 3584

  •    8.2.2.5. Pharmacological treatment: general aspects 3584

  •    8.2.2.6. Specific cardiomyopathies 3584

  •    8.2.2.7. Peripartum cardiomyopathy 3585

  •  8.3. Recommendations for non-cardiac surgery 3585

  • 9. Requirements for specialized cardiomyopathy units 3586

  • 10. Living with cardiomyopathy: advice for patients 3586

  • 11. Sex differences in cardiomyopathies 3587

  • 12. Comorbidities and cardiovascular risk factors in cardiomyopathies 3587

  •  12.1. Cardiovascular risk factors 3587

  •  12.2. Dilated cardiomyopathy 3588

  •  12.3. Hypertrophic cardiomyopathy 3588

  •  12.4. Arrhythmogenic right ventricular cardiomyopathy 3588

  • 13. Coronavirus disease (COVID-19) and cardiomyopathies 3588

  • 14. Key messages 3588

  • 15. Gaps in evidence 3589

  • 16. ‘What to do’ and ‘What not to do’ messages from the Guidelines 3591

  • 17. Supplementary data 3595

  • 18. Data availability statement 3595

  • 19. Author information 3595

  • 20. Appendix 3596

  • 21. Acknowledgements 3597

  • 22. References 3597

Tables of Recommendations

  • Recommendation Table 1 — Recommendations for the provision of service of multidisciplinary cardiomyopathy teams 3519

  • Recommendation Table 2 — Recommendations for diagnostic work-up in cardiomyopathies 3520

  • Recommendation Table 3 — Recommendations for laboratory tests in the diagnosis of cardiomyopathies 3524

  • Recommendation Table 4 — Recommendation for echocardiographic evaluation in patients with cardiomyopathy 3524

  • Recommendation Table 5 — Recommendations for cardiac magnetic resonance indication in patients with cardiomyopathy 3526

  • Recommendation Table 6 — Recommendations for computed tomography and nuclear imaging 3528

  • Recommendation Table 7 — Recommendation for endomyocardial biopsy in patients with cardiomyopathy 3528

  • Recommendation Table 8 — Recommendations for genetic counselling and testing in cardiomyopathies 3537

  • Recommendation Table 9 — Recommendations for cardiac transplantation in patients with cardiomyopathy 3540

  • Recommendation Table 10 — Recommendation for left ventricular assist device therapy in patients with cardiomyopathy 3540

  • Recommendation Table 11 — Recommendations for management of atrial fibrillation and atrial flutter in patients with cardiomyopathy 3543

  • Recommendation Table 12 — Recommendations for implantable cardioverter defibrillator in patients with cardiomyopathy 3545

  • Recommendation Table 13 — Recommendations for routine follow-up of patients with cardiomyopathy 3546

  • Recommendation Table 14 — Recommendations for family screening and follow-up evaluation of relatives 3546

  • Recommendation Table 15 — Recommendations for psychological support in patients and family members with cardiomyopathies 3549

  • Recommendation Table 16 — Recommendation for evaluation of left ventricular outflow tract obstruction 3549

  • Recommendation Table 17 — Additional recommendation for cardiovascular magnetic resonance evaluation in hypertrophic cardiomyopathy 3550

  • Recommendation Table 18 — Recommendations for treatment of left ventricular outflow tract obstruction (general measures) 3553

  • Recommendation Table 19 — Recommendations for medical treatment of left ventricular outflow tract obstruction 3554

  • Recommendation Table 20 — Recommendations for septal reduction therapy 3556

  • Recommendation Table 21 — Recommendations for indications for cardiac pacing in patients with obstruction 3557

  • Recommendation Table 22 — Recommendations for chest pain on exertion in patients without left ventricular outflow tract obstruction 3557

  • Recommendation Table 23 — Additional recommendations for prevention of sudden cardiac death in patients with hypertrophic cardiomyopathy 3561

  • Recommendation Table 24 — Recommendations for an implantable cardioverter defibrillator in patients with dilated cardiomyopathy 3566

  • Recommendation Table 25 — Recommendation for resting and ambulatory electrocardiogram monitoring in patients with non-dilated left ventricular cardiomyopathy 3567

  • Recommendation Table 26 — Recommendations for an implantable cardioverter defibrillator in patients with non-dilated left ventricular cardiomyopathy 3568

  • Recommendation Table 27 — Recommendation for resting and ambulatory electrocardiogram monitoring in patients with arrhythmogenic right ventricular cardiomyopathy 3569

  • Recommendation Table 28 — Recommendations for the antiarrhythmic management of patients with arrhythmogenic right ventricular cardiomyopathy 3570

  • Recommendation Table 29 — Recommendations for sudden cardiac death prevention in patients with arrhythmogenic right ventricular cardiomyopathy 3571

  • Recommendation Table 30 — Recommendations for the management of patients with restrictive cardiomyopathy 3574

  • Recommendation Table 31 — Exercise recommendations for patients with cardiomyopathy 3582

  • Recommendation Table 32 — Recommendations for reproductive issues in patients with cardiomyopathy 3585

  • Recommendation Table 33 — Recommendations for non-cardiac surgery in patients with cardiomyopathy 3585

  • Recommendation Table 34 — Recommendation for management of cardiovascular risk factors in patients with cardiomyopathy 3588

List of tables

  • Table 1 Classes for recommendations 3510

  • Table 2 Levels of evidence 3510

  • Table 3 Morphological and functional traits used to describe cardiomyopathy phenotypes 3514

  • Table 4 Key epidemiological metrics in adults and children for the different cardiomyopathy phenotypes 3517

  • Table 5 Examples of inheritance patterns that should raise the suspicion of specific genetic aetiologies, grouped according to cardiomyopathy phenotype 3521

  • Table 6 Examples of signs and symptoms that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype 3522

  • Table 7 Examples of electrocardiographic features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype 3523

  • Table 8 First-level (to be performed in each patient) and second-level (to be performed in selected patients following specialist evaluation to identify specific aetiologies) laboratory tests, grouped by cardiomyopathy phenotype 3525

  • Table 9 Frequently encountered actionable results on multimodality imaging 3528

  • Table 10 Overview of genes associated with monogenic, non-syndromic cardiomyopathies, and their relative contributions to different cardiomyopathic phenotypes 3530

  • Table 11 Utility of genetic testing in cardiomyopathies 3533

  • Table 12 Specific issues to consider when counselling children 3534

  • Table 13 Key discussion points of pre- and post-test genetic counselling 3536

  • Table 14 Pre-natal and pre-implantation options and implications 3536

  • Table 15 Atrial fibrillation burden and management in cardiomyopathies 3542

  • Table 16 Psychological considerations 3548

  • Table 17 Imaging evaluation in hypertrophic cardiomyopathy 3550

  • Table 18 Echocardiographic features that suggest specific aetiologies in hypertrophic cardiomyopathy 3551

  • Table 19 Major clinical features associated with an increased risk of sudden cardiac death 3558

  • Table 20 Non-genetic causes of dilated cardiomyopathy 3563

  • Table 21 High-risk genotypes and associated predictors of sudden cardiac death 3566

  • Table 22 Clinical features and management of syndromic and metabolic cardiomyopathies 3575

  • Table 23 Anderson–Fabry disease red flags 3577

  • Table 24 General guidance for daily activity for patients with cardiomyopathies 3586

  • Table 25 Modulators of the phenotypic expression of cardiomyopathies 3588

List of figures

  • Figure 1 Central illustration 3512

  • Figure 2 Clinical diagnostic workflow of cardiomyopathy 3513

  • Figure 3 Examples of non-dilated left ventricular cardiomyopathy phenotypes and their aetiological correlates 3515

  • Figure 4 Worked example of the non-dilated left ventricular cardiomyopathy phenotype 3516

  • Figure 5 Multidisciplinary care of cardiomyopathies 3519

  • Figure 6 Multimodality imaging process in cardiomyopathies 3526

  • Figure 7 Examples of cardiac magnetic resonance imaging tissue characterization features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype 3527

  • Figure 8 The genetic architecture of the cardiomyopathies 3533

  • Figure 9 A patient-centred approach to cascade genetic testing of children 3535

  • Figure 10 Clinical approach to infantile and childhood cardiomyopathy 3538

  • Figure 11 Algorithm for the approach to family screening and follow-up of family members 3547

  • Figure 12 Protocol for the assessment and treatment of left ventricular outflow tract obstruction 3551

  • Figure 13 Algorithm for the treatment of heart failure in hypertrophic cardiomyopathy 3552

  • Figure 14 Flow chart on the management of left ventricular outflow tract obstruction 3554

  • Figure 15 Pre-assessment checklist for patients being considered for invasive septal reduction therapies 3555

  • Figure 16 Flow chart for implantation of an implantable cardioverter defibrillator in patients with hypertrophic cardiomyopathy 3561

  • Figure 17 Implantation of implantable cardioverter defibrillators in patients with dilated cardiomyopathy or non-dilated left ventricular cardiomyopathy flowchart 3565

  • Figure 18 Algorithm to approach implantable cardioverter defibrillator decision-making in patients with arrhythmogenic right ventricular cardiomyopathy 3572

  • Figure 19 Spectrum of restrictive heart diseases 3573

  • Figure 20 Anderson–Fabry disease diagnostic algorithm 3578

  • Figure 21 Screening for cardiac amyloidosis 3580

  • Figure 22 Diagnosis of cardiac amyloidosis 3581

Abbreviations and acronyms

     
  • 18F-FDG

    18F-fluorodeoxyglucose

  •  
  • 2D

    Two-dimensional

  •  
  • 3D

    Three-dimensional

  •  
  • 99mTc

    99mTechnetium

  •  
  • AAD

    Antiarrhythmic drug

  •  
  • ABC

    Atrial Fibrillation Better Care approach

  •  
  • ACE

    Angiotensin-converting enzyme

  •  
  • ACE-I

    Angiotensin-converting enzyme inhibitor

  •  
  • ACM

    Arrhythmogenic cardiomyopathy

  •  
  • AD

    Autosomal dominant

  •  
  • AED

    Automated external defibrillator

  •  
  • AF

    Atrial fibrillation

  •  
  • AFD

    Anderson–Fabry disease

  •  
  • AHA/ACC

    American Heart Association/American College of Cardiology

  •  
  • AL

    Monoclonal immunoglobulin light chain amyloidosis

  •  
  • ALCAPA

    Anomalous left coronary artery from the pulmonary artery

  •  
  • ALT

    Alanine aminotransferase

  •  
  • ALVC

    Arrhythmogenic left ventricular cardiomyopathy

  •  
  • APHRS

    Asia Pacific Heart Rhythm Society

  •  
  • AR

    Autosomal recessive

  •  
  • ARB

    Angiotensin receptor blocker

  •  
  • ARNI

    Angiotensin receptor neprilysin inhibitor

  •  
  • ARVC

    Arrhythmogenic right ventricular cardiomyopathy

  •  
  • ASA

    Alcohol septal ablation

  •  
  • AST

    Aspartate transaminase

  •  
  • ATPase

    Adenosine triphosphatase

  •  
  • ATTR

    Transthyretin amyloidosis

  •  
  • ATTR-CA

    Transthyretin cardiac amyloidosis

  •  
  • ATTR-CM

    Transthyretin amyloid cardiomyopathy

  •  
  • ATTRv

    Hereditary transthyretin amyloidosis

  •  
  • ATTRwt

    Wild-type OR Acquired transthyretin amyloidosis

  •  
  • AV

    Atrioventricular

  •  
  • b.p.m.

    Beats per minute

  •  
  • BAG3

    BAG cochaperone-3

  •  
  • BNP

    Brain natriuretic peptide

  •  
  • CAD

    Coronary artery disease

  •  
  • CCB

    Calcium channel blocker

  •  
  • CHA2DS2-VASc

    Congestive heart failure or left ventricular dysfunction, hypertension, age ≥75 (doubled), diabetes, stroke (doubled)-vascular disease, age 65–74, sex category (female) (score)

  •  
  • CHD

    Congenital heart disease

  •  
  • CK

    Creatinine kinase

  •  
  • CMR

    Cardiac magnetic resonance

  •  
  • COVID-19

    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection

  •  
  • CPET

    Cardio-pulmonary exercise testing

  •  
  • CPR

    Cardio-pulmonary resuscitation

  •  
  • CRT

    Cardiac resynchronization therapy

  •  
  • CrCl

    Creatinine clearance

  •  
  • CT

    Computed tomography

  •  
  • CTCA

    Computed tomography coronary angiography

  •  
  • DBS

    Deep brain stimulation

  •  
  • DCM

    Dilated cardiomyopathy

  •  
  • DES

    Desmin

  •  
  • DMD

    Duchenne muscular dystrophy

  •  
  • DOAC

    Direct-acting oral anticoagulant

  •  
  • DPD

    3,3-diphosphono-1,2-propanodicarboxylic acid

  •  
  • DSP

    Desmoplakin

  •  
  • EAST-AFNET

    Early Treatment of Atrial Fibrillation for Stroke Prevention Trial

  •  
  • ECG

    Electrocardiogram

  •  
  • ECHO

    Echocardiogram

  •  
  • ECV

    Extracellular volume

  •  
  • EF

    Ejection fraction

  •  
  • EHRA

    European Heart Rhythm Association

  •  
  • EMB

    Endomyocardial biopsy

  •  
  • EMF

    Endomyocardial fibrosis

  •  
  • EORP

    EURObservational Research Programme

  •  
  • ERN

    European Reference Network

  •  
  • ERT

    Enzyme replacement therapy

  •  
  • FLNC

    Filamin C

  •  
  • FRA

    Friedreich ataxia

  •  
  • FTX

    Frataxin

  •  
  • Gb3

    Globotriaosylceramide

  •  
  • GDMT

    Guideline-directed medical therapy

  •  
  • GSD

    Glycogen storage disorder

  •  
  • GWAS

    Genome-wide association study

  •  
  • HbA1c

    Haemoglobin A1C

  •  
  • HBP

    His-Bundle pacing

  •  
  • HCM

    Hypertrophic cardiomyopathy

  •  
  • HCMR

    Hypertrophic Cardiomyopathy Registry

  •  
  • 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

  •  
  • HMDP

    Hydroxymethylene diphosphonate

  •  
  • HR

    Hazard ratio

  •  
  • HRS

    Heart Rhythm Society

  •  
  • hs-cTnT

    High-sensitivity cardiac troponin T

  •  
  • ICD

    Implantable cardioverter defibrillator

  •  
  • INR

    International normalized ratio

  •  
  • ITFC

    International Task Force Consensus statement

  •  
  • IVF

    In vitro fertilization

  •  
  • LA

    Left atrium

  •  
  • LAHRS

    Latin American Heart Rhythm Society

  •  
  • LBBB

    Left bundle branch block

  •  
  • LGE

    Late gadolinium enhancement

  •  
  • LMNA

    Lamin A/C

  •  
  • LMWH

    Low-molecular-weight heparin

  •  
  • LSD

    Lysosomal storage disease

  •  
  • LV

    Left ventricular

  •  
  • LVAD

    LV assist device

  •  
  • LVEDV

    Left ventricular end-diastolic volume

  •  
  • LVEF

    Left ventricular ejection fraction

  •  
  • LVH

    Left ventricular hypertrophy

  •  
  • LVNC

    Left ventricular non-compaction

  •  
  • LVOT

    Left ventricular outflow tract

  •  
  • LVSD

    Left ventricular systolic dysfunction

  •  
  • LVOTO

    Left ventricular outflow tract obstruction

  •  
  • MCS

    Mechanical circulatory support

  •  
  • MELAS

    Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (syndrome)

  •  
  • MERRF

    Mitochondrial epilepsy with ragged-red fibres

  •  
  • MGUS

    Monoclonal gammopathy of undetermined significance

  •  
  • MICONOS

    Mitochondrial Protection with Idebenone in Cardiac or Neurological Outcome (study group)

  •  
  • MLVWT

    Maximum left ventricular wall thickness

  •  
  • MRA

    Mineralocorticoid receptor antagonist

  •  
  • MRI

    Magnetic resonance imaging

  •  
  • MV

    Mitral valve

  •  
  • mWHO

    Modified World Health Organization (classification)

  •  
  • NCS

    Non-cardiac surgery

  •  
  • NDLVC

    Non-dilated left ventricular cardiomyopathy

  •  
  • NGS

    Next-generation sequencing

  •  
  • NSML

    Noonan syndrome with multiple lentigines

  •  
  • NSVT

    Non-sustained ventricular tachycardia

  •  
  • NT-proBNP

    N-terminal pro-brain natriuretic peptide

  •  
  • NYHA

    New York Heart Association

  •  
  • OMT

    Optimal medical therapy

  •  
  • P/LP

    Pathogenic/likely pathogenic

  •  
  • PES

    Programmed electrical stimulation

  •  
  • PET

    Positron emission tomography

  •  
  • PKP2

    Plakophilin 2

  •  
  • PLN

    Phospholamban

  •  
  • PPCM

    Peripartum cardiomyopathy

  •  
  • PRKAG2

    Protein kinase AMP-activated non-catalytic subunit gamma 2

  •  
  • PRS

    Polygenic risk scores

  •  
  • PTH

    Parathyroid hormone

  •  
  • PVR

    Pulmonary vascular resistance

  •  
  • PYP

    Pyrophosphate

  •  
  • QoL

    Quality of life

  •  
  • QRS

    Q, R, and S waves of an ECG

  •  
  • RAS-HCM

    RASopathy-associated HCM

  •  
  • RBBB

    Right bundle branch block

  •  
  • RBM20

    RNA binding motif protein

  •  
  • RCM

    Restrictive cardiomyopathy

  •  
  • RCT

    Randomized controlled trial

  •  
  • RV

    Right ventricular

  •  
  • RVEF

    Right ventricular ejection fraction

  •  
  • RVOTO

    Right ventricular outflow tract obstruction

  •  
  • RWMA

    Regional wall motion abnormality

  •  
  • SAECG

    Signal-averaged electrocardiogram

  •  
  • SAM

    Systolic anterior motion

  •  
  • SCD

    Sudden cardiac death

  •  
  • SGLT2i

    Sodium–glucose co-transporter 2 inhibitor

  •  
  • SMVT

    Sustained monomorphic ventricular tachycardia

  •  
  • SPECT

    Single-photon emission computed tomography

  •  
  • SRT

    Septal reduction therapy

  •  
  • TIA

    Transient ischaemic attack

  •  
  • TMEM43

    transmembrane protein 43

  •  
  • TRED-HF

    Therapy withdrawal in REcovered Dilated cardiomyopathyHeart Failure

  •  
  • TTE

    Transthoracic echocardiography

  •  
  • TTN

    Titin

  •  
  • TTNtv

    Titin gene truncating variants

  •  
  • TTR

    Transthyretin

  •  
  • TWI

    T wave inversion

  •  
  • UFH

    Unfractionated heparin

  •  
  • VALOR-HCM

    A Study to Evaluate Mavacamten in Adults With Symptomatic Obstructive HCM Who Are Eligible for Septal Reduction Therapy

  •  
  • VE

    Ventricular extrasystole

  •  
  • VF

    Ventricular fibrillation

  •  
  • VKA

    Vitamin K antagonist

  •  
  • VT

    Ventricular tachycardia

  •  
  • VUS

    Variant of unknown significance

  •  
  • WHO

    World Health Organization

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, where appropriate, 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. ESC Policies and Procedures for formulating and issuing ESC Guidelines can be found on the ESC website (https://www.escardio.org/Guidelines).

The Members of this Task Force were selected by the ESC to represent professionals involved with the medical care of patients with this pathology. The selection procedure 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 evaluation of 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 below. 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.

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 and 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. The Task Force received its entire financial support from the ESC without any involvement from the healthcare industry.

Table 1

Classes for recommendations

graphic
graphic
Table 1

Classes for recommendations

graphic
graphic
Table 2

Levels of evidence

graphic
graphic
Table 2

Levels of evidence

graphic
graphic

The ESC Clinical Practice Guidelines (CPG) Committee supervises and co-ordinates the preparation of new guidelines and is responsible for the approval process. ESC Guidelines undergo extensive review by the CPG Committee and external experts, including members from across the whole of the ESC region and from relevant ESC Subspecialty Communities and National Cardiac Societies. After appropriate revisions, the guidelines are signed off by all the experts involved in the Task Force. The finalized document is signed off by the CPG Committee for publication in the European Heart Journal. The guidelines were developed after careful consideration of the scientific and medical knowledge and the evidence available at the time of their writing. Tables of evidence summarizing the findings of studies informing development of the guidelines are included. The ESC warns readers that the technical language may be misinterpreted and declines any responsibility in this respect.

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

The objective of this European Society of Cardiology (ESC) Guideline is to help healthcare professionals diagnose and manage patients with cardiomyopathies according to the best available evidence. Uniquely for relatively common cardiovascular diseases, there are very few randomized controlled clinical trials in patients with cardiomyopathies. For this reason, the majority of the recommendations in this guideline are based on observational cohort studies and expert consensus opinion. The aim is to provide healthcare professionals with a practical diagnostic and treatment framework for patients of all ages and, as an increasing number of patients have a known genetic basis for their disease, the guideline also considers the implications of a diagnosis for families and provides advice on reproduction and contraception. As cardiomyopathies can present at any age and can affect individuals and families across the entire life course, this guideline follows the principle of considering cardiomyopathies in all age groups as single disease entities, with recommendations applicable to children and adults with cardiomyopathy throughout, while accepting that the evidence base for many of the recommendations is significantly more limited for children. Age-related differences are specifically highlighted.

This is a new guideline, not an update of existing guidelines, with the exception of the section on hypertrophic cardiomyopathy (HCM), in which we have provided a focused update to the 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy.1 As such, most of the recommendations in this guideline are new. It is beyond the scope of this guideline to provide detailed descriptions and recommendations for each individual cardiomyopathy phenotype; instead, the aim is to provide a guide to the diagnostic approach to cardiomyopathies, highlight general evaluation and management issues, and signpost the reader to the relevant evidence base for the recommendations.

Adoption of morphological and functional disease definitions means that the number of possible aetiologies is considerable, particularly in young children. As it is impractical to provide an exhaustive compendium of all possible causes of cardiomyopathy, the guideline focuses on the most common disease phenotypes, but additional references for less common disorders are also provided. Similarly, treatment recommendations focus largely on generic management issues but refer to specific rare diseases when appropriate. The central illustration (Figure 1) highlights key aspects in the evaluation and management of cardiomyopathies addressed in this guideline.

Central illustration.
Figure 1

Central illustration.

Key aspects in the evaluation and management of cardiomyopathies. ARVC, arrhythmogenic right ventricular cardiomyopathy; CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; GDMT, guideline-directed medical therapy; HCM, hypertrophic cardiomyopathy; HF, heart failure ICD, implantable cardioverter defibrillator; LVOTO, left ventricular outflow tract obstruction; MCS, mechanical circulatory support; NDLVC, non-dilated left ventricular cardiomyopathy; PVR, pulmonary vascular resistance; RCM, restrictive cardiomyopathy; SCD, sudden cardiac death.

This is the first major international guideline to address cardiomyopathies other than HCM. Other major innovations include:

  • A new phenotypic description of cardiomyopathies, including updated descriptions of dilated and non-dilated left ventricular (LV) cardiomyopathy phenotypes, and highlighting the key role of ventricular myocardial scar assessment using cardiac magnetic resonance (CMR) imaging.

  • A focus on the patient pathway, from presentation, through initial assessment and diagnosis, to management, highlighting the importance of considering cardiomyopathy as a cause of common clinical presentations (e.g. heart failure, arrhythmia) and the importance of utilizing a multiparametric approach following the identification of the presenting phenotype to arrive at an aetiological diagnosis.

  • Updated recommendations for clinical and genetic cascade screening for relatives of individuals with cardiomyopathies.

  • A focus on cardiomyopathies across the life course, from paediatric to adult age (including transition), and considering the different clinical phases (e.g. concealed, overt, end stage).

  • New recommendations on sudden cardiac death (SCD) risk stratification for different cardiomyopathy phenotypes, including in childhood, and highlighting the important role of genotype in the assessment of sudden death risk.

  • Updated recommendations for the management of left ventricular outflow tract obstruction (LVOTO) in HCM.

  • A multidisciplinary approach to cardiomyopathies that has the patient and their family at its heart.

3. Phenotypic approach to cardiomyopathies

In medicine, classification systems are used to standardize disease nomenclature by grouping disorders according to shared characteristics. In 2008, the ESC promoted a pragmatic system for the clinical description of cardiomyopathies in which a historical focus on ventricular morphology and function was maintained, while signposting aetiological diversity through subdivision into genetic and non-genetic subtypes.2 Since then, knowledge of cardiomyopathies has increased substantially through the application of new imaging and molecular technologies.

In this guideline, the Task Force took a number of considerations into account when deciding its approach to disease description. These included: (i) a historical legacy which, while still useful, has led to contradictory and confusing terminology in many situations; (ii) the evolving nature of cardiomyopathies over a lifetime; (iii) aetiological complexity with multiple disease processes contributing to disease phenotypes; (iv) differential disease expression in families; and (v) emerging aetiology-focused therapies.

The Task Force concluded that a single classification system that embraces all possible causes of disease and every clinical scenario remains an aspiration that is outside the scope of this clinical guideline. Instead, the Task Force updated the existing clinical classification to include new phenotypic descriptions and to simplify terminology, while simultaneously providing a conceptual framework for diagnosis and treatment. This nomenclature prompts clinicians to consider cardiomyopathy as the cause of several clinical presentations (e.g. arrhythmia, heart failure), and focuses on morphological and functional characteristics of the myocardium (Figure 2). It is important to recognize that different cardiomyopathy phenotypes may coexist in the same family, and that disease progression in an individual patient can include evolution from one cardiomyopathy phenotype to another. Nevertheless, the Task Force recommends an approach to disease nomenclature and diagnosis that is based on the predominant cardiac phenotype at presentation.

Clinical diagnostic workflow of cardiomyopathy.
Figure 2

Clinical diagnostic workflow of cardiomyopathy.

ARVC, arrhythmogenic right ventricular cardiomyopathy; CMP, cardiomyopathy; CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; ECG, electrocardiogram; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy.

While recognizing the fact that genes encoding cardiac ion channels may be implicated in some patients with dilated cardiomyopathy (DCM), conduction disorders, and arrhythmias, the Task Force was not persuaded that there is sufficient evidence to consider cardiac channelopathies as cardiomyopathies, in keeping with the approach taken by other recent ESC Guidelines.3

The most important changes in this guideline relate to the group of conditions variously included under the umbrella term ‘arrhythmogenic cardiomyopathies’. This term refers to a group of conditions that feature structural and functional abnormalities of the myocardium (identified by cardiac imaging and/or macroscopic and microscopic pathological investigation) and ventricular arrhythmia. This nosology has evolved in response to the recognition of the clinical and genetic overlap between right ventricular (RV) and LV cardiomyopathies, but a lack of a generally accepted definition has meant that the term encompasses a broad range of diverse pathologies and has introduced a number of inconsistencies and contradictions when applied in a clinical setting.4 The term ‘arrhythmogenic right ventricular (dysplasia/) cardiomyopathy’ (ARVC) was originally used by physicians who first discovered the disease, in the pre-genetic and pre-CMR era, to describe a new heart muscle disease predominantly affecting the right ventricle, whose cardinal clinical manifestation was the occurrence of malignant ventricular arrhythmias. Subsequently, autopsy investigations, genotype–phenotype correlation studies and the increasing use of contrast-enhancement CMR led to the identification of fibro-fatty replacement of the myocardium as a key phenotypic feature of the disease that affects the myocardium of both ventricles, with LV involvement which may even exceed the severity of RV involvement. This has led to the catch-all term of arrhythmogenic cardiomyopathy (ACM), which represents the evolution of the original term of ARVC.5 Consistent with its general approach, the Task Force agreed to highlight the vital importance of arrhythmia as a diagnostic red flag and prognostic marker across a range of clinical phenotypes, but did not recommend the use of the term ACM as a distinct cardiomyopathy subtype as it lacks a morphological or functional definition consistent with the existing classification scheme. While acknowledging that ‘ACM’ as an umbrella term that encompasses diverse clinical phenotypes has been previously used, this decision will, it is hoped, help to resolve many of the circular arguments that currently bedevil the field. The fundamental tenet throughout this guideline is that aetiology is vital to the management of patients with heart muscle disease and that a careful and consistent description of the morphological and functional phenotype is a crucial first step in the diagnostic pathway, while the final diagnosis will ideally describe aetiology alongside the phenotype.6,7

3.1. Definitions

A cardiomyopathy is defined as ‘a myocardial disorder in which the heart muscle is structurally and functionally abnormal, in the absence of coronary artery disease (CAD), hypertension, valvular disease, and congenital heart disease (CHD) sufficient to cause the observed myocardial abnormality’.2 This definition applies to both children and adults and makes no a priori assumptions about aetiology (which can be familial/genetic or acquired) or myocardial pathology. While the focus of this guideline is on genetic cardiomyopathies, the systematic approach to diagnosis starting from the phenotype at presentation described in this guideline enables clinicians to reach precise diagnoses that may also include non-genetic (e.g. inflammatory, toxic, and multisystem diseases) causes. It is important to note that cardiomyopathies can coexist with ischaemic, valvular, and hypertensive disease and that the presence of one does not exclude the possibility of the other.

The morphological and functional traits used to describe the cardiomyopathy phenotypes are shown in Table 3. The major innovation is the specific inclusion of myocardial tissue characterization traits, including non-ischaemic ventricular scarring or fatty replacement, which can occur with and without ventricular dilatation, wall motion abnormalities, or global systolic or diastolic dysfunction. This phenotype is important to recognize, as it may be the sole clue to the diagnosis of a cardiomyopathy and has prognostic significance that varies with the underlying aetiology.

Table 3

Morphological and functional traits used to describe cardiomyopathy phenotypes

Morphological traits
Ventricular hypertrophy: left and/or right
Ventricular dilatation: left and/or right
Non-ischaemic ventricular scar and other myocardial tissue characterization features on cardiac magnetic resonance
Functional traits
Ventricular systolic dysfunction (global, regional)
Ventricular diastolic dysfunction (restrictive physiology)
Morphological traits
Ventricular hypertrophy: left and/or right
Ventricular dilatation: left and/or right
Non-ischaemic ventricular scar and other myocardial tissue characterization features on cardiac magnetic resonance
Functional traits
Ventricular systolic dysfunction (global, regional)
Ventricular diastolic dysfunction (restrictive physiology)
Table 3

Morphological and functional traits used to describe cardiomyopathy phenotypes

Morphological traits
Ventricular hypertrophy: left and/or right
Ventricular dilatation: left and/or right
Non-ischaemic ventricular scar and other myocardial tissue characterization features on cardiac magnetic resonance
Functional traits
Ventricular systolic dysfunction (global, regional)
Ventricular diastolic dysfunction (restrictive physiology)
Morphological traits
Ventricular hypertrophy: left and/or right
Ventricular dilatation: left and/or right
Non-ischaemic ventricular scar and other myocardial tissue characterization features on cardiac magnetic resonance
Functional traits
Ventricular systolic dysfunction (global, regional)
Ventricular diastolic dysfunction (restrictive physiology)

Atrial dilatation (left and/or right) is an important additional clinical finding in the phenotypic description of cardiomyopathies. Ultra-rare, usually autosomal recessive, cases of pure dilated atrial cardiomyopathy are reported,8 but these are outside the scope of this guideline.

3.2. Cardiomyopathy phenotypes

3.2.1. Hypertrophic cardiomyopathy

Hypertrophic cardiomyopathy (HCM) is defined as the presence of increased LV wall thickness (with or without RV hypertrophy) or mass that is not solely explained by abnormal loading conditions.2

3.2.2. Dilated cardiomyopathy

Dilated cardiomyopathy (DCM) is defined as the presence of LV dilatation and global or regional systolic dysfunction unexplained solely by abnormal loading conditions (e.g. hypertension, valve disease, CHD) or CAD.2 Very rarely, LV dilatation can occur with normal ejection fraction (EF) in the absence of athletic remodelling or other environmental factors; this is not in itself a cardiomyopathy, but may represent an early manifestation of DCM. The preferred term for this is isolated left ventricular dilatation.

Right ventricular dilatation and dysfunction may be present but are not necessary for the diagnosis. When dilatation or wall motion abnormalities are confined or predominant to the right ventricle, the possibility of ARVC should be considered (see Section 3.2.4).

3.2.3. Non-dilated left ventricular cardiomyopathy

Hitherto, the definition of DCM had a number of important limitations, most notably the exclusion of genetic and acquired disorders that manifest as intermediate phenotypes that do not meet standard disease definitions in spite of the presence of myocardial disease on cardiac imaging or tissue analysis. In a previous ESC statement, this phenomenon inspired the creation of a new disease category, hypokinetic non-dilated cardiomyopathy.9 In this guideline, we propose replacement of this term with non-dilated left ventricular cardiomyopathy (NDLVC), which can be further characterized by the presence or absence of systolic dysfunction (regional or global). Isolated LV dysfunction (regional or global) without scarring should also be considered under this diagnostic category. The NDLVC phenotype is defined as the presence of non-ischaemic LV scarring or fatty replacement regardless of the presence of global or regional wall motion abnormalities (RWMAs), or isolated global LV hypokinesia without scarring.

The NDLVC phenotype will include individuals that up until now may have variably been described as having DCM (but without LV dilatation), arrhythmogenic left ventricular cardiomyopathy (ALVC), left-dominant ARVC, or arrhythmogenic DCM (but often without fulfilling diagnostic criteria for ARVC) (Figure 3). The simple worked example (Figure 4) shows how the identification of an NDLVC phenotype should trigger a multiparametric approach that leads to a specific aetiological diagnosis, with implications for clinical treatment.

Examples of non-dilated left ventricular cardiomyopathy phenotypes and their aetiological correlates.
Figure 3

Examples of non-dilated left ventricular cardiomyopathy phenotypes and their aetiological correlates.

BAG3, BAG cochaperone-3; DMD, Duchenne muscular dystrophy; DSP, desmoplakin; FLNC, filamin C; LGE, late gadolinium enhancement; LMNA, lamin A/C; NDLVC, non-dilated left ventricular cardiomyopathy; PLN, phospholamban; RBM20, RNA binding motif protein 20; TTN, titin. Distribution of LGE (arrowheads) in NDLVC and aetiological correlates. Desmoplakin (DSP), filamin C (FLNC) and phospholamban (PLN) genotypes show a characteristic subepicardial, ring-like LGE pattern, whereas titin (TTN), BAG3 (BAG3), lamin A/C (LMNA), DMD, RBM20 genotypes and myocarditis are more heterogeneous, but with overall less scar (sometimes without) and lower left ventricular ejection fraction.

Worked example of the non-dilated left ventricular cardiomyopathy phenotype.
Figure 4

Worked example of the non-dilated left ventricular cardiomyopathy phenotype.

ACM, arrhythmogenic cardiomyopathy; ALVC, arrhythmogenic left ventricular cardiomyopathy; DCM, dilated cardiomyopathy; DSP, desmoplakin; ECG, electrocardiogram; EF, ejection fraction; LV, left ventricular; NDLVC, non-dilated left ventricular cardiomyopathy; SCD, sudden cardiac death; VE, ventricular extrasystole. Worked example of the NDLVC phenotype showing how a systematic multiparametric approach to clinical phenotyping, starting from the recognition of a clinical phenotype and integrating extended phenotypic information and targeted diagnostics, including genetic testing, can be used to arrive at highly specific phenotypic descriptions that can result in personalized treatment plans. In this worked example, the diagnosis transforms from a simplistic categorization to a complex genetic disorder characterized by myocardial scar and a propensity to ventricular arrhythmia.

3.2.4. Arrhythmogenic right ventricular cardiomyopathy

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is defined as the presence of predominantly RV dilatation and/or dysfunction in the presence of histological involvement and/or electrocardiographic abnormalities in accordance with published criteria.10

For decades, ARVC has been one of the principal cardiomyopathy subtypes. It has been defined in accordance with published consensus criteria that comprise RV dysfunction (global or regional), histological abnormalities in the form of fibro-fatty replacement of cardiomyocytes, electrocardiographic characteristics, ventricular arrhythmia of RV origin, and the presence of familial disease and/or pathogenic variants in desmosomal protein genes.

Over time, the clinical paradigm of ARVC has moved from a focus on severe RV disease and malignant ventricular arrhythmia to a broader concept that includes concealed or subclinical phenotypes and biventricular or even left-dominant disease. This has led to a plethora of new terms, including ‘arrhythmogenic left ventricular cardiomyopathy (ALVC)’, ‘left and right dominant cardiomyopathy’, ‘arrhythmogenic dilated cardiomyopathy’, and most recently, the catch-all term ‘arrhythmogenic cardiomyopathy’. The term ARVC can be used to describe the original variant in which ventricular dilatation or wall motion abnormalities are predominantly confined to the right ventricle, with or without LV involvement, and the 2010 modified Task Force criteria for the diagnosis of ARVC can be applied.10 Predominant LV disease can also occur in the same family;5 see Section 7.3 for recommendations on assessment and management of this phenotype.

3.2.5. Restrictive cardiomyopathy

Restrictive cardiomyopathy (RCM) is defined as restrictive left and/or RV pathophysiology in the presence of normal or reduced diastolic volumes (of one or both ventricles), normal or reduced systolic volumes, and normal ventricular wall thickness.2

Restrictive cardiomyopathy commonly presents as biatrial enlargement. Left ventricular systolic function can be preserved, but it is rare for contractility to be completely normal. Restrictive pathophysiology may not be present throughout the natural history, but only at an initial stage (with an evolution towards a hypokinetic-dilated phase).11 Restrictive physiology can also occur in patients with end-stage hypertrophic and dilated cardiomyopathy; the preferred terms are ‘hypertrophic’ or ‘dilated cardiomyopathy with restrictive physiology’. Restrictive ventricular physiology can also be caused by endocardial pathology (fibrosis, fibroelastosis, and thrombosis) that impairs diastolic function.

3.3. Other traits and syndromes associated with cardiomyopathy phenotypes

3.3.1. Left ventricular hypertrabeculation (left ventricular non-compaction)

The term ‘left ventricular non-compaction’ (LVNC) has been used to describe a ventricular phenotype characterized by prominent LV trabeculae and deep intertrabecular recesses. The myocardial wall is often thickened with a thin, compacted epicardial layer and a thicker endocardial layer. In some patients, this abnormal trabecular architecture is associated with LV dilatation and systolic dysfunction. Left ventricular non-compaction is frequently a familial trait and is associated with variants in a range of genes, including those encoding proteins of the sarcomere, Z-disc, cytoskeleton, and nuclear envelope.12–16

Left ventricular non-compaction has also been used to describe an acquired and sometimes transient phenomenon of excessive LV trabeculation (e.g. in athletes, during pregnancy, or following vigorous activity)17–19 that must reflect increased prominence of an otherwise normal myocardial architecture, given that cardiomyocytes are terminally differentiated and the formation of new cardiac structures is impossible.20

The Task Force does not consider LVNC to be a cardiomyopathy in the general sense. Instead, it is seen as a phenotypic trait that can occur either in isolation or in association with other developmental abnormalities, ventricular hypertrophy, dilatation, and/or systolic dysfunction. Given the lack of morphometric evidence for ventricular compaction in humans,21,22 the term ‘hypertrabeculation’, rather than LVNC, is recommended, particularly when the phenomenon is transient or clearly of adult onset.

3.3.2. Takotsubo syndrome

Transient LV apical ballooning syndrome, or takotsubo syndrome, is characterized, in its most typical variant, by transient regional systolic dysfunction, dilatation, and oedema involving the LV apex and/or mid-ventricle in the absence of obstructive coronary disease on coronary angiography.23 Patients present with an abrupt onset of angina-like chest pain and have diffuse T wave inversion (TWI), sometimes preceded by ST-segment elevation and mild cardiac enzyme elevation. Most reported cases occur in post-menopausal women. Symptoms are often preceded by emotional or physical stress. Norepinephrine concentration is elevated in most patients and a transient, dynamic outflow tract pressure gradient is reported in some cases. Left ventricular function usually normalizes over a period of days to weeks, and recurrence is rare. The same kind of reversible myocardial dysfunction is occasionally encountered in patients with intracranial haemorrhage or other acute cerebral accidents (neurogenic myocardial stunning).

Takotsubo syndrome is sometimes referred to as takotsubo or stress cardiomyopathy. Given the transient nature of the phenomenon, the Task Force does not recommend its classification as a cardiomyopathy.

4. Epidemiology

Cardiomyopathies have a variable expression throughout life.24 Geographical distribution of genetic variants influences estimated prevalence in different populations, ethnicities, regions, and countries. The complexity of diagnostic criteria for some conditions, such as ARVC, limits the evaluation of the true prevalence of the disease in the general population. Moreover, epidemiological data are often not collected systematically at population level. For example, the prevalence of idiopathic DCM has been recently estimated to be almost 10 times higher based on several population-based estimates and indirect assumptions of the prevalence of genetic variants associated with the disease in general populations,25 and with less stringent diagnostic criteria.9

There are no specific data on the epidemiology of the NDLVC phenotype, but patients affected by it have previously been included in DCM or ARVC cohorts, from which extrapolations may be possible. Contemporary epidemiological metrics for the main cardiomyopathies are shown in Table 4. Further details on the epidemiology of cardiomyopathies can be found in the Supplementary data online, Section 1.

Table 4

Key epidemiological metrics in adults and children for the different cardiomyopathy phenotypes

Cardiomyopathy phenotypeAdultsChildren
HCMPrevalence: 0.2%26–33Childhood incidence: 0.002–0.005%34–36
Childhood prevalence: 0.029%36
DCMPrevalence: 0.036–0.400%25,37Childhood incidence: 0.003–0.006%
Childhood prevalence: 0.026%36
Infantile incidence: 0.038–0.046%34–36,38
NDLVCTo be determinedTo be determined
ARVCPrevalence: 0.078%39–41Very rare in infancy and early childhood; to be determined in older children and adolescents
RCMRareChildhood incidence: 0.0003%34
Cardiomyopathy phenotypeAdultsChildren
HCMPrevalence: 0.2%26–33Childhood incidence: 0.002–0.005%34–36
Childhood prevalence: 0.029%36
DCMPrevalence: 0.036–0.400%25,37Childhood incidence: 0.003–0.006%
Childhood prevalence: 0.026%36
Infantile incidence: 0.038–0.046%34–36,38
NDLVCTo be determinedTo be determined
ARVCPrevalence: 0.078%39–41Very rare in infancy and early childhood; to be determined in older children and adolescents
RCMRareChildhood incidence: 0.0003%34

ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy.

© ESC 2023
Table 4

Key epidemiological metrics in adults and children for the different cardiomyopathy phenotypes

Cardiomyopathy phenotypeAdultsChildren
HCMPrevalence: 0.2%26–33Childhood incidence: 0.002–0.005%34–36
Childhood prevalence: 0.029%36
DCMPrevalence: 0.036–0.400%25,37Childhood incidence: 0.003–0.006%
Childhood prevalence: 0.026%36
Infantile incidence: 0.038–0.046%34–36,38
NDLVCTo be determinedTo be determined
ARVCPrevalence: 0.078%39–41Very rare in infancy and early childhood; to be determined in older children and adolescents
RCMRareChildhood incidence: 0.0003%34
Cardiomyopathy phenotypeAdultsChildren
HCMPrevalence: 0.2%26–33Childhood incidence: 0.002–0.005%34–36
Childhood prevalence: 0.029%36
DCMPrevalence: 0.036–0.400%25,37Childhood incidence: 0.003–0.006%
Childhood prevalence: 0.026%36
Infantile incidence: 0.038–0.046%34–36,38
NDLVCTo be determinedTo be determined
ARVCPrevalence: 0.078%39–41Very rare in infancy and early childhood; to be determined in older children and adolescents
RCMRareChildhood incidence: 0.0003%34

ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy.

© ESC 2023

4.1. Special populations

Several forms of cardiomyopathy previously considered secondary to external factors were recently proved to have genetic contributors, leading to the ‘second hit theory’, and a genetic aetiology should be kept in mind for family history taking and genetic testing.

  • Titin gene truncating variants (TTNtv) represent a prevalent genetic predisposition for alcoholic cardiomyopathy (present in 13.5% of patients vs. 2.9% in controls), as they are associated with a worse left ventricular ejection fraction (LVEF) in DCM patients who consume alcohol above recommended levels.42

  • Unrecognized rare variants in cardiomyopathy-associated genes, particularly TTNtv (in 7.5% of cases), appear to be associated with an increased risk of cancer therapy-induced cardiomyopathy in children and adults.43

  • Rare truncating variants in eight genes are found in 15% of women with peripartum cardiomyopathy (PPCM), and two-thirds are TTNtv (10% of patients vs. 1.4% of the reference population).44,45 Additionally, other truncating variants are identified in the DSP (1%), FLNC (1%), and BAG3 (0.2%) genes.45

  • Anderson–Fabry disease is found in 0.94% of males and 0.90% of females in cardiac screening programmes for left ventricular hypertrophy (LVH) in selected populations and HCM.46

  • Screening with bone scintigraphy found a high prevalence of transthyretin cardiac amyloidosis (ATTR-CA) in specific populations: 8% in severe aortic stenosis, 12% in heart failure with preserved ejection fraction (HFpEF) with LVH, 7% in LVH/HCM depending on the age, and 7% in carpal tunnel syndrome undergoing surgery (a higher prevalence if it is bilateral), mainly for the wild-type form.47,48

  • Disease-causing variants in genes implicated in DCM, NDLVC, and ARVC have been identified in 8–22% of adults and children presenting with acute myocarditis.49–51 Individuals with an acute myocarditis presentation and desmosomal protein gene variants were shown to have a higher rate of myocarditis recurrence and ventricular arrhythmia compared with myocarditis patients without a desmosomal variant identified.52

5. Integrated patient management

The diagnosis, assessment, and management of patients with cardiomyopathy requires a co-ordinated, systematic, and individualized pathway that delivers optimized care by a multidisciplinary and expert team. Central to this approach is not only the individual patient, but also the family as a whole; clinical findings in relatives are essential for understanding what happens to the patient, and vice versa.53,54

5.1. Multidisciplinary cardiomyopathy teams

Healthcare professionals encounter diseases affecting the myocardium in many and varied clinical settings. Some may manifest for the first time with an acute event, including sudden unexplained death, whereas others present with progressive symptoms or are detected incidentally. Patients with cardiomyopathy can also have extracardiac manifestations (e.g. neurological, neuromuscular, ophthalmological, nephrological). Patient care requires the collaboration of different specialties.55 The composition of the multidisciplinary team will depend on the patient’s and family’s needs and the local availability of services (Figure 5). Patients with complex needs benefit from a multidisciplinary team, including relevant specialties as well as the general cardiologist, general practitioner, and the family/carer. In addition, the integration of genetics into mainstream cardiology services requires expertise from different specialties:

  • Adult and paediatric cardiologists subspecialized in cardiogenetic conditions.

  • Cardiac imaging specialists (technicians, cardiologists, radiologists), including CMR experts.

  • Specialist nurses and/or genetic counsellors with skills in family history taking, drawing pedigrees, and patient/family management, particularly when the number of disciplines or the complexity implicated in a patient’s/family’s care increases.

  • Clinical psychologists to support patients and their relatives.

  • Geneticists and bioinformaticians to interpret results of genetic investigations.

  • Expert pathologists to interpret findings by endomyocardial biopsy (EMB) and autopsy of individuals dying from a suspected inherited cardiac condition. Specialist cardiovascular pathology centres play a crucial role in the autopsy diagnosis of cardiomyopathy when local expertise is not available.56,57

Multidisciplinary care of cardiomyopathies.
Figure 5

Multidisciplinary care of cardiomyopathies.

aThe list presented is not exhaustive and represents examples of specialties that often interact in the care of cardiomyopathy patients.

Finally, patients’ associations should be promoted and integrated into the healthcare process for rare and very rare cardiac conditions.

One particularly important aspect of the multidisciplinary approach to patient care in cardiomyopathies is the need for appropriate transition of care from paediatric to adult services. Children with a genetic cardiomyopathy generally need lifelong cardiac follow-up. The transition to adulthood, including the transfer of care to adult cardiomyopathy services, can be challenging for both the child and the parents. The process of transition should include adequate and timely preparation and joint consultations, taking into consideration the child’s wishes, and level of understanding and independence at different life stages. Evidence from the field of CHD highlights the importance of specific interventions that can help the process of transition of clinical care, including adequate and timely preparation for transition and joint consultations.58,59

5.2. Co-ordination between different levels of care

A shared care approach between cardiomyopathy specialists and general adult and paediatric cardiology centres is strongly recommended. While referral cardiomyopathy units are essential for complex cases with diagnostic and/or treatment difficulties that require expertise that may only be available in high-volume centres, general adult and paediatric cardiologists have a key role to play in the diagnosis, management, and follow-up of patients with cardiomyopathy (see Section 9). A shared approach between cardiomyopathy units and between general cardiologist/paediatric cardiologist is strongly recommended. This approach can be facilitated by the implementation of telemedical contact between units and the use of remote monitoring with patients.60 The creation of local/regional/national/international networks, such as the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart) (https://guardheart.ern-net.eu) allows clinicians and health professionals to share information about these pathologies, for the benefit of cardiomyopathy patients.61

Recommendation Table 1

Recommendations for the provision of service of multidisciplinary cardiomyopathy teams

graphic
graphic
Recommendation Table 1

Recommendations for the provision of service of multidisciplinary cardiomyopathy teams

graphic
graphic

6. The patient pathway

The diagnosis of cardiomyopathy rests on the identification of structural and/or functional myocardial abnormalities, including myocardial fibrosis, that are not explained solely by abnormal loading conditions or CAD. However, disease phenotypes can also include arrhythmic and electrocardiographic manifestations, morphological abnormalities of the cardiac valves, and abnormal coronary microcirculatory function. As a key theme throughout this guideline, the Task Force highlights the importance of using a systematic approach to the identification and assessment of patients with a suspected cardiomyopathy. Central to this is the need for clinicians to consider a diagnosis of cardiomyopathy as the cause of several common adult and paediatric clinical presentations. The identification of a cardiomyopathy phenotype is only the beginning of the diagnostic process and should prompt a systematic search for the underlying aetiology, which may be genetic or acquired.

6.1. Clinical presentation

Patients with cardiomyopathy may access health services through several pathways. Referral from primary care (e.g. general practitioners and general paediatricians) may be triggered by symptoms (most commonly dyspnoea, chest pain, palpitation, syncope) or incidental findings (e.g. an abnormal electrocardiogram [ECG] in the context of community, school, work-related medical check-ups, or sports pre-participation screening; the incidental detection of a murmur; or, increasingly, genotype-first identification as a result of secondary findings during research or clinical sequencing for other indications). In secondary and tertiary care (general cardiology and paediatric cardiology), patients with cardiomyopathy may present to the heart failure clinic with symptoms of heart failure with reduced ejection fraction (HFrEF), mildly reduced ejection fraction (HFmrEF), or preserved ejection fraction (HFpEF); to the arrhythmia clinic with early-onset conduction disease, atrial arrhythmia, or ventricular arrhythmia; or to the emergency department with suspected myocarditis. Frequently, patients enter the cardiomyopathy pathway in primary, secondary, or tertiary care as a result of family screening following the diagnosis of cardiomyopathy or a sudden death in a relative, and may also be identified as part of the work-up for multiorgan disease known to be associated with cardiovascular involvement. Clinicians in all these settings therefore need to consider the possibility of cardiomyopathy as a cause and use a systematic, cardiomyopathy-oriented approach to clinical evaluation.

6.2. Initial work-up

The cardiomyopathy-oriented approach is based on interpreting clinical and instrumental findings to suspect and ultimately generate a phenotype-based aetiological diagnosis to guide disease-specific management.62 This approach requires deliberate analysis of multiparametric investigations in the individual and their relatives and an integrated probabilistic analysis of clinical investigations. Re-analysis of clinical data is required as new information emerges, and family information can provide important clues to the diagnosis, given the variable expression and incomplete penetrance of most cardiomyopathies, and can result in differences in diagnostic criteria between probands and relatives. In this context, relatives of individuals with cardiomyopathy can have non-diagnostic morphological and electrocardiographic abnormalities that can indicate mild and early phenotypic expression of disease and can increase diagnostic accuracy for predicting disease in genotyped populations. The identification of diagnostic clues, or red flags, is a crucial aspect of the initial work-up.

6.3. Systematic approach to diagnosis of cardiomyopathy

A multiparametric approach to the evaluation of patients with suspected cardiomyopathy is recommended, with the aims of: (i) establishing and characterizing the presence of a cardiomyopathy phenotype; and (ii) identifying the underlying aetiological diagnosis.62 Clinicians should approach a patient with suspected cardiomyopathy using a ‘cardiomyopathy mindset’ (Figure 2):

  • Use multimodality imaging to characterize the phenotype and identify abnormal ventricular morphology (e.g. hypertrophy, dilatation) and function (systolic/diastolic, global/regional), and detect abnormalities of tissue characterization (e.g. non-ischaemic myocardial scar and fatty replacement).

  • Use a combination of personal and family history, clinical examination, electrocardiography, and laboratory investigations to achieve an aetiological diagnosis, looking for specific signs and symptoms and laboratory markers suggestive of a specific diagnosis; the presence of ventricular and atrial arrhythmia and conduction disease to aid diagnosis, suggest specific causes, and monitor disease progression and risk stratification; and clues from the pedigree to suggest specific inheritance patterns and identify at-risk relatives. This approach should result in a timely and accurate diagnosis to enable early treatment of symptoms and prevention of disease-related complications.

Recommendation Table 2

Recommendations for diagnostic work-up in cardiomyopathies

graphic
graphic
Recommendation Table 2

Recommendations for diagnostic work-up in cardiomyopathies

graphic
graphic

6.4. History and physical examination

Age is one of the most important factors to take into account when considering the possible causes of cardiomyopathy. For example, inherited metabolic disorders and congenital dysmorphic syndromes are more common in neonates and infants (see Section 6.9.1) than in older children or adults, whereas wild-type transthyretin amyloidosis (ATTRwt) is a disease mostly of adults over the age of 65 years (see Section 7.6).

Construction of a three- to four-generation family pedigree helps to identify Mendelian forms of inheritance and identifies other family members who may be at risk of disease development.62 Specific features to note in the family history include premature deaths (taking into account that SCDs may sometimes be reported as accidental deaths, e.g. drowning, unexplained traffic accident, and, rarely, as stillbirth or sudden infant death syndromes), unexplained heart failure, cardiac transplantation, pacemaker and defibrillator implants, and evidence for systemic disease (e.g. stroke at a young age, skeletal muscle weakness, renal dysfunction, diabetes, deafness). Most Mendelian forms of cardiomyopathy are autosomal dominant and are therefore characterized by the presence of affected individuals across generations, with transmission from parents of either sex (including male-to-male) and a 50% risk of allele transmission to offspring (although, due to incomplete penetrance, the proportion of affected individuals in an individual pedigree will be lower). X-linked inheritance should be suspected if males are the most severely affected individuals and there is no male-to-male transmission. Autosomal recessive inheritance, the least common pattern, is likely when both parents of the proband are unaffected and consanguineous, although severe autosomal recessive cardiomyopathies can also occur in the absence of familial consanguinity.67,68 When women—but not men—transmit the disease to children of either sex, mitochondrial DNA variants should be considered (Table 5). It is important to note that the absence of familial disease does not exclude a genetic origin (see Section 6.8).

Table 5

Examples of inheritance patterns that should raise the suspicion of specific genetic aetiologies, grouped according to cardiomyopathy phenotype

Cardiomyopathy phenotypeADARX-linkedMatrilineal
HCMSarcomericX
Anderson–FabryX
DanonX
TTR amyloidosisX
RASopathyX(X)
Friedreich ataxiaX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
DCMLMNAX
RBM20X
SarcomericX
DystrophinX
EmerinX
Barth syndromeX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
NDLVCLMNAX
DESXX
FLNCX
PLNX
TMEM43X
RBM20X
ARVCPLNX
DesmosomalXX
TMEM43X
RCMSarcomericX
DESXX
FLNCX
BAG3X
RASopathyX(X)
Cardiomyopathy phenotypeADARX-linkedMatrilineal
HCMSarcomericX
Anderson–FabryX
DanonX
TTR amyloidosisX
RASopathyX(X)
Friedreich ataxiaX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
DCMLMNAX
RBM20X
SarcomericX
DystrophinX
EmerinX
Barth syndromeX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
NDLVCLMNAX
DESXX
FLNCX
PLNX
TMEM43X
RBM20X
ARVCPLNX
DesmosomalXX
TMEM43X
RCMSarcomericX
DESXX
FLNCX
BAG3X
RASopathyX(X)

AD, autosomal dominant; AR, autosomal recessive; ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy; TTR, transthyretin; DNA, deoxyribonucleic acid; RASopathies, Ras/mitogen-activated protein kinase pathway dysregulation.

(X) indicates the presence of a correlation between a cardiomyopathy and a pattern of inheritance.

Table 5

Examples of inheritance patterns that should raise the suspicion of specific genetic aetiologies, grouped according to cardiomyopathy phenotype

Cardiomyopathy phenotypeADARX-linkedMatrilineal
HCMSarcomericX
Anderson–FabryX
DanonX
TTR amyloidosisX
RASopathyX(X)
Friedreich ataxiaX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
DCMLMNAX
RBM20X
SarcomericX
DystrophinX
EmerinX
Barth syndromeX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
NDLVCLMNAX
DESXX
FLNCX
PLNX
TMEM43X
RBM20X
ARVCPLNX
DesmosomalXX
TMEM43X
RCMSarcomericX
DESXX
FLNCX
BAG3X
RASopathyX(X)
Cardiomyopathy phenotypeADARX-linkedMatrilineal
HCMSarcomericX
Anderson–FabryX
DanonX
TTR amyloidosisX
RASopathyX(X)
Friedreich ataxiaX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
DCMLMNAX
RBM20X
SarcomericX
DystrophinX
EmerinX
Barth syndromeX
Mitochondrial
 Mitochondrial DNAX
 Nuclear DNAXXX
NDLVCLMNAX
DESXX
FLNCX
PLNX
TMEM43X
RBM20X
ARVCPLNX
DesmosomalXX
TMEM43X
RCMSarcomericX
DESXX
FLNCX
BAG3X
RASopathyX(X)

AD, autosomal dominant; AR, autosomal recessive; ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy; TTR, transthyretin; DNA, deoxyribonucleic acid; RASopathies, Ras/mitogen-activated protein kinase pathway dysregulation.

(X) indicates the presence of a correlation between a cardiomyopathy and a pattern of inheritance.

Patients with cardiomyopathy may experience dyspnoea, chest pain, palpitation, and syncope and/or pre-syncope, although many individuals complain of few, if any, symptoms (see Section 6.4 for assessment of symptoms in specific cardiomyopathy subtypes). A number of non-cardiac symptoms act as pointers for specific diagnoses (Table 6). Similarly, general physical examination can provide diagnostic clues in patients with syndromic or metabolic causes of cardiomyopathy.62

Table 6

Examples of signs and symptoms that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype

FindingCardiomyopathy phenotype
HCMDCMNDLVCARVCRCM
Learning difficulties, developmental delayMitochondrial diseasesDystrophinopathiesNoonan syndrome
Noonan syndromeMitochondrial diseases
Danon diseaseMyotonic dystrophy
FKTN variants
Sensorineural deafnessMitochondrial diseasesEpicardin variants
NSMLMitochondrial diseases
Visual impairmentMitochondrial diseasesCRYAB
ATTRv or hereditary ATTRType 2 myotonic dystrophy
Danon disease
Anderson–Fabry diseasea
Gait disturbanceFriedreich ataxiaDystrophinopathiesMyofibrillar myopathies
Sarcoglycanopathies
Myofibrillar myopathies
MyotoniaMyotonic dystrophy
Paraesthesia/sensory abnormalities/neuropathic painAmyloidosisAmyloidosis
Anderson–Fabry disease
Carpal tunnel syndromeTTR-related amyloidosis
Muscle weaknessMitochondrial diseasesDystrophinopathiesLaminopathiesDesminopathies
GlycogenosesSarcoglycanopathiesDesminopathies
FHL1 variantsLaminopathies
Myotonic dystrophy
Desminopathies
Palpebral ptosisMitochondrial diseasesMitochondrial diseases
Myotonic dystrophy
LentiginesNSML
AngiokeratomataAnderson–Fabry disease
Pigmentation of skin and scarsHaemochromatosis
Palmoplantar keratoderma and woolly hairCarvajal syndromeNaxos and Carvajal syndromes
DSP variantsDSP variantsDSP variants
FindingCardiomyopathy phenotype
HCMDCMNDLVCARVCRCM
Learning difficulties, developmental delayMitochondrial diseasesDystrophinopathiesNoonan syndrome
Noonan syndromeMitochondrial diseases
Danon diseaseMyotonic dystrophy
FKTN variants
Sensorineural deafnessMitochondrial diseasesEpicardin variants
NSMLMitochondrial diseases
Visual impairmentMitochondrial diseasesCRYAB
ATTRv or hereditary ATTRType 2 myotonic dystrophy
Danon disease
Anderson–Fabry diseasea
Gait disturbanceFriedreich ataxiaDystrophinopathiesMyofibrillar myopathies
Sarcoglycanopathies
Myofibrillar myopathies
MyotoniaMyotonic dystrophy
Paraesthesia/sensory abnormalities/neuropathic painAmyloidosisAmyloidosis
Anderson–Fabry disease
Carpal tunnel syndromeTTR-related amyloidosis
Muscle weaknessMitochondrial diseasesDystrophinopathiesLaminopathiesDesminopathies
GlycogenosesSarcoglycanopathiesDesminopathies
FHL1 variantsLaminopathies
Myotonic dystrophy
Desminopathies
Palpebral ptosisMitochondrial diseasesMitochondrial diseases
Myotonic dystrophy
LentiginesNSML
AngiokeratomataAnderson–Fabry disease
Pigmentation of skin and scarsHaemochromatosis
Palmoplantar keratoderma and woolly hairCarvajal syndromeNaxos and Carvajal syndromes
DSP variantsDSP variantsDSP variants

ARVC, arrhythmogenic right ventricular cardiomyopathy; ATTR, transthyretin amyloidosis; ATTRv, hereditary transthyretin amyloidosis; DCM, dilated cardiomyopathy; DSP, desmoplakin; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; NSML, Noonan syndrome with multiple lentigines; RCM, restrictive cardiomyopathy; TTR, transthyretin.

Cornea verticillata, characteristic of Anderson–Fabry disease, does not cause visual impairment per se.

Table 6

Examples of signs and symptoms that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype

FindingCardiomyopathy phenotype
HCMDCMNDLVCARVCRCM
Learning difficulties, developmental delayMitochondrial diseasesDystrophinopathiesNoonan syndrome
Noonan syndromeMitochondrial diseases
Danon diseaseMyotonic dystrophy
FKTN variants
Sensorineural deafnessMitochondrial diseasesEpicardin variants
NSMLMitochondrial diseases
Visual impairmentMitochondrial diseasesCRYAB
ATTRv or hereditary ATTRType 2 myotonic dystrophy
Danon disease
Anderson–Fabry diseasea
Gait disturbanceFriedreich ataxiaDystrophinopathiesMyofibrillar myopathies
Sarcoglycanopathies
Myofibrillar myopathies
MyotoniaMyotonic dystrophy
Paraesthesia/sensory abnormalities/neuropathic painAmyloidosisAmyloidosis
Anderson–Fabry disease
Carpal tunnel syndromeTTR-related amyloidosis
Muscle weaknessMitochondrial diseasesDystrophinopathiesLaminopathiesDesminopathies
GlycogenosesSarcoglycanopathiesDesminopathies
FHL1 variantsLaminopathies
Myotonic dystrophy
Desminopathies
Palpebral ptosisMitochondrial diseasesMitochondrial diseases
Myotonic dystrophy
LentiginesNSML
AngiokeratomataAnderson–Fabry disease
Pigmentation of skin and scarsHaemochromatosis
Palmoplantar keratoderma and woolly hairCarvajal syndromeNaxos and Carvajal syndromes
DSP variantsDSP variantsDSP variants
FindingCardiomyopathy phenotype
HCMDCMNDLVCARVCRCM
Learning difficulties, developmental delayMitochondrial diseasesDystrophinopathiesNoonan syndrome
Noonan syndromeMitochondrial diseases
Danon diseaseMyotonic dystrophy
FKTN variants
Sensorineural deafnessMitochondrial diseasesEpicardin variants
NSMLMitochondrial diseases
Visual impairmentMitochondrial diseasesCRYAB
ATTRv or hereditary ATTRType 2 myotonic dystrophy
Danon disease
Anderson–Fabry diseasea
Gait disturbanceFriedreich ataxiaDystrophinopathiesMyofibrillar myopathies
Sarcoglycanopathies
Myofibrillar myopathies
MyotoniaMyotonic dystrophy
Paraesthesia/sensory abnormalities/neuropathic painAmyloidosisAmyloidosis
Anderson–Fabry disease
Carpal tunnel syndromeTTR-related amyloidosis
Muscle weaknessMitochondrial diseasesDystrophinopathiesLaminopathiesDesminopathies
GlycogenosesSarcoglycanopathiesDesminopathies
FHL1 variantsLaminopathies
Myotonic dystrophy
Desminopathies
Palpebral ptosisMitochondrial diseasesMitochondrial diseases
Myotonic dystrophy
LentiginesNSML
AngiokeratomataAnderson–Fabry disease
Pigmentation of skin and scarsHaemochromatosis
Palmoplantar keratoderma and woolly hairCarvajal syndromeNaxos and Carvajal syndromes
DSP variantsDSP variantsDSP variants

ARVC, arrhythmogenic right ventricular cardiomyopathy; ATTR, transthyretin amyloidosis; ATTRv, hereditary transthyretin amyloidosis; DCM, dilated cardiomyopathy; DSP, desmoplakin; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; NSML, Noonan syndrome with multiple lentigines; RCM, restrictive cardiomyopathy; TTR, transthyretin.

Cornea verticillata, characteristic of Anderson–Fabry disease, does not cause visual impairment per se.

6.5. Resting and ambulatory electrocardiography

The resting 12-lead ECG is often the first test that suggests the possibility of cardiomyopathy. Although the ECG can be normal in a small proportion of individuals with cardiomyopathy, standard ECG abnormalities are common in all cardiomyopathy subtypes and can precede the development of an overt morphological or functional phenotype by many years; for example, in genotype-positive individuals identified during family screening. When interpreted in conjunction with findings on echocardiography and CMR imaging, features that would normally indicate other conditions, such as myocardial ischaemia or infarction, can—with age at diagnosis, inheritance pattern, and associated clinical features—suggest an underlying diagnosis or provide clues to the underlying diagnosis. For this reason, the ECG is recommended at the first clinic visit in all individuals with known or suspected cardiomyopathy and should be repeated whenever there is a change in symptoms in patients with an established diagnosis. Although the ECG is often non-specific, there are particular features that can suggest a certain aetiology or morphological diagnosis, including atrioventricular (AV) block, ventricular pre-excitation pattern, distribution of repolarization abnormalities, and high or low QRS voltages (Table 7).

Table 7

Examples of electrocardiographic features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype

Cardiomyopathy phenotypeFindingSpecific diseases to be considered
HCMShort PR interval/pre-excitationGlycogenosis
Danon disease
PRKAG2 cardiomyopathy
Anderson–Fabry disease
Mitochondrial disease
AV blockAmyloidosis
Anderson–Fabry disease (late stage)
Danon disease
Sarcoidosis
PRKAG2 cardiomyopathy
Extreme LVHDanon disease
Glycogenosis (e.g. Pompe disease)
PRKAG2 cardiomyopathy
Low QRS voltageaAmyloidosis
Friedreich ataxia
Superior QRS axis (‘northwest axis’)Noonan syndrome
Q waves/pseudoinfarction patternAmyloidosis
DCMAV blockLaminopathy
Emery–Dreifuss 1
Myocarditis (esp. Chagas disease, Lyme disease, diphtheria)
Sarcoidosis
Desminopathy
Myotonic dystrophy
Low P wave amplitudeEmery–Dreifuss 1 and 2
Atrial standstillEmery–Dreifuss 1 and 2
Posterolateral infarction patternDystrophinopathy
Limb-girdle muscular dystrophy
Sarcoidosis
Extremely low QRS amplitudePLN variant
NDLVCAV blockLaminopathy
Desminopathy
Extremely low QRS amplitudePLN variant
Low QRS voltage + atypical RBBBDesmosomal variants
ARVCT wave inversion V1-V3 + terminal activation delay +/− low right ventricular voltages +/− atypical RBBB
RCMAV blockDesminopathy
Amyloidosis
Cardiomyopathy phenotypeFindingSpecific diseases to be considered
HCMShort PR interval/pre-excitationGlycogenosis
Danon disease
PRKAG2 cardiomyopathy
Anderson–Fabry disease
Mitochondrial disease
AV blockAmyloidosis
Anderson–Fabry disease (late stage)
Danon disease
Sarcoidosis
PRKAG2 cardiomyopathy
Extreme LVHDanon disease
Glycogenosis (e.g. Pompe disease)
PRKAG2 cardiomyopathy
Low QRS voltageaAmyloidosis
Friedreich ataxia
Superior QRS axis (‘northwest axis’)Noonan syndrome
Q waves/pseudoinfarction patternAmyloidosis
DCMAV blockLaminopathy
Emery–Dreifuss 1
Myocarditis (esp. Chagas disease, Lyme disease, diphtheria)
Sarcoidosis
Desminopathy
Myotonic dystrophy
Low P wave amplitudeEmery–Dreifuss 1 and 2
Atrial standstillEmery–Dreifuss 1 and 2
Posterolateral infarction patternDystrophinopathy
Limb-girdle muscular dystrophy
Sarcoidosis
Extremely low QRS amplitudePLN variant
NDLVCAV blockLaminopathy
Desminopathy
Extremely low QRS amplitudePLN variant
Low QRS voltage + atypical RBBBDesmosomal variants
ARVCT wave inversion V1-V3 + terminal activation delay +/− low right ventricular voltages +/− atypical RBBB
RCMAV blockDesminopathy
Amyloidosis

ARVC, arrhythmogenic right ventricular cardiomyopathy; AV, atrioventricular; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVH, left ventricular hypertrophy; NDLVC, non-dilated left ventricular cardiomyopathy; PKP2, plakophilin 2; PLN, phospholamban; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; QRS, Q, R, and S waves of an ECG; RBBB, right bundle branch block; RCM, restrictive cardiomyopathy.

In the absence of obesity, pericardial effusion, chronic obstructive pulmonary disease, abnormalities of the chest, or other reasons that may cause low voltage.

Adapted from Rapezzi et al.62

Table 7

Examples of electrocardiographic features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype

Cardiomyopathy phenotypeFindingSpecific diseases to be considered
HCMShort PR interval/pre-excitationGlycogenosis
Danon disease
PRKAG2 cardiomyopathy
Anderson–Fabry disease
Mitochondrial disease
AV blockAmyloidosis
Anderson–Fabry disease (late stage)
Danon disease
Sarcoidosis
PRKAG2 cardiomyopathy
Extreme LVHDanon disease
Glycogenosis (e.g. Pompe disease)
PRKAG2 cardiomyopathy
Low QRS voltageaAmyloidosis
Friedreich ataxia
Superior QRS axis (‘northwest axis’)Noonan syndrome
Q waves/pseudoinfarction patternAmyloidosis
DCMAV blockLaminopathy
Emery–Dreifuss 1
Myocarditis (esp. Chagas disease, Lyme disease, diphtheria)
Sarcoidosis
Desminopathy
Myotonic dystrophy
Low P wave amplitudeEmery–Dreifuss 1 and 2
Atrial standstillEmery–Dreifuss 1 and 2
Posterolateral infarction patternDystrophinopathy
Limb-girdle muscular dystrophy
Sarcoidosis
Extremely low QRS amplitudePLN variant
NDLVCAV blockLaminopathy
Desminopathy
Extremely low QRS amplitudePLN variant
Low QRS voltage + atypical RBBBDesmosomal variants
ARVCT wave inversion V1-V3 + terminal activation delay +/− low right ventricular voltages +/− atypical RBBB
RCMAV blockDesminopathy
Amyloidosis
Cardiomyopathy phenotypeFindingSpecific diseases to be considered
HCMShort PR interval/pre-excitationGlycogenosis
Danon disease
PRKAG2 cardiomyopathy
Anderson–Fabry disease
Mitochondrial disease
AV blockAmyloidosis
Anderson–Fabry disease (late stage)
Danon disease
Sarcoidosis
PRKAG2 cardiomyopathy
Extreme LVHDanon disease
Glycogenosis (e.g. Pompe disease)
PRKAG2 cardiomyopathy
Low QRS voltageaAmyloidosis
Friedreich ataxia
Superior QRS axis (‘northwest axis’)Noonan syndrome
Q waves/pseudoinfarction patternAmyloidosis
DCMAV blockLaminopathy
Emery–Dreifuss 1
Myocarditis (esp. Chagas disease, Lyme disease, diphtheria)
Sarcoidosis
Desminopathy
Myotonic dystrophy
Low P wave amplitudeEmery–Dreifuss 1 and 2
Atrial standstillEmery–Dreifuss 1 and 2
Posterolateral infarction patternDystrophinopathy
Limb-girdle muscular dystrophy
Sarcoidosis
Extremely low QRS amplitudePLN variant
NDLVCAV blockLaminopathy
Desminopathy
Extremely low QRS amplitudePLN variant
Low QRS voltage + atypical RBBBDesmosomal variants
ARVCT wave inversion V1-V3 + terminal activation delay +/− low right ventricular voltages +/− atypical RBBB
RCMAV blockDesminopathy
Amyloidosis

ARVC, arrhythmogenic right ventricular cardiomyopathy; AV, atrioventricular; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVH, left ventricular hypertrophy; NDLVC, non-dilated left ventricular cardiomyopathy; PKP2, plakophilin 2; PLN, phospholamban; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; QRS, Q, R, and S waves of an ECG; RBBB, right bundle branch block; RCM, restrictive cardiomyopathy.

In the absence of obesity, pericardial effusion, chronic obstructive pulmonary disease, abnormalities of the chest, or other reasons that may cause low voltage.

Adapted from Rapezzi et al.62

Patients with cardiomyopathy may seek cardiology evaluation due to arrhythmia-related symptoms or documented arrhythmia, including bradyarrhythmias and tachyarrhythmias, ranging from symptomatic atrial/ventricular premature beats to life-threating ventricular arrhythmias. The frequency of arrhythmias detected during ambulatory electrocardiographic monitoring is age related and variable across different cardiomyopathy subtypes. Some arrhythmias are relatively common in the context of cardiomyopathy (e.g. atrial fibrillation [AF] or ventricular premature beats), while others may suggest a specific diagnosis. ECG monitoring is therefore useful at the initial clinical assessment and at regular intervals to assess the risk of SCD and stroke.

6.6. Laboratory tests

Routine laboratory testing aids the detection of extracardiac conditions that cause or exacerbate ventricular dysfunction (e.g. thyroid disease, renal dysfunction, and diabetes mellitus) and secondary organ dysfunction in patients with severe heart failure. High levels of brain natriuretic peptide (BNP), N-terminal pro-brain natriuretic peptide (NT-proBNP), and high-sensitivity cardiac troponin T (hs-cTnT) are associated with cardiovascular events, heart failure, and death, and may have diagnostic, prognostic, and therapeutic monitoring value.69 Routine blood tests for comorbidities, including full blood count, renal and liver function parameters and electrolytes, thyroid function, fasting glucose, and Haemoglobin A1C (HbA1c) are recommended in all patients with heart failure symptoms.69 Persistently elevated serum creatinine kinase (CK) levels can be suggestive of myopathies or neuromuscular disorders including dystrophinopathies (e.g. Becker muscular dystrophy or X-linked DCM), laminopathies, desminopathies, or less often, a myofibrillar myopathy.62 Elevated C-reactive protein levels may be present in patients with ARVC and NDLVC, particularly in the context of recurrent myocarditis-like episodes.70 Elevated serum levels of iron and ferritin and high transferrin saturation can suggest a diagnosis of haemochromatosis and should trigger further aetiological refinement (primary vs. secondary) based on genetic testing. Lactic acidosis, myoglobinuria, and leucocytopaenia can be suggestive of mitochondrial diseases. A list of recommended laboratory tests in adults and children is shown in Table 8. Following specialist evaluation, additional tests to detect rare metabolic causes are often required in children, including measurement of lactate, pyruvate, pH, uric acid, ammonia, ketones, free fatty acids, carnitine profile, urine organic acids, and amino acids (see Section 6.9).

Table 8

First-level (to be performed in each patient) and second-level (to be performed in selected patients following specialist evaluation to identify specific aetiologies) laboratory tests, grouped by cardiomyopathy phenotype

LevelHCMDCMNDLVCARVCRCM
First
  • CK

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Troponin

  • Calcium

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Serum iron

  • Thyroid function

  • Troponin

  • Vitamin D (children)

  • Calcium

  • CK

  • C-reactive protein

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Troponin

  • C-reactive protein

  • Liver function

  • NT-proBNPa

  • Renal function

  • Troponin

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Serum angiotensin-converting enzyme

  • Serum iron

  • Troponin

  • Urine and plasma protein immunofixation, free light chains

Second
  • Alpha-galactosidase A levels (males) and lyso-Gb3

  • Carnitine profile

  • Free fatty acids

  • Immunofixation and free light chains

  • Lactic acid

  • Myoglobinuria

  • Pyruvate

  • PTH

  • Urine and plasma protein

  • Urine organic acids and plasma amino acids

  • Carnitine profile

  • Free fatty acids

  • Lactic acid

  • Organ- and non–organ-specific serum autoantibodies

  • Serum angiotensin-converting enzyme

  • Thiamine

  • Viral serology

  • Urine organic acids and plasma amino acids

  • Organ- and non-organ-specific serum autoantibodies

  • Viral serology

  • Organ- and non–organ-specific autoantibodies

  • Serum angiotensin-converting enzyme

LevelHCMDCMNDLVCARVCRCM
First
  • CK

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Troponin

  • Calcium

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Serum iron

  • Thyroid function

  • Troponin

  • Vitamin D (children)

  • Calcium

  • CK

  • C-reactive protein

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Troponin

  • C-reactive protein

  • Liver function

  • NT-proBNPa

  • Renal function

  • Troponin

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Serum angiotensin-converting enzyme

  • Serum iron

  • Troponin

  • Urine and plasma protein immunofixation, free light chains

Second
  • Alpha-galactosidase A levels (males) and lyso-Gb3

  • Carnitine profile

  • Free fatty acids

  • Immunofixation and free light chains

  • Lactic acid

  • Myoglobinuria

  • Pyruvate

  • PTH

  • Urine and plasma protein

  • Urine organic acids and plasma amino acids

  • Carnitine profile

  • Free fatty acids

  • Lactic acid

  • Organ- and non–organ-specific serum autoantibodies

  • Serum angiotensin-converting enzyme

  • Thiamine

  • Viral serology

  • Urine organic acids and plasma amino acids

  • Organ- and non-organ-specific serum autoantibodies

  • Viral serology

  • Organ- and non–organ-specific autoantibodies

  • Serum angiotensin-converting enzyme

ARVC, arrhythmogenic right ventricular cardiomyopathy; BNP, brain natriuretic peptide; CK, creatinine kinase; DCM, dilated cardiomyopathy; Gb3, globotriaosylceramide; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; NT-proBNP, N-terminal pro-brain natriuretic peptide; PTH, parathyroid hormone; RCM, restrictive cardiomyopathy.

Alternatively, BNP can be considered depending on the local availability.

Table 8

First-level (to be performed in each patient) and second-level (to be performed in selected patients following specialist evaluation to identify specific aetiologies) laboratory tests, grouped by cardiomyopathy phenotype

LevelHCMDCMNDLVCARVCRCM
First
  • CK

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Troponin

  • Calcium

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Serum iron

  • Thyroid function

  • Troponin

  • Vitamin D (children)

  • Calcium

  • CK

  • C-reactive protein

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Troponin

  • C-reactive protein

  • Liver function

  • NT-proBNPa

  • Renal function

  • Troponin

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Serum angiotensin-converting enzyme

  • Serum iron

  • Troponin

  • Urine and plasma protein immunofixation, free light chains

Second
  • Alpha-galactosidase A levels (males) and lyso-Gb3

  • Carnitine profile

  • Free fatty acids

  • Immunofixation and free light chains

  • Lactic acid

  • Myoglobinuria

  • Pyruvate

  • PTH

  • Urine and plasma protein

  • Urine organic acids and plasma amino acids

  • Carnitine profile

  • Free fatty acids

  • Lactic acid

  • Organ- and non–organ-specific serum autoantibodies

  • Serum angiotensin-converting enzyme

  • Thiamine

  • Viral serology

  • Urine organic acids and plasma amino acids

  • Organ- and non-organ-specific serum autoantibodies

  • Viral serology

  • Organ- and non–organ-specific autoantibodies

  • Serum angiotensin-converting enzyme

LevelHCMDCMNDLVCARVCRCM
First
  • CK

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Troponin

  • Calcium

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Serum iron

  • Thyroid function

  • Troponin

  • Vitamin D (children)

  • Calcium

  • CK

  • C-reactive protein

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Phosphate

  • Proteinuria

  • Renal function

  • Troponin

  • C-reactive protein

  • Liver function

  • NT-proBNPa

  • Renal function

  • Troponin

  • CK

  • Ferritin

  • Full blood count

  • Liver function

  • NT-proBNPa

  • Proteinuria

  • Renal function

  • Serum angiotensin-converting enzyme

  • Serum iron

  • Troponin

  • Urine and plasma protein immunofixation, free light chains

Second
  • Alpha-galactosidase A levels (males) and lyso-Gb3

  • Carnitine profile

  • Free fatty acids

  • Immunofixation and free light chains

  • Lactic acid

  • Myoglobinuria

  • Pyruvate

  • PTH

  • Urine and plasma protein

  • Urine organic acids and plasma amino acids

  • Carnitine profile

  • Free fatty acids

  • Lactic acid

  • Organ- and non–organ-specific serum autoantibodies

  • Serum angiotensin-converting enzyme

  • Thiamine

  • Viral serology

  • Urine organic acids and plasma amino acids

  • Organ- and non-organ-specific serum autoantibodies

  • Viral serology

  • Organ- and non–organ-specific autoantibodies

  • Serum angiotensin-converting enzyme

ARVC, arrhythmogenic right ventricular cardiomyopathy; BNP, brain natriuretic peptide; CK, creatinine kinase; DCM, dilated cardiomyopathy; Gb3, globotriaosylceramide; HCM, hypertrophic cardiomyopathy; NDLVC, non-dilated left ventricular cardiomyopathy; NT-proBNP, N-terminal pro-brain natriuretic peptide; PTH, parathyroid hormone; RCM, restrictive cardiomyopathy.

Alternatively, BNP can be considered depending on the local availability.

Recommendation Table 3

Recommendations for laboratory tests in the diagnosis of cardiomyopathies

graphic
graphic
Recommendation Table 3

Recommendations for laboratory tests in the diagnosis of cardiomyopathies

graphic
graphic

6.7. Multimodality imaging

6.7.1. General considerations

Non-invasive imaging modalities represent the backbone of diagnosis and follow-up in patients with cardiomyopathies, including ultrasound-based techniques, CMR imaging, computed tomography (CT), and nuclear techniques, such as positron emission tomography (PET) and scintigraphy (Figure 6).1,71,72 Physicians should always consider the yield of actionable results vs. the costs, advantages, and limitations of each technique, as well as patient safety and patient exposure to ionizing radiation and contrast media. Standardized algorithms should be in place to move hierarchically from simpler and cheaper to more complex and expensive tests. A bi-directional flow of information between the clinician and the imager is key to maximizing appropriateness: clinicians should formulate and share clear pre-test hypotheses, based on available information, to aid the interpretation of novel findings. The imager should respond in a similarly focused fashion, assessing the likelihood of alternative diagnoses and refraining from diagnoses that are not compatible/plausible based on the overall clinical context.

Multimodality imaging process in cardiomyopathies.
Figure 6

Multimodality imaging process in cardiomyopathies.

CMR, cardiac magnetic resonance; CTCA, computed tomography coronary angiography; LGE, late gadolinium enhancement; LV, left ventricular; PET, positron emission tomography.

6.7.2. Echocardiography

The non-invasive nature and widespread availability of echocardiography make it the main imaging tool, from initial diagnosis to follow-up. Transthoracic echocardiography (TTE) provides relevant information on global and regional RV and LV anatomy and function as well as valve function and the presence of dynamic obstruction, pulmonary hypertension, or pericardial effusions.71–73 Myocardial deformation imaging (speckle tracking or tissue Doppler) with global longitudinal strain is a more sensitive marker than EF to detect subtle ventricular dysfunction (e.g. in genotype-positive HCM, DCM, and ARVC family members72,74,75), and may help discriminate between different aetiologies of hypertrophy76 (e.g. amyloidosis, HCM, and athlete’s heart). Mechanical dispersion is a marker of contraction inhomogeneity and highlights fine structural changes that may be missed by other modalities.77–80 Three-dimensional echocardiography reliably assesses volumes of cardiac chambers but needs an adequate acoustic window. Contrast agents can be considered for better endocardial delineation to depict the presence of hypertrabeculation, apical HCM, or apical aneurysms, and to exclude thrombus. Stress echocardiography can be helpful in selected patients to evaluate myocardial ischaemia and exercise echocardiography is useful to identify provocable LVOTO in symptomatic patients with HCM (see Section 7.1.1.3). Transoesophageal echocardiography is limited to selected indications, such as the exclusion of atrial thrombi related to AF, elucidating the mechanism of mitral regurgitation, or in planning invasive interventions (e.g. septal myectomy in HCM).

When measuring cardiac dimensions and wall thickness in children, it is important to correct for body size, using z-scores (defined as the number of standard deviations from the population mean). Of note, there are inherent limitations with the use of z-scores in the diagnosis of cardiomyopathies, including the fact that there are many different normative data published resulting in significant variation in z-scores for the same patient.81 In addition, there are no normative data for wall thickness other than at the basal interventricular septum or posterior wall. The Task Force recommends using the normative data from the Paediatric Heart Network consortium.82

Recommendation Table 4

Recommendation for echocardiographic evaluation in patients with cardiomyopathy

graphic
graphic
Recommendation Table 4

Recommendation for echocardiographic evaluation in patients with cardiomyopathy

graphic
graphic

6.7.3. Cardiac magnetic resonance

Cardiac magnetic resonance imaging (MRI) combines the advantages of non-invasiveness and independence of acoustic window with the ability for tissue characterization. The latter advantage is particularly important in the diagnosis of NDLVC, ARVC, myocarditis, amyloidosis, sarcoidosis and other forms of inflammatory disease, and iron overload/haemochromatosis. Cardiac magnetic resonance is particularly useful if echocardiography provides poor image quality. Initial evaluation should routinely include cine imaging sequences, T2-weighted sequences, pre- and post-contrast T1 mapping, and late gadolinium enhancement (LGE). When suspecting haemochromatosis, T2* mapping should be employed. Cardiac magnetic resonance findings can provide important aetiological clues (Figure 7), with potential therapeutic implications (Table 9) and should be assessed collectively with genetic results and other clinical features by experienced operators in cardiac imaging and the evaluation of heart muscle disease. Serial follow-up CMR, every 2–5 years depending on initial severity and clinical course, can assist in evaluating disease progression as well as the benefits of therapy (e.g. evaluation of extracellular volume [ECV] in amyloidosis, or of iron deposition in haemochromatosis), and should be considered in all patients with cardiomyopathy.

Examples of cardiac magnetic resonance imaging tissue characterization features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype.
Figure 7

Examples of cardiac magnetic resonance imaging tissue characterization features that should raise the suspicion of specific aetiologies, grouped according to cardiomyopathy phenotype.

ARVC, arrhythmogenic right ventricular cardiomyopathy; CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; DES, desmin; DSP, desmoplakin; EMF, endomyocardial fibrosis; FLNC, filamin C; HCM, hypertrophic cardiomyopathy; LGE, late gadolinium enhancement; LV, left ventricular; LVH, left ventricular hypertrophy; NDLVC, non-dilated left ventricular cardiomyopathy; RCM, restrictive cardiomyopathy; RV, right ventricular. Examples of CMR tissue characterization features that should raise the suspicion of specific aetiologies (column 4), grouped according to cardiomyopathy phenotype (column 1). CMR images features (column 3) correspond to the listed findings (column 2).

Table 9

Frequently encountered actionable results on multimodality imaging

Parameter/findingAction
RWMAs on echocardiography or CMRRaise suspicion of concomitant CAD, myocarditis, ARVC, NDLVC, or sarcoidosis
Systolic impairment on echocardiography or CMRAssessment of risk in DCM, NDLVC, and ARVC; evaluation of treatment efficacy
Measurement of the wall thickness on echocardiography or CMRDiagnosis of HCM (when echocardiography is inconclusive); risk stratification in HCM
Diastolic dysfunction on echocardiographyExplain symptoms; evaluation of treatment efficacy
Left atrial size on echocardiographySCD risk prediction in HCM; systematic screening for AF in case of left atrial enlargement
LVOTO in HCM on resting/exercise echocardiographyExplain symptoms; guide management
Non-invasive evaluation of pulmonary pressuresExplain symptoms; guide management
Tissue characterization on CMRDiagnosis; risk assessment
Inflammation on CMR or 18F-FDG-PETDiagnosis; evaluation of treatment efficacy in inflammatory cardiomyopathies
Parameter/findingAction
RWMAs on echocardiography or CMRRaise suspicion of concomitant CAD, myocarditis, ARVC, NDLVC, or sarcoidosis
Systolic impairment on echocardiography or CMRAssessment of risk in DCM, NDLVC, and ARVC; evaluation of treatment efficacy
Measurement of the wall thickness on echocardiography or CMRDiagnosis of HCM (when echocardiography is inconclusive); risk stratification in HCM
Diastolic dysfunction on echocardiographyExplain symptoms; evaluation of treatment efficacy
Left atrial size on echocardiographySCD risk prediction in HCM; systematic screening for AF in case of left atrial enlargement
LVOTO in HCM on resting/exercise echocardiographyExplain symptoms; guide management
Non-invasive evaluation of pulmonary pressuresExplain symptoms; guide management
Tissue characterization on CMRDiagnosis; risk assessment
Inflammation on CMR or 18F-FDG-PETDiagnosis; evaluation of treatment efficacy in inflammatory cardiomyopathies

18F-FDG-PET, fluorodeoxyglucose positron emission tomography; AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; AF, atrial fibrillation; CAD, coronary artery disease; CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVOTO, left ventricular outflow tract obstruction; NDLVC, non-dilated left ventricular cardiomyopathy; RWMA, regional wall motion abnormality; SCD, sudden cardiac death.

Table 9

Frequently encountered actionable results on multimodality imaging

Parameter/findingAction
RWMAs on echocardiography or CMRRaise suspicion of concomitant CAD, myocarditis, ARVC, NDLVC, or sarcoidosis
Systolic impairment on echocardiography or CMRAssessment of risk in DCM, NDLVC, and ARVC; evaluation of treatment efficacy
Measurement of the wall thickness on echocardiography or CMRDiagnosis of HCM (when echocardiography is inconclusive); risk stratification in HCM
Diastolic dysfunction on echocardiographyExplain symptoms; evaluation of treatment efficacy
Left atrial size on echocardiographySCD risk prediction in HCM; systematic screening for AF in case of left atrial enlargement
LVOTO in HCM on resting/exercise echocardiographyExplain symptoms; guide management
Non-invasive evaluation of pulmonary pressuresExplain symptoms; guide management
Tissue characterization on CMRDiagnosis; risk assessment
Inflammation on CMR or 18F-FDG-PETDiagnosis; evaluation of treatment efficacy in inflammatory cardiomyopathies
Parameter/findingAction
RWMAs on echocardiography or CMRRaise suspicion of concomitant CAD, myocarditis, ARVC, NDLVC, or sarcoidosis
Systolic impairment on echocardiography or CMRAssessment of risk in DCM, NDLVC, and ARVC; evaluation of treatment efficacy
Measurement of the wall thickness on echocardiography or CMRDiagnosis of HCM (when echocardiography is inconclusive); risk stratification in HCM
Diastolic dysfunction on echocardiographyExplain symptoms; evaluation of treatment efficacy
Left atrial size on echocardiographySCD risk prediction in HCM; systematic screening for AF in case of left atrial enlargement
LVOTO in HCM on resting/exercise echocardiographyExplain symptoms; guide management
Non-invasive evaluation of pulmonary pressuresExplain symptoms; guide management
Tissue characterization on CMRDiagnosis; risk assessment
Inflammation on CMR or 18F-FDG-PETDiagnosis; evaluation of treatment efficacy in inflammatory cardiomyopathies

18F-FDG-PET, fluorodeoxyglucose positron emission tomography; AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; AF, atrial fibrillation; CAD, coronary artery disease; CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVOTO, left ventricular outflow tract obstruction; NDLVC, non-dilated left ventricular cardiomyopathy; RWMA, regional wall motion abnormality; SCD, sudden cardiac death.

6.7.3.1. Special considerations
  • Recently developed rapid CMR techniques allow scans to be performed without general anaesthesia even in very young children.103 In children (and adults) unable to undergo CMR without general anaesthesia, the relative risks and benefits of the procedure should be considered.

  • Imaging artefacts caused by cardiac implantable electronic devices have posed limitations for CMR imaging in the past.104–110 A number of solutions are available to reduce artefacts, including reducing inhomogeneity, technical adjustments, and the use of special sequences, which reduce the rate of uninterpretable studies to one in five.111,112 Cardiac magnetic resonance can therefore be considered in patients with conditional devices and nearly all non-conditional devices provided appropriate protocols are put in place.113

  • Nephrogenic systemic fibrosis is a rare complication reported in patients with first-generation linear unstable gadolinium chelates and severe renal disease.114 However, gadolinium-based contrast agents can be safely administered for patients with an estimated glomerular filtration rate >30 mL/min/1.73 m2, and nephrogenic systemic fibrosis is virtually unreported with the use of newer linear or macrocyclic gadolinium contrasts. For patients with severe renal impairment, new CMR modalities and mapping procedures, which are very informative and do not require the use of contrast, are particularly valuable when assessing Anderson–Fabry disease and cardiac amyloidosis.115–117

  • The use of gadolinium contrast is generally not advised in pregnancy due to the potential for adverse outcomes in the foetus and neonate.118

Recommendation Table 5

Recommendations for cardiac magnetic resonance indication in patients with cardiomyopathy

graphic
graphic
graphic
graphic
Recommendation Table 5

Recommendations for cardiac magnetic resonance indication in patients with cardiomyopathy

graphic
graphic
graphic
graphic

6.7.4. Computed tomography and nuclear medicine techniques

Other imaging modalities, including nuclear medicine-based techniques and CT, are indicated in selected subsets of patients with cardiomyopathy.160,161 Indications and the risk-benefit ratio should be evaluated on an individual patient basis, always taking into account radioprotection issues, which are particularly relevant in the young. Nuclear medicine is particularly helpful in the aetiological diagnosis of cardiac amyloidosis (see Section 7.7). 18FDG-PET is useful in the identification of myocardial inflammation associated with active sarcoidosis and, potentially, in other atypical forms of myocarditis.162–164 However, a negative scan does not exclude sarcoidosis in its inactive form. In patients with HCM, DCM, and Anderson–Fabry disease, H215O or 13NH3 dipyridamole or regadenoson PET has been used to evaluate microvascular dysfunction, an important predictor of adverse outcome.165 However, this test does not currently have a role in aetiological diagnosis (e.g. in distinguishing phenocopies) and is largely confined to research purposes.

Computed tomography-based imaging is primarily used in patients with a suspicion of cardiomyopathy to rule out CAD, either as an alternative diagnosis (e.g. in individuals with DCM, NDLVC, or ARVC phenotypes) or as a comorbidity affecting clinical manifestations and course. In children and adolescents, CT angiography can be useful to exclude congenital vascular malformations (e.g. anomalous left coronary artery from the pulmonary artery [ALCAPA] or anomalous pulmonary venous return). Standard CT imaging provides additional information regarding concomitant pulmonary disease (e.g. sarcoidosis), pericardial disease, and chest wall deformities affecting the heart.

Recommendation Table 6

Recommendations for computed tomography and nuclear imaging

graphic
graphic
Recommendation Table 6

Recommendations for computed tomography and nuclear imaging

graphic
graphic

6.7.5. Endomyocardial biopsy

Endomyocardial biopsy (EMB) with immunohistochemical quantification of inflammatory cells and identification of viral genomes remains the gold standard for the identification of cardiac inflammation. It may confirm the diagnosis of autoimmune disease in patients with unexplained heart failure and suspected giant cell myocarditis, eosinophilic myocarditis, vasculitis, and sarcoidosis. Electron microscopy should be employed when storage or mitochondrial cardiomyopathies are suspected. Endomyocardial biopsy should be reserved for specific situations where its results may affect treatment after careful evaluation of the risk-benefit ratio. Importantly, EMB is not completely risk-free and should be performed by experienced teams. Likewise, the diagnostic work-up of a biopsy should be performed by pathologists with expertise in cardiomyopathies.

Recommendation Table 7

Recommendation for endomyocardial biopsy in patients with cardiomyopathy

graphic
graphic
Recommendation Table 7

Recommendation for endomyocardial biopsy in patients with cardiomyopathy

graphic
graphic

6.8. Genetic testing and counselling

6.8.1. Genetic architecture

Familial forms of cardiomyopathies show diverse modes of inheritance. Gene identification has, over the last three decades, primarily focused on the identification of Mendelian (monogenic) disease genes that most commonly display autosomal dominant inheritance, although other inheritance patterns including autosomal recessive, X-linked, and mitochondrial (matrilineal) are also observed (Table 5). Major genes currently associated with different types of cardiomyopathies are listed in Table 10. Cardiomyopathies are characterized by a marked genetic and allelic heterogeneity, that is, many different variants in many different genes can cause the same phenotype. Rare pathogenic variants associated with cardiomyopathies often exhibit the phenomena of incomplete and age-related penetrance, and variable expressivity.178,179 That is, not all individuals carrying a causative variant manifest the disease and, among those who do, there is broad variability in age of onset and disease severity. Thus, while some individuals may have severe disease necessitating cardiac transplantation at a young age, others may remain unaffected throughout their lives or are only mildly affected. This variability could be due to heterogeneity among causative variants, the additional contribution of non-genetic (clinical, environmental) factors (e.g. hypertension in HCM,180 exercise in ARVC181), and the co-inheritance of additional genetic factors, which act to exacerbate or attenuate the effect of the principal Mendelian genetic variant on the phenotype. This is an active area of research, and recent genome-wide association studies conducted in patients with HCM have provided strong evidence for the modulatory role of common genetic variants of individually small effect that collectively modulate the effects of Mendelian variants (Figure 8).182,183

The genetic architecture of the cardiomyopathies.
Figure 8

The genetic architecture of the cardiomyopathies.

GWAS, genome-wide association studies; MAF, minor allele frequency. Cardiomyopathy can be Mendelian, caused by genetic variants that are ultra-rare in the general population and have large effect sizes. Such variants can display complete penetrance; i.e. all individuals with the variant in the family manifest the disease (panel A). However, individual variants are often insufficient to yield a disease phenotype in isolation, and their effect is modulated by the co-inheritance of modulatory genetic factors and by non-genetic factors (panel B). Besides increasing disease penetrance, such modulatory variants also affect the severity of the disease (panel B). Modulatory genetic factors are thought to comprise common variants with individually small-effect sizes and intermediate-effect variants that have population frequencies and effect sizes between rare and common variants. Some patients have a more complex aetiology (non-Mendelian/polygenic inheritance) in which a substantial number of non-Mendelian genetic factors and non-genetic factors are required to reach the threshold for disease (panel C). Such patients typically have a sporadic presentation or present with a less pronounced familial clustering of the disease. Family trees demonstrate the male (square) and female (circle) family members that are affected (black filled), with incomplete phenotype (grey filled) or unaffected (white filled). The presence or absence of the variant of interest is noted with “+” or “-”, respectively.

Table 10

Overview of genes associated with monogenic, non-syndromic cardiomyopathies, and their relative contributions to different cardiomyopathic phenotypes

graphic
graphic
graphic
graphic
graphic
graphic
Table 10

Overview of genes associated with monogenic, non-syndromic cardiomyopathies, and their relative contributions to different cardiomyopathic phenotypes

graphic
graphic
graphic
graphic
graphic
graphic

Across the different cardiomyopathies, the proportion of cases with a confident genetic diagnosis (that is with identification of a likely causal Mendelian genetic variant) is relatively low (e.g. as low as ∼40% in HCM124 and ∼30% in DCM184–186). Genome-wide association studies of common variants in HCM and DCM have provided empirical evidence for substantial polygenic inheritance in these cardiomyopathies.182,183,187 Contrary to Mendelian inheritance, where a single, large-effect variant primarily determines susceptibility to the disorder, complex inheritance rests on the co-inheritance of multiple susceptibility variants. Although not yet studied systematically, besides common variants of small effect, intermediate-effect variants with effect sizes and frequencies between common and Mendelian variants are also expected to contribute to such complex inheritance.188 It is likely that cardiomyopathies span a continuum of genetic complexity, with Mendelian forms at one end, determined primarily by the inheritance of an ultra-rare large-effect genetic variant, and highly polygenic forms at the other (see Figure 8). Variants that contribute to disease susceptibility in the setting of complex inheritance likely overlap with those that modulate disease penetrance and expressivity in the Mendelian form of the disease.182,183

6.8.2. Genetic testing

Genetic testing of Mendelian cardiomyopathy genes has become a standard aspect of clinical management in affected families.3 First-line testing should be focused on genes robustly associated with the presenting phenotype. If initial testing does not reveal a cause, but suspicion of a monogenic cause remains high, then more extended sequencing or analysis may be indicated, depending on the family structure and other factors. Once a genetic cause is established in one family member, then other family members may undergo testing for only the causative variant.

Genetic testing in an individual with cardiomyopathy (known as confirmatory testing or diagnostic testing) is recommended for their direct benefit: (i) to confirm the diagnosis; (ii) where it may inform prognosis; (iii) where it may inform treatment selection; or (iv) where it may inform their reproductive management. Genetic testing of an affected individual may be indicated, even if it is unlikely to alter their management, if there are relatives who may benefit from testing, particularly if there are relatives who will be enrolled in longitudinal surveillance if the genetic aetiology is not established and who may be spared this burden if a genetic diagnosis is made in the family (Table 11). Testing may also be helpful in broader contexts, even when not obviously informative for immediate management; for example, a genetic diagnosis may provide psychological benefit in a patient struggling to understand their disease.

Table 11

Utility of genetic testing in cardiomyopathies

For the patient
  • Diagnosis: for the affected individual, the diagnosis of cardiomyopathy is primarily made on the basis of a phenotypic definition of disease, without reference to genetic aetiology. However, with appropriate genetic counselling and acknowledging the caveat that the finding will only be clinically actionable when a P/LP variant is found, genetic testing may be of value in clarifying borderline cases (e.g. where LVH is observed in the context of mild or controlled hypertension, but the clinician is not able to confidently distinguish between early sarcomeric HCM and a hypertensive phenocopy). Genetic testing can also identify genocopies: distinct genetic conditions that mimic a particular cardiomyopathy.

  • Prognosis: for an increasing number of conditions, a genetic diagnosis can provide prognostic information. For example, DCM due to variants in LMNA has an adverse prognosis requiring more frequent surveillance and shifting therapeutic decision thresholds with a lower threshold for primary prevention ICD implantation.

  • Therapy: a genetic diagnosis may directly stratify choice of therapy. In addition to decisions on primary prevention ICD implantation, an increasing number of treatments are either established or under trial for a specific molecular subtype of cardiomyopathy. In addition, with an increasingly sophisticated toolbox for manipulation of the genome, further waves of therapies aiming to replace, alter, or remove abnormal genes and transcripts responsible for cardiomyopathies are anticipated once a precise molecular aetiology is established in a patient.

  • Reproductive advice: a genetic diagnosis informs reproductive advice and management for an affected adult and/or the parents of an affected child, enabling tailored advice on inheritance patterns and the risk of transmission to future children, and opening the door to management of risk; e.g. through pre-natal diagnostics or pre-implantation genetic diagnosis.

For relatives
  • Cardiomyopathies display incomplete and age-related penetrance, with great variability, therefore it is very difficult to identify clinically those relatives who are not at risk of developing cardiomyopathy. A normal one-off assessment is of limited value, and relatives without cardiomyopathy on initial evaluation may require long-term longitudinal surveillance. Genetic testing can eliminate this uncertainty: an individual who does not carry the genetic variant proved to be responsible for disease in their family can be confidently reassured and discharged without surveillance, while an individual who carries a disease-causing variant can be followed closely, and potentially treated early.

For the patient
  • Diagnosis: for the affected individual, the diagnosis of cardiomyopathy is primarily made on the basis of a phenotypic definition of disease, without reference to genetic aetiology. However, with appropriate genetic counselling and acknowledging the caveat that the finding will only be clinically actionable when a P/LP variant is found, genetic testing may be of value in clarifying borderline cases (e.g. where LVH is observed in the context of mild or controlled hypertension, but the clinician is not able to confidently distinguish between early sarcomeric HCM and a hypertensive phenocopy). Genetic testing can also identify genocopies: distinct genetic conditions that mimic a particular cardiomyopathy.

  • Prognosis: for an increasing number of conditions, a genetic diagnosis can provide prognostic information. For example, DCM due to variants in LMNA has an adverse prognosis requiring more frequent surveillance and shifting therapeutic decision thresholds with a lower threshold for primary prevention ICD implantation.

  • Therapy: a genetic diagnosis may directly stratify choice of therapy. In addition to decisions on primary prevention ICD implantation, an increasing number of treatments are either established or under trial for a specific molecular subtype of cardiomyopathy. In addition, with an increasingly sophisticated toolbox for manipulation of the genome, further waves of therapies aiming to replace, alter, or remove abnormal genes and transcripts responsible for cardiomyopathies are anticipated once a precise molecular aetiology is established in a patient.

  • Reproductive advice: a genetic diagnosis informs reproductive advice and management for an affected adult and/or the parents of an affected child, enabling tailored advice on inheritance patterns and the risk of transmission to future children, and opening the door to management of risk; e.g. through pre-natal diagnostics or pre-implantation genetic diagnosis.

For relatives
  • Cardiomyopathies display incomplete and age-related penetrance, with great variability, therefore it is very difficult to identify clinically those relatives who are not at risk of developing cardiomyopathy. A normal one-off assessment is of limited value, and relatives without cardiomyopathy on initial evaluation may require long-term longitudinal surveillance. Genetic testing can eliminate this uncertainty: an individual who does not carry the genetic variant proved to be responsible for disease in their family can be confidently reassured and discharged without surveillance, while an individual who carries a disease-causing variant can be followed closely, and potentially treated early.

DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; ICD, implantable cardioverter defibrillator; LMNA, lamin A/C; LVH, left ventricular hypertrophy; P/LP, pathogenic/likely pathogenic.

Table 11

Utility of genetic testing in cardiomyopathies

For the patient
  • Diagnosis: for the affected individual, the diagnosis of cardiomyopathy is primarily made on the basis of a phenotypic definition of disease, without reference to genetic aetiology. However, with appropriate genetic counselling and acknowledging the caveat that the finding will only be clinically actionable when a P/LP variant is found, genetic testing may be of value in clarifying borderline cases (e.g. where LVH is observed in the context of mild or controlled hypertension, but the clinician is not able to confidently distinguish between early sarcomeric HCM and a hypertensive phenocopy). Genetic testing can also identify genocopies: distinct genetic conditions that mimic a particular cardiomyopathy.

  • Prognosis: for an increasing number of conditions, a genetic diagnosis can provide prognostic information. For example, DCM due to variants in LMNA has an adverse prognosis requiring more frequent surveillance and shifting therapeutic decision thresholds with a lower threshold for primary prevention ICD implantation.

  • Therapy: a genetic diagnosis may directly stratify choice of therapy. In addition to decisions on primary prevention ICD implantation, an increasing number of treatments are either established or under trial for a specific molecular subtype of cardiomyopathy. In addition, with an increasingly sophisticated toolbox for manipulation of the genome, further waves of therapies aiming to replace, alter, or remove abnormal genes and transcripts responsible for cardiomyopathies are anticipated once a precise molecular aetiology is established in a patient.

  • Reproductive advice: a genetic diagnosis informs reproductive advice and management for an affected adult and/or the parents of an affected child, enabling tailored advice on inheritance patterns and the risk of transmission to future children, and opening the door to management of risk; e.g. through pre-natal diagnostics or pre-implantation genetic diagnosis.

For relatives
  • Cardiomyopathies display incomplete and age-related penetrance, with great variability, therefore it is very difficult to identify clinically those relatives who are not at risk of developing cardiomyopathy. A normal one-off assessment is of limited value, and relatives without cardiomyopathy on initial evaluation may require long-term longitudinal surveillance. Genetic testing can eliminate this uncertainty: an individual who does not carry the genetic variant proved to be responsible for disease in their family can be confidently reassured and discharged without surveillance, while an individual who carries a disease-causing variant can be followed closely, and potentially treated early.

For the patient
  • Diagnosis: for the affected individual, the diagnosis of cardiomyopathy is primarily made on the basis of a phenotypic definition of disease, without reference to genetic aetiology. However, with appropriate genetic counselling and acknowledging the caveat that the finding will only be clinically actionable when a P/LP variant is found, genetic testing may be of value in clarifying borderline cases (e.g. where LVH is observed in the context of mild or controlled hypertension, but the clinician is not able to confidently distinguish between early sarcomeric HCM and a hypertensive phenocopy). Genetic testing can also identify genocopies: distinct genetic conditions that mimic a particular cardiomyopathy.

  • Prognosis: for an increasing number of conditions, a genetic diagnosis can provide prognostic information. For example, DCM due to variants in LMNA has an adverse prognosis requiring more frequent surveillance and shifting therapeutic decision thresholds with a lower threshold for primary prevention ICD implantation.

  • Therapy: a genetic diagnosis may directly stratify choice of therapy. In addition to decisions on primary prevention ICD implantation, an increasing number of treatments are either established or under trial for a specific molecular subtype of cardiomyopathy. In addition, with an increasingly sophisticated toolbox for manipulation of the genome, further waves of therapies aiming to replace, alter, or remove abnormal genes and transcripts responsible for cardiomyopathies are anticipated once a precise molecular aetiology is established in a patient.

  • Reproductive advice: a genetic diagnosis informs reproductive advice and management for an affected adult and/or the parents of an affected child, enabling tailored advice on inheritance patterns and the risk of transmission to future children, and opening the door to management of risk; e.g. through pre-natal diagnostics or pre-implantation genetic diagnosis.

For relatives
  • Cardiomyopathies display incomplete and age-related penetrance, with great variability, therefore it is very difficult to identify clinically those relatives who are not at risk of developing cardiomyopathy. A normal one-off assessment is of limited value, and relatives without cardiomyopathy on initial evaluation may require long-term longitudinal surveillance. Genetic testing can eliminate this uncertainty: an individual who does not carry the genetic variant proved to be responsible for disease in their family can be confidently reassured and discharged without surveillance, while an individual who carries a disease-causing variant can be followed closely, and potentially treated early.

DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; ICD, implantable cardioverter defibrillator; LMNA, lamin A/C; LVH, left ventricular hypertrophy; P/LP, pathogenic/likely pathogenic.

Genetic testing in a clinically unaffected relative of an individual with cardiomyopathy may be indicated irrespective of age, even in very young children, if a genetic diagnosis has been established with confidence in the affected individual (known as cascade testing, predictive testing, or pre-symptomatic testing). Once a pathogenic/likely pathogenic (P/LP) variant has been identified within an index patient following investigations of relevant disease genes associated with the specific phenotype, it is possible to offer cascade genetic testing of first-degree at-risk relatives, including pre-test genetic counselling (see Section 6.8.3). In a scenario where a first-degree relative has died, evaluation of close relatives of the deceased individual (i.e. second-degree relatives of the index patient) should also be considered.

Individuals who are found not to harbour the familial variant can usually be discharged from clinical follow-up; those who do carry the familial variant are recommended to undergo clinical evaluation and usually ongoing surveillance. Cascade testing is not indicated when a variant of uncertain significance is identified in the proband.

Sequencing may also be indicated for segregation analysis (rather than as a diagnostic test) to inform interpretation of a variant of uncertain significance found in an affected individual. This is usually limited to individuals who are clearly affected, or to testing of the parents to identify a de novo variant. Genetic counselling in this circumstance would involve clear communication to family members that this is not a diagnostic test, but rather is contributing to clarifying the pathogenicity of the uncertain variant.

Finally, the evaluation of cardiac genes for secondary findings where data are generated in the setting of genetic testing for another clinical indication (also referred to as opportunistic screening) may be reasonable where the balance of benefits and harm is known, and if the cost is acceptable. Broader population screening might also prove reasonable if the balance of benefits and harm proves favourable. At present there is insufficient data to evaluate the balance of benefits and harm in either context, and this should currently only be performed in a research context in order to obtain such data. Careful genetic counselling to fully explain benefits and risks in this setting is critical. At present, there are very little data to evaluate this balance and this is an important evidence gap. In the United States of America, the American College of Medical Genetics and Genomics has recommended that cardiomyopathy-associated genes be evaluated for secondary findings whenever broad clinical sequencing is undertaken, regardless of the initial indication for testing.192,193 There is currently no international consensus around this recommendation.

6.8.2.1. Non-Mendelian cardiomyopathies and implications for genetic testing

The preceding discussion has focused on genetic testing to identify monogenic forms of cardiomyopathy. The recognition that an important proportion of cardiomyopathies have a more complex genetic architecture has important implications for the use of genetic tests.

The absence of a monogenic disease-causing variant on conventional genetic testing (i.e. sequencing for rare variants of large effect) leaves three possibilities: (i) either there is a monogenic cause that has not been identified (i.e. not detected or recognized as causative by current testing); (ii) the cardiomyopathy does not have a genetic aetiology; or (iii) the cardiomyopathy is attributable to the effects of multiple variants of individually smaller effect (Figure 8). Recent data suggest that for many cardiomyopathies, the absence of a rare causative variant on comprehensive testing indicates that the disease is unlikely to have a monogenic aetiology.182,183,194 This, in turn, implies a different inheritance pattern, with a lower risk to first-degree relatives, such that ongoing surveillance may not be indicated if an initial clinical evaluation is reassuring. The use of genetic testing to identify families in whom the disease is unlikely to be monogenic represents a likely new application of conventional testing, which is gathering evidence but not yet established.

Polygenic risk scores (PRS) (sometimes known as genomic risk scores) are another form of genetic test that may, in the future, have relevance in the management of cardiomyopathies. Instead of trying to identify a single genetic variant that is responsible for disease, many variants across the genome are evaluated, each associated with a small effect on disease risk, and a score representing the aggregate risk is calculated.182,183,195–197 To date, the value of a PRS in the clinical management of cardiomyopathies has not yet been demonstrated, and access to genetic counselling will be even more important in conveying risks and uncertainties to patients and families.

6.8.2.2. Genetic test reports and variant interpretation

Many genetic diagnostic laboratories use a standardized framework to interpret and report diagnostic genetic test results.3,198–200 A negative genetic test result in a proband indicates that no causative variant has been found in a known disease-associated gene. This does not necessarily mean the patient does not have a genetic disease, but reflects our limited knowledge of the genetic architecture of inherited cardiomyopathies at this point in time. Aspects concerning the genetic testing approach, genetic testing methods, and variant interpretation are further elaborated in the Supplementary data online, Section 2, and in the European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases.3

6.8.3. Genetic counselling

Genetic counselling is a process that aims to support patients and their families to understand and adapt to the medical, psychosocial, and familial impact of genetic diseases.201,202 It should be performed by healthcare professionals with specific training, such as genetic counsellors, genetic nurses, or clinical/medical geneticists, regardless of whether genetic testing is being considered. Genetic counselling can include a discussion of inheritance risks, provide education including the need for clinical evaluation, perform pre- and post-genetic test counselling, review variant classifications, obtain a three-generation family history, and provide psychosocial support.203–205 For patients with a new diagnosis of cardiomyopathy, there can be difficulty adjusting to life with an inherited cardiomyopathy, challenges living with an implantable cardioverter defibrillator (ICD), and ongoing trauma and grief for those who have experienced a young SCD in their family. Attention to the psychological support needs of patients is therefore critical (see Section 6.12). Indeed, in the general setting, genetic counselling can improve knowledge, recall, and patient empowerment; increase satisfaction with decision-making; and reduce anxiety.206–209

6.8.3.1. Genetic counselling in children

There are specific issues to be considered when counselling children and their families and considering clinical screening and cascade genetic testing,75,210,211 (Table 12) and a patient-centred approach that takes in to account the experiences and values of the family is needed (Figure 9). The guiding principle remains that any testing, clinical or genetic, should be in the best interests of the child and have an impact on management, lifestyle, and/or ongoing clinical testing.75 With appropriate multidisciplinary support in a paediatric setting, psychosocial outcomes in children undergoing clinical screening and cascade genetic testing are no different than those of the general population.212

A patient-centred approach to cascade genetic testing of children.
Figure 9

A patient-centred approach to cascade genetic testing of children.

Factors to consider when supporting families to decide whether to pursue cascade genetic testing in children.

Table 12

Specific issues to consider when counselling children

IssueImplications
AutonomyCompetence of child to decide on testing
Informed consentAppropriate to understanding of child
Right to know/not to know the resultConsider wishes of child and family
ConfidentialityContext of family history
Incomplete and age-related penetranceSymptoms/features of disease may not become apparent for many years
LifestyleSchool, sports, employment
Life stages and transitionMoving from primary to secondary education; transition to adult medical services
IssueImplications
AutonomyCompetence of child to decide on testing
Informed consentAppropriate to understanding of child
Right to know/not to know the resultConsider wishes of child and family
ConfidentialityContext of family history
Incomplete and age-related penetranceSymptoms/features of disease may not become apparent for many years
LifestyleSchool, sports, employment
Life stages and transitionMoving from primary to secondary education; transition to adult medical services
Table 12

Specific issues to consider when counselling children

IssueImplications
AutonomyCompetence of child to decide on testing
Informed consentAppropriate to understanding of child
Right to know/not to know the resultConsider wishes of child and family
ConfidentialityContext of family history
Incomplete and age-related penetranceSymptoms/features of disease may not become apparent for many years
LifestyleSchool, sports, employment
Life stages and transitionMoving from primary to secondary education; transition to adult medical services
IssueImplications
AutonomyCompetence of child to decide on testing
Informed consentAppropriate to understanding of child
Right to know/not to know the resultConsider wishes of child and family
ConfidentialityContext of family history
Incomplete and age-related penetranceSymptoms/features of disease may not become apparent for many years
LifestyleSchool, sports, employment
Life stages and transitionMoving from primary to secondary education; transition to adult medical services
6.8.3.2. Pre- and post-test genetic counselling (proband)

One critical role for genetic counselling is that it should be done alongside genetic testing (see Section 6.8.2).3 This includes a discussion prior to a decision to undertake genetic testing (pre-test), and at the time of the return of the results (post-test). Key discussion points during pre- and post-test counselling are summarized in Table 13.

Table 13

Key discussion points of pre- and post-test genetic counselling

Pre-test genetic counsellingDetailed family history
Genetic education
Process and logistics of genetic testing and return of the result
Explanation of all possible outcomes
Implications for clinical care
Lifestyle implications including sport, exercise, and employment
Implications for the family
Risk of reclassification
Secondary genetic findings
Potential insurance implications (country dependent)
Exploration of feelings and understanding
Psychosocial support
Post-test genetic counsellingRe-cap on key points of pre-test session
Result disclosure
Specific implications for clinical care
Specific implications for the family and how to approach relatives
Risk of reclassification, plan for resolving uncertain variant status if applicable
Exploration of feelings and understanding
Provision of details about how family members can access genetic counselling
Offer information about reproductive genetic testing options for those with a genetic diagnosis
Psychosocial support
Pre-test genetic counsellingDetailed family history
Genetic education
Process and logistics of genetic testing and return of the result
Explanation of all possible outcomes
Implications for clinical care
Lifestyle implications including sport, exercise, and employment
Implications for the family
Risk of reclassification
Secondary genetic findings
Potential insurance implications (country dependent)
Exploration of feelings and understanding
Psychosocial support
Post-test genetic counsellingRe-cap on key points of pre-test session
Result disclosure
Specific implications for clinical care
Specific implications for the family and how to approach relatives
Risk of reclassification, plan for resolving uncertain variant status if applicable
Exploration of feelings and understanding
Provision of details about how family members can access genetic counselling
Offer information about reproductive genetic testing options for those with a genetic diagnosis
Psychosocial support

Modified from Ingles et al.213

Table 13

Key discussion points of pre- and post-test genetic counselling

Pre-test genetic counsellingDetailed family history
Genetic education
Process and logistics of genetic testing and return of the result
Explanation of all possible outcomes
Implications for clinical care
Lifestyle implications including sport, exercise, and employment
Implications for the family
Risk of reclassification
Secondary genetic findings
Potential insurance implications (country dependent)
Exploration of feelings and understanding
Psychosocial support
Post-test genetic counsellingRe-cap on key points of pre-test session
Result disclosure
Specific implications for clinical care
Specific implications for the family and how to approach relatives
Risk of reclassification, plan for resolving uncertain variant status if applicable
Exploration of feelings and understanding
Provision of details about how family members can access genetic counselling
Offer information about reproductive genetic testing options for those with a genetic diagnosis
Psychosocial support
Pre-test genetic counsellingDetailed family history
Genetic education
Process and logistics of genetic testing and return of the result
Explanation of all possible outcomes
Implications for clinical care
Lifestyle implications including sport, exercise, and employment
Implications for the family
Risk of reclassification
Secondary genetic findings
Potential insurance implications (country dependent)
Exploration of feelings and understanding
Psychosocial support
Post-test genetic counsellingRe-cap on key points of pre-test session
Result disclosure
Specific implications for clinical care
Specific implications for the family and how to approach relatives
Risk of reclassification, plan for resolving uncertain variant status if applicable
Exploration of feelings and understanding
Provision of details about how family members can access genetic counselling
Offer information about reproductive genetic testing options for those with a genetic diagnosis
Psychosocial support

Modified from Ingles et al.213

6.8.3.3. Genetic counselling for cascade testing

Once a P/LP variant has been identified within an index patient following investigations of relevant disease genes associated with the specific phenotype, it is possible to offer cascade genetic testing of first-degree at-risk relatives, including pre-test genetic counselling (see Section 6.8). In a scenario where a first-degree relative has died, evaluation of close relatives of the deceased individual (i.e. second-degree relatives of the index patient) should also be considered.

The right assignment of the level of pathogenicity of a variant is crucial for cascade genetic testing. Inappropriate use of genetic testing in a family has the potential to introduce unnecessary worry and fear, as well as potential harm related to the misinterpretation of genetic variants. Variants should therefore be classified by a specialized multidisciplinary cardiac genetic team with an appropriate level of expertise. Systematic reclassification of identified variants and communication to families is crucial. Conveying information on the importance of clinical and genetic testing of at-risk relatives is typically reliant on the proband in the family understanding the information and passing it on to the appropriate relatives. Common barriers to communication can include poor family relationships, guilt regarding passing a causative variant on to children, psychosocial factors including distress, and comprehension of the result.214,215 A patient will often selectively communicate genetic information to relatives, assessing their ability to understand and cope with the information, their life stage, and their risk status.216 Poor health literacy is an important barrier to effectively communicating genetic risk information to relatives, highlighting the need for targeted resources and mechanisms for support.217

6.8.3.4. Pre-natal or pre-implantation genetic diagnosis

Pre-natal or pre-implantation genetic testing can be offered to parents who have had a previous affected child with an inherited cardiomyopathy due to a single or multiple pathogenic variant(s), or to couples where one or both partners carries a known pathogenic (familial) variant. The decision to pursue pre-natal or pre-implantation genetic testing should consider a spectrum of disease- and parent-related aspects, including cultural, religious, legal, and availability issues.218 Options for pre-natal or pre-implantation genetic diagnosis should be discussed as part of the genetic counselling process and in a timely manner. If pre-natal diagnostics are performed, it should be done early enough in pregnancy to give the patient options regarding pregnancy continuation, or co-ordination of pregnancy, delivery, and neonatal care.219

Options for pre-natal and pre-implantation genetic diagnosis are summarized in Table 14. Most reproductive diagnostic testing options are for established pregnancies, except pre-implantation genetic diagnosis which allows for selective implantation of unaffected embryos.

Table 14

Pre-natal and pre-implantation options and implications

IssueImplications
Chorionic villus sampling
  • Transcervical or transabdominal sampling of the placenta at 10–14 weeks of gestation. The procedure-related foetal loss rate is ∼0.2%.220

  • Performed at early gestational age; short testing turnaround time.

Amniocentesis
  • Direct sampling of amniotic fluid is performed after 15 weeks of gestation The loss rate is ∼0.1%.220

Non-invasive pre-natal testing
  • Performed for a single gene disorder.

  • Cell-free foetal DNA isolated from maternal plasma sample.

  • Offered in early pregnancy (approximately week 9); miscarriage risk not increased.

  • Not widely available (method still largely under development and therefore not readily available).

Pre-implantation genetic diagnosis
  • IVF procedure with a success rate of 25–30% per embryo transfer though dependent on the mother’s age and fertility, followed by biopsy and genetic testing of a single cell of the embryo.

  • Risks to mother and offspring of IVF, such as multiple birth, premature labour and low birth weight, as well as emotional health effects for those undergoing the procedure.

  • Availability and methods differ across countries.

IssueImplications
Chorionic villus sampling
  • Transcervical or transabdominal sampling of the placenta at 10–14 weeks of gestation. The procedure-related foetal loss rate is ∼0.2%.220

  • Performed at early gestational age; short testing turnaround time.

Amniocentesis
  • Direct sampling of amniotic fluid is performed after 15 weeks of gestation The loss rate is ∼0.1%.220

Non-invasive pre-natal testing
  • Performed for a single gene disorder.

  • Cell-free foetal DNA isolated from maternal plasma sample.

  • Offered in early pregnancy (approximately week 9); miscarriage risk not increased.

  • Not widely available (method still largely under development and therefore not readily available).

Pre-implantation genetic diagnosis
  • IVF procedure with a success rate of 25–30% per embryo transfer though dependent on the mother’s age and fertility, followed by biopsy and genetic testing of a single cell of the embryo.

  • Risks to mother and offspring of IVF, such as multiple birth, premature labour and low birth weight, as well as emotional health effects for those undergoing the procedure.

  • Availability and methods differ across countries.

IVF, in vitro fertilization.

Table 14

Pre-natal and pre-implantation options and implications

IssueImplications
Chorionic villus sampling
  • Transcervical or transabdominal sampling of the placenta at 10–14 weeks of gestation. The procedure-related foetal loss rate is ∼0.2%.220

  • Performed at early gestational age; short testing turnaround time.

Amniocentesis
  • Direct sampling of amniotic fluid is performed after 15 weeks of gestation The loss rate is ∼0.1%.220

Non-invasive pre-natal testing
  • Performed for a single gene disorder.

  • Cell-free foetal DNA isolated from maternal plasma sample.

  • Offered in early pregnancy (approximately week 9); miscarriage risk not increased.

  • Not widely available (method still largely under development and therefore not readily available).

Pre-implantation genetic diagnosis
  • IVF procedure with a success rate of 25–30% per embryo transfer though dependent on the mother’s age and fertility, followed by biopsy and genetic testing of a single cell of the embryo.

  • Risks to mother and offspring of IVF, such as multiple birth, premature labour and low birth weight, as well as emotional health effects for those undergoing the procedure.

  • Availability and methods differ across countries.

IssueImplications
Chorionic villus sampling
  • Transcervical or transabdominal sampling of the placenta at 10–14 weeks of gestation. The procedure-related foetal loss rate is ∼0.2%.220

  • Performed at early gestational age; short testing turnaround time.

Amniocentesis
  • Direct sampling of amniotic fluid is performed after 15 weeks of gestation The loss rate is ∼0.1%.220

Non-invasive pre-natal testing
  • Performed for a single gene disorder.

  • Cell-free foetal DNA isolated from maternal plasma sample.

  • Offered in early pregnancy (approximately week 9); miscarriage risk not increased.

  • Not widely available (method still largely under development and therefore not readily available).

Pre-implantation genetic diagnosis
  • IVF procedure with a success rate of 25–30% per embryo transfer though dependent on the mother’s age and fertility, followed by biopsy and genetic testing of a single cell of the embryo.

  • Risks to mother and offspring of IVF, such as multiple birth, premature labour and low birth weight, as well as emotional health effects for those undergoing the procedure.

  • Availability and methods differ across countries.

IVF, in vitro fertilization.

Recommendation Table 8

Recommendations for genetic counselling and testing in cardiomyopathies

graphic
graphic
graphic
graphic
Recommendation Table 8

Recommendations for genetic counselling and testing in cardiomyopathies

graphic
graphic
graphic
graphic

6.9. Diagnostic approach to paediatric patients

Traditionally, cardiomyopathies in children have been considered to be distinct entities from adolescent and adult cardiomyopathies, with different aetiologies, natural history, and management. Although substantially rarer than in adults, contemporary data have shown that, beyond the first year of life, in most cases, paediatric cardiomyopathies represent part of the spectrum of the same diseases that are seen in adolescents and adults.245 Given their rarity, data on clinical management and outcomes are more limited than in adults, but large population-based or international consortium data have provided important information on clinical presentation, natural history, and outcomes of cardiomyopathies in children.245 Paediatric-onset cardiomyopathies often represent two opposite ends of the spectrum of heart muscle disease: (i) severe, early-onset disease, with rapid disease progression and poor prognosis, in keeping with the most severe presentations in adults; or (ii) early phenotypic expression of adult cardiomyopathy phenotypes, increasingly identified as a result of family screening. For this reason, the Task Force highlights the principle of considering cardiomyopathies in all age groups as single disease entities, with recommendations applicable to paediatric and adult populations throughout this guideline, accepting that the evidence base for many of the recommendations is significantly more limited for children. Where there are age-related differences, these are specifically highlighted.

The general approach to paediatric and adult cardiomyopathies is based on age of onset, clinical presentation, and cardiac and systemic phenotype.246 When a syndromic or metabolic disease is suspected, a step-by-step approach taking into consideration age of onset, consanguinity and family history, cardiac and systemic involvement, ECG and imaging, and laboratory work-up is recommended to define phenotype, aetiology, and tailored management.247 As in adults, clinical presentation varies, from an absence of symptoms to SCD as the first and unique manifestation.35,81,248,249

6.9.1. Infantile and early childhood-onset cardiomyopathy

In contrast, the aetiology, natural history, and outcomes of infant-onset (<1 year of age) cardiomyopathies can be substantially different than those seen in older children, adolescents, and adults. In infantile and early childhood-onset cardiomyopathies, clinical presentation, cardiac phenotype, and aetiology are the main determinants of management.2 Severe clinical onset of infantile cardiomyopathies is generally managed in intensive or subintensive care units by neonatologists and paediatric cardiologists, for respiratory distress and/or metabolic acidosis, and/or hypoglycaemia, and/or hypotonia.247,250–252 A comprehensive clinical approach, taking into consideration both the cardiac and systemic phenotype (consanguinity; dysmorphisms or skeletal anomalies; mental retardation; muscle hypotonia and weakness; hypoglycaemia with or without metabolic acidosis; increased CK and transaminases; presence of urine ketones, organic aciduria, acylcarnitine, and free fatty acid profiles; and calcium and vitamin D metabolism), and involving a multidisciplinary team (geneticist and experts in metabolic and neurological diseases), is mandatory to guide management when reversible or specific diseases are present (Figure 10).

Clinical approach to infantile and childhood cardiomyopathy.
Figure 10

Clinical approach to infantile and childhood cardiomyopathy.

ALT, alanine aminotransferase; ARVC, arrhythmogenic right ventricular cardiomyopathy; AST, aspartate transaminase; CHD, congenital heart disease; DCM, dilated cardiomyopathy; ECG, electrocardiogram; GSD, glycogen storage disorder; HCM, hypertrophic cardiomyopathy; LSD, lysosomal storage disease; LVH, left ventricular hypertrophy; MYH7, myosin heavy chain 7; NDLVC, non-dilated left ventricular cardiomyopathy; NSML, Noonan syndrome with multiple lentigines; RCM, restrictive cardiomyopathy. aSee Table 5. bSee Table 7.

In infants with HCM, after exclusion of reversible causes (maternal diabetes,253 twin–twin syndrome, corticosteroid use254,255), it is important to define, along with the pattern of hypertrophy (asymmetric, concentric, biventricular), the presence of LVOTO, diastolic and/or systolic dysfunction,1,256 and RV involvement. Early-onset sarcomeric disease (including double/compound variants) should be excluded even in the absence of a family history for HCM and SCD; these infants present with severe heart failure symptoms, and survival beyond the first year of life is uncommon.257 In contrast, clinical presentation with heart failure is rare in infants with heterozygous sarcomeric disease compared with malformation syndromes or metabolic disorders, in whom survival rates are <90% and <70% at 1 year, respectively.248,258,259 In infants with HCM, in the presence of biventricular outflow tract obstruction and ≥1 red flag for a neurocardiofaciocutaneous syndrome (dysmorphisms, cutaneous abnormalities, skeletal anomalies, etc.), a diagnosis of RASopathies should be strongly suspected.260–263 Severe LVOTO in RASopathy-related HCM often requires high-dose beta blockade and, in some cases, consideration of septal myectomy.264–267 In infants with HCM, biventricular hypertrophy, often presenting with signs of heart failure and systolic dysfunction, and ≥1 red flag for metabolic disease (muscle hypotonia, increased CK, and transaminases, consanguinity or matrilineal pattern of inheritance), it is mandatory to exclude inborn errors of metabolism, including glycogenosis type II (Pompe disease), fatty acid oxidation defects, and mitochondrial disorders.268–272 In infants with Pompe disease, enzyme replacement therapy (ERT) has been shown to result in reversal of LVH.269,273–275

In infants with DCM, reversible causes (i.e. hypocalcaemic vitamin D-dependent rickets) and CHD (aortic coarctation and ALCAPA, requiring immediate surgical management) should be ruled out.249,276,277 Viral myocarditis should also be excluded by non-invasive (i.e. laboratory) and invasive (EMB) investigations, in selected cases.278,279 Neuromuscular (dystrophin- and sarcoglycan-related cardiomyopathies) should be excluded in patients presenting with muscle hypotonia and increased CK, and a multidisciplinary approach involving a neurologist and experts in metabolic disease is required.280–282 When a DCM phenotype is associated with LV hypertrabeculation, other mitochondrial/metabolic diseases, including Barth syndrome, should be considered.283–285

Isolated RCM is rare in infants, but a mixed RCM/HCM phenotype is more frequently encountered. Familial cases are frequent, particularly in patients with an RCM/HCM phenotype.286–289 Independently of the phenotype, it is generally associated with poor prognosis, though the RCM/HCM phenotype has significantly better transplant-free survival than isolated RCM.286

Arrhythmogenic RV cardiomyopathy and non-dilated LV cardiomyopathy phenotypes are very rare in infants, and are most commonly autosomal recessive forms associated with cutaneous manifestations (e.g. Naxos disease and Carvajal syndrome),290–292 although this may reflect a lack of systematic clinical screening for these conditions in early childhood. Recent data suggest that ∼15% of ARVC patients present with paediatric-onset disease and paediatric ARVC patients more often present with severe phenotype and higher risk of SCD.293 Increasingly, children with ARVC and NDLVC phenotypes presenting with acute myocarditic presentations are recognized.294–297

6.10. General principles in the management of patients with cardiomyopathy

6.10.1. Assessment of symptoms

Some people with subtle structural abnormalities with cardiomyopathy remain asymptomatic and have a normal lifespan; however, others may develop symptoms, often many years after the appearance of ECG or imaging evidence of disease. In infants, symptoms and signs of heart failure include tachypnoea, poor feeding, excessive sweating, and failure to thrive. Older children, adolescents, and adults complain of fatigue and dyspnoea as well as chest pain, palpitations, and syncope. Because the New York Heart Association (NYHA) classification to grade heart failure is not applicable to children under the age of 5 years, the Ross Heart Failure classification has been adopted in children <5 years of age but has not been validated against outcomes.298 Systematic two-dimensional (2D) and Doppler echocardiography, resting and ambulatory ECG monitoring, and exercise testing are usually sufficient to determine the most likely cause of symptoms. Additional investigations (e.g. coronary CT scanning or coronary angiography, cardio-pulmonary exercise testing [CPET], electrophysiological study, loop recorder implantation) should be considered to investigate specific symptoms of chest pain, syncope, and palpitation, according to established clinical practice and guidelines.1,4,69,299–301 Cardiac catheterization to evaluate right and left heart function and pulmonary arterial resistance, and CPET with simultaneous measurement of respiratory gases, is not a standard part of the work-up, but remains recommended in severely symptomatic patients with systolic and/or diastolic LV dysfunction when uncertainty about filling status exists, or for those being considered for heart transplantation or mechanical circulatory support.69

6.10.2. Heart failure management

The clinical management of heart failure has been described in the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure.69 In that document, recommendations are generally independent from the aetiology of heart failure and include current medical therapy, devices, and LV assist device (LVAD)/transplantation. As such, the treatment recommendations must be regarded as generic and not specific to the different forms of cardiomyopathy. Medical therapies for HFrEF based on randomized controlled trials (RCTs) from large cohorts, including angiotensin-converting enzyme inhibitors (ACE-I)/angiotensin receptor neprilysin inhibitors (ARNIs), beta-blockers, mineralocorticoid receptor antagonists (MRA), and sodium–glucose co-transporter 2 inhibitors (SGLT2i), would be mostly applicable to genetic DCM, NDLVC, and other phenotypes associated with LV dysfunction (e.g. end-stage HCM, RCM, and ARVC). Indications for a cardiac resynchronization therapy (CRT) device and heart transplant would also be generally applicable accordingly. Recommendations for management of HFpEF would be mainly applicable to non-obstructive HCM, RCM, and cardiac amyloidosis. A Focused Update is due to be published in 2023.69a

Individual response to heart failure therapies may not be the same for different specific genetic causes, as has been demonstrated in several observational studies.302,303 Further management considerations applicable to specific cardiomyopathy subtypes in adults and children, and in particular contexts, such as pregnancy and rare metabolic genocopies, are rapidly developing304 and are discussed in the specific cardiomyopathy sections (see Sections 7.6 and 8.2.2).

Cardiac amyloidosis and some forms of RCM deserve special consideration regarding heart failure management. Fluid control and maintenance of euvolaemia are central. If heart failure symptoms are present, loop diuretics should be given, although orthostatic hypotension may cause intolerance, and excessive fluid loss may worsen symptoms due to restriction (e.g. in HCM or amyloidosis). The role of beta-blockers, ACE-Is, angiotensin receptor blockers (ARBs), or ARNIs in the treatment of these patients has not been determined and they may not be well tolerated because of hypotension.305 Moreover, withdrawal of these drugs frequently leads to improvement in symptoms and should be considered.

Heart failure with an LVEF >40–50% recovered from HFrEF or HFmrEF (improved LVEF306) is not separately considered in the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure, but is particularly important for genetic DCM, as a substantial proportion of patients with HFrEF or HFmrEF will improve their LVEF with guideline-directed medical therapy (GDMT).69 Patients and physicians are faced with the dilemma of whether to continue lifelong pharmacotherapy or wean at some point. The TRED-HF (Therapy withdrawal in REcovered Dilated cardiomyopathyHeart Failure) trial is the only RCT that evaluated if weaning GDMT is safe. The results showed that a large proportion of the patients had recurrent LV dysfunction or heart failure, so current recommendations caution against weaning.307

6.10.2.1. Preventive heart failure medical therapy of asymptomatic carriers/early disease expression

Heart failure therapy should be guided according to the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure for HFrEF, HFmrEF, and HFpEF in patients with cardiomyopathy and heart failure symptoms.69,69a Evidence for treatment recommendations in asymptomatic LV dysfunction is scarce, which presents a challenge for genetic cardiomyopathies, where a sizeable proportion of the patients are young with no or only mild symptoms, and where asymptomatic patients are frequently discovered through cascade screening. Because heart failure medication has proved to affect LV remodelling in symptomatic patients with LV dysfunction, first-line heart failure therapy may be considered in patients with early forms of DCM/NDLVC to prevent progression of LV dilatation and dysfunction (e.g. ACE-I, ARBs, beta-blockers and MRAs, Class IIb Level C). Biomarkers may help to identify pre-symptomatic patients who might benefit from early neuro-hormonal blockade.308 The effect of heart failure drugs to prevent progression into overt disease in genetic carriers of DCM-/NDLVC-causing variants is currently unsettled. A placebo-controlled trial (EARLY-Gene trial) is under way to test the utility of candesartan to prevent LV dysfunction/dilatation in this scenario (EudraCT: 2021-004577-30).

Management in other asymptomatic affected patients with diagnoses of HCM, ARVC, and RCM should be decided individually, as medication has not been proved to affect disease expression.

There is no evidence to support the use of current pharmacological agents for the prevention of disease development in non-affected carriers. Randomized controlled trials are warranted in order to address the value of new pharmacologic agents in this scenario.309

Heart failure therapies are given to children with cardiomyopathies, applying the evidence from adults to children or based on a limited number of clinical studies.310 Heart failure therapies routinely used in children with LV dysfunction are ACE-Is, beta-blockers, diuretics, and aldosterone antagonists. Angiotensin receptor blockers are an alternative for ACE-Is. Early results of the multicentric randomized control PANORAMA-HF Trial and the subsequent Food and Drug Administration (FDA) approval for ARNI in children have paved the way for this newer class of drugs for paediatric patients with symptomatic heart failure with systemic left ventricle systolic dysfunction, 1 year of age and older. Dosing recommendations in younger children are currently pending,311 but for children <40 kg a starting dose of 1.6 mg/kg titrated to a maximum of 3.1 mg/kg has been suggested.312 There are currently no clinical trial or efficacy data available for SGLT-2 inhibitors in children.

6.10.2.2. Cardiac transplantation

Orthotopic cardiac transplantation should be considered in patients with moderate-to-severe drug-refractory symptoms (NYHA functional class III–IV) who meet standard eligibility criteria (see the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure).69 This may include patients with RCM and HCM with normal LVEF but severe drug-refractory symptoms (NYHA functional class III–IV) caused by diastolic dysfunction.313–316 In patients with refractory ventricular arrhythmias that cannot be solely attributed to an acute decompensation in the setting of end-stage heart failure, a comprehensive evaluation of all potential therapeutic options (e.g. pharmacotherapy; ventricular tachycardia [VT] ablation including epicardial access if indicated and feasible; cardiac sympathetic denervation in patients with electrical storm and/or refractory polymorphic VT or rapid monomorphic VT) should be undertaken before recommending cardiac transplantation (see Section 6.10.4).

Recommendation Table 9

Recommendations for cardiac transplantation in patients with cardiomyopathy

graphic
graphic
Recommendation Table 9

Recommendations for cardiac transplantation in patients with cardiomyopathy

graphic
graphic
6.10.2.3. Left ventricular assist devices

As there are increasing numbers of patients with end-stage heart failure, and the organ donor pool remains limited, mechanical circulatory support (MCS) with an LVAD or biventricular assist device is increasingly used as a bridge to transplant. Long-term MCS should also be considered as destination therapy for cardiomyopathy patients with advanced heart failure despite optimal medical therapy who are not eligible for transplantation.69

Recommendation Table 10

Recommendation for left ventricular assist device therapy in patients with cardiomyopathy

graphic
graphic
Recommendation Table 10

Recommendation for left ventricular assist device therapy in patients with cardiomyopathy

graphic
graphic

6.10.3. Management of atrial arrhythmias

Atrial fibrillation is the most common arrhythmia in all subtypes of cardiomyopathies and is associated with an increased risk of cardio-embolic events, heart failure, and death.331–333 Data from 3208 consecutive adult patients in the EURObservational Research Programme (EORP) Cardiomyopathy Registry showed an AF prevalence of 28.2% at baseline and 31.1% during follow-up,331–333 although it differed among cardiomyopathy types (see Table 15). Overall, annual incidence in this registry was 3.0%.332,333 In patients with cardiomyopathies, the presence of AF is associated with more severe symptoms, an increased prevalence of cardiovascular risk factors and comorbidities, and an increased incidence of stroke and death (from any cause and from heart failure).332,334–336

Table 15

Atrial fibrillation burden and management in cardiomyopathies

ConditionAF epidemiologyAF management
PrevalenceAnnual incidenceAnticoagulationLong-term rate controlLong-term rhythm control
HCM17–39%331–334,365,413,421–4282.8–4.8%332,333,365Always (if no contraindication)371,429Beta-blockers (preferred)
Verapamil or diltiazem (only if preserved LVEF)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
Amiodarone, dofetilide disopyramide, sotalol,a dronedaronebAblation397,412,415,416,418,430–435
DCM25–49%331–333,426,436,437 LMNA related438–4413.8–5.5%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)c
Beta-blockers (preferred)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Amiodarone, sotalolaAblation
NDLVC39.2–43.1%d  442–4444.4–12%d  442,444,445According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Digoxin
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee, amiodarone, sotalolaAblation446
ARVC9–30%331–333,437,447–4512.1–2.8%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee (associated with beta-blockers)
Amiodarone, sotalola
Ablation
RCM45–51%331–3334.5–10.3%332,333Always (if no contraindication)Beta-blockersd (preferred)
Digoxinf
Verapamil or diltiazem (only if ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
AmiodaroneNo data
ConditionAF epidemiologyAF management
PrevalenceAnnual incidenceAnticoagulationLong-term rate controlLong-term rhythm control
HCM17–39%331–334,365,413,421–4282.8–4.8%332,333,365Always (if no contraindication)371,429Beta-blockers (preferred)
Verapamil or diltiazem (only if preserved LVEF)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
Amiodarone, dofetilide disopyramide, sotalol,a dronedaronebAblation397,412,415,416,418,430–435
DCM25–49%331–333,426,436,437 LMNA related438–4413.8–5.5%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)c
Beta-blockers (preferred)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Amiodarone, sotalolaAblation
NDLVC39.2–43.1%d  442–4444.4–12%d  442,444,445According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Digoxin
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee, amiodarone, sotalolaAblation446
ARVC9–30%331–333,437,447–4512.1–2.8%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee (associated with beta-blockers)
Amiodarone, sotalola
Ablation
RCM45–51%331–3334.5–10.3%332,333Always (if no contraindication)Beta-blockersd (preferred)
Digoxinf
Verapamil or diltiazem (only if ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
AmiodaroneNo data

AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; AV, atrioventricular; CrCl, creatinine clearance; CRT, cardiac magnetic resonance; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; LMNA, lamin A/C; LV, left ventricular; LVEF, left ventricular ejection fraction; NDLVC, non-dilated left ventricular cardiomyopathy; QRS, Q, R, and S waves of an ECG; RCM, restrictive cardiomyopathy.

Use with caution as evidence suggests that it may be associated with increased all-cause mortality.452

Dronedarone is not contraindicated in LV hypertrophy but has no significant studies in HCM.

LMNA-related DCM: increased risk of stroke (8–22%).368,440

Extrapolated from studies reporting prevalent and incident AF in HFpEF.

Contraindicated in patients with ischaemic heart disease or reduced LVEF. Should not be used in patients with CrCl <35 mL/min/1.73 m2 and significant liver disease. Should be discontinued in case of QRS widening >25% above baseline and patients with left bundle branch block or any other conduction block >120 ms. Caution when sinoatrial/atrioventricular conduction disturbances.

In cardiac amyloidosis, beta-blockers in low dosage and digoxin with caution.453,454 Non-dihydropyridine calcium channel blockers may worsen LV systolic function and heart failure.455

Table 15

Atrial fibrillation burden and management in cardiomyopathies

ConditionAF epidemiologyAF management
PrevalenceAnnual incidenceAnticoagulationLong-term rate controlLong-term rhythm control
HCM17–39%331–334,365,413,421–4282.8–4.8%332,333,365Always (if no contraindication)371,429Beta-blockers (preferred)
Verapamil or diltiazem (only if preserved LVEF)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
Amiodarone, dofetilide disopyramide, sotalol,a dronedaronebAblation397,412,415,416,418,430–435
DCM25–49%331–333,426,436,437 LMNA related438–4413.8–5.5%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)c
Beta-blockers (preferred)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Amiodarone, sotalolaAblation
NDLVC39.2–43.1%d  442–4444.4–12%d  442,444,445According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Digoxin
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee, amiodarone, sotalolaAblation446
ARVC9–30%331–333,437,447–4512.1–2.8%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee (associated with beta-blockers)
Amiodarone, sotalola
Ablation
RCM45–51%331–3334.5–10.3%332,333Always (if no contraindication)Beta-blockersd (preferred)
Digoxinf
Verapamil or diltiazem (only if ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
AmiodaroneNo data
ConditionAF epidemiologyAF management
PrevalenceAnnual incidenceAnticoagulationLong-term rate controlLong-term rhythm control
HCM17–39%331–334,365,413,421–4282.8–4.8%332,333,365Always (if no contraindication)371,429Beta-blockers (preferred)
Verapamil or diltiazem (only if preserved LVEF)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
Amiodarone, dofetilide disopyramide, sotalol,a dronedaronebAblation397,412,415,416,418,430–435
DCM25–49%331–333,426,436,437 LMNA related438–4413.8–5.5%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)c
Beta-blockers (preferred)
Digoxin
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Amiodarone, sotalolaAblation
NDLVC39.2–43.1%d  442–4444.4–12%d  442,444,445According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Digoxin
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee, amiodarone, sotalolaAblation446
ARVC9–30%331–333,437,447–4512.1–2.8%332,333According to cardio-embolic risk
(always if HF or reduced LVEF)
Beta-blockers (preferred)
Verapamil or diltiazem (only if LVEF ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control preferred in case of symptoms or/and heart failure or LV dysfunction
Flecainidee (associated with beta-blockers)
Amiodarone, sotalola
Ablation
RCM45–51%331–3334.5–10.3%332,333Always (if no contraindication)Beta-blockersd (preferred)
Digoxinf
Verapamil or diltiazem (only if ≥40%)
AV node ablation + CRT or physiological pacing388–390
Rhythm control is preferred
AmiodaroneNo data

AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; AV, atrioventricular; CrCl, creatinine clearance; CRT, cardiac magnetic resonance; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; LMNA, lamin A/C; LV, left ventricular; LVEF, left ventricular ejection fraction; NDLVC, non-dilated left ventricular cardiomyopathy; QRS, Q, R, and S waves of an ECG; RCM, restrictive cardiomyopathy.

Use with caution as evidence suggests that it may be associated with increased all-cause mortality.452

Dronedarone is not contraindicated in LV hypertrophy but has no significant studies in HCM.

LMNA-related DCM: increased risk of stroke (8–22%).368,440

Extrapolated from studies reporting prevalent and incident AF in HFpEF.

Contraindicated in patients with ischaemic heart disease or reduced LVEF. Should not be used in patients with CrCl <35 mL/min/1.73 m2 and significant liver disease. Should be discontinued in case of QRS widening >25% above baseline and patients with left bundle branch block or any other conduction block >120 ms. Caution when sinoatrial/atrioventricular conduction disturbances.

In cardiac amyloidosis, beta-blockers in low dosage and digoxin with caution.453,454 Non-dihydropyridine calcium channel blockers may worsen LV systolic function and heart failure.455

Both the 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation and the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure recommend an integrated and structured approach to facilitate guideline-adherent management. The Atrial Fibrillation Better Care (ABC) approach has been shown to reduce the risk of stroke and systemic embolism, myocardial infarction, and mortality in the general population.337–361 Although this approach has not been specifically assessed in patients with cardiomyopathies, heart failure was present in ∼20% of the individuals of these studies and, where specified, cardiomyopathy in ∼5.5–6.5%. In particular, two RCTs support integrated care.347,361 The RACE 3, combining the components of the ABC pathway into structured care, resulted in reduced AF burden and better rhythm control among 245 patients with early persistent AF and stable heart failure (119 randomized to targeted and 126 to conventional therapy).347 The mobile Atrial Fibrillation App Trial (mAFA-II), which included 714 patients with heart failure (21.5%), 54 with HCM (1.6%), and 105 with DCM (3.2%), showed the superiority of integrated care supported by mobile technology in the composite outcome of ‘ischaemic stroke/systemic thrombo-embolism, death, and re-hospitalization’ (1.9% vs. 6.0%; hazard ratio [HR] 0.39; 95% confidence interval [CI], 0.22 to 0.67; P < 0.001) and re-hospitalization rates (1.2% vs. 4.5%; HR 0.32; 95% CI, 0.17 to 0.60; P < 0.001).361 Adherence to the mobile health technology beyond 1 year was good, and was associated with a reduction in adverse clinical outcomes.362

6.10.3.1. Anticoagulation

Thrombo-embolic risk varies in different cardiomyopathy phenotypes (see Section 7).332,363–367 Cardiac amyloidosis, HCM, and RCM368 are associated with a particularly increased risk of stroke.332,365,369,370 The EORP registry indicated a worse prognosis for the population with cardiomyopathy and concurrent AF with an annual incidence of stroke/transient ischaemic attack (TIA) about three times higher in the cardiomyopathy group with AF.332,334 Hence, considering anticoagulation is key in patients with any type of AF or atrial flutter.

Importantly, patients with cardiomyopathy and AF have more cardio-embolic risk factors, including greater age, more advanced NYHA class and more frequent history of stroke/TIA, hypertension, and diabetes mellitus, among others.332,333 The CHA2DS2-VASc (congestive heart failure or left ventricular dysfunction, hypertension, age ≥75 [doubled], diabetes, stroke [doubled]-vascular disease, age 65–74, sex category [female]) score has not been specifically tested in patients with cardiomyopathies,369 and retrospective evidence suggests that it may perform suboptimally with respect to stroke prediction in HCM and ATTR amyloidosis.334,365,371–374 For this reason, although there are no RCTs evaluating the role of anticoagulation among patients with HCM, given the high incidence of stroke, prophylactic anticoagulation is recommended in all patients with HCM and AF.334,371,372,374 A similar recommendation is given in patients with AF and RCM or cardiac amyloidosis.375 In patients with DCM, NDLVC, or ARVC and AF, chronic oral anticoagulation should be considered on an individual basis, taking into consideration the CHA2DS2-VASc score, as proposed by the 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation.336 Atrial fibrillation is a rare finding in children with genetic cardiomyopathies and no data are available regarding the performance of CHA2DS2-VASc or any other risk stratification score, nor the risk and benefit of prescribing oral anticoagulation. There are no data on long-term prophylactic anticoagulation in children with DCM in sinus rhythm.

In the general population, direct-acting oral anticoagulants (DOACs) are preferred for the prevention of thrombo-embolic events in patients with AF and without severe mitral stenosis and/or mechanical valve prosthesis, as they have similar efficacy to vitamin K antagonists (VKAs) but a lower risk of intracranial haemorrhage.376 There are no randomized data comparing direct oral anticoagulants with VKAs in patients with cardiomyopathy, although data suggest that they may be used in a similar manner as the general population.373,374,377–380

6.10.3.2. Rate control

Rate control should be considered in any patient with cardiomyopathy presenting with AF.336 A strict rate control (resting heart rate <80 beats per minute [b.p.m.] and heart rate during moderate exercise <110 b.p.m.) did not show any benefit over lenient rate control (resting heart rate <110 b.p.m.) in RACE II381 and a pooled analysis of RACE II and AFFIRM.382 However, only 8–12% of patients had a history of cardiomyopathy (type unspecified) in the RACE II trial, and only 10% of the patients in RACE II and 17% of those in the pooled analysis had a history of heart failure hospitalization or NYHA class II or III, respectively.381,382 No data are available for the different cardiomyopathy subtypes, but observational studies suggest that higher heart rates are associated with worse outcomes in patients with heart failure.383,384 Accordingly, the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure consider lenient rate control to be acceptable as an initial approach but to target a lower heart rate in case of persistent symptoms or suspicion of associated tachycardia-induced cardiac dysfunction.69

Very little data are available regarding the choice of pharmacological treatment for rate control in patients with cardiomyopathies. Beta-blockers are the preferred choice in patients with cardiomyopathies given their long-established safety in the presence of LV dysfunction.385,386 Digoxin is an alternative, particularly in patients with contraindication or intolerance to beta-blockers and among patients with AF and heart failure symptoms (RATE-AF trial), having shown no difference in quality of life (QoL) at 6 months compared with bisoprolol.387 When administering digoxin, close monitoring of plasma drug levels is needed, as observational data suggest higher mortality in patients with AF, regardless of heart failure; the risk of death was related to serum digoxin concentration and was highest in patients with concentrations ≥1.2 ng/mL. On the contrary, a lower mortality with beta-blocker therapy in AF patients with concomitant heart failure has been observed. Non-dihydropyridine calcium channel blockers (CCBs) (verapamil or diltiazem) may only be used in patients with LVEF ≥40%.336

Atrioventricular node ablation is also an alternative in patients with poor ventricular rate control despite medical treatment not eligible for rhythm control by catheter ablation or in patients with biventricular pacing.336 In patients with symptomatic persistent AF (>6 months) unsuitable for AF ablation or in which AF ablation had failed, narrow QRS and at least one admission for heart failure, AV node ablation in association with CRT has been shown to be superior to rate control with pharmacological therapy, reducing the composite outcome of death due to heart failure, or hospitalization due to heart failure, or worsening heart failure,388 and all-cause mortality,389 irrespective of baseline EF (APAF-CRT Trial). Whether conduction system-pacing is a (better) alternative to CRT needs to be further explored with only one small crossover trial (ALTERNATIVE-AF) comparing His-Bundle pacing (HBP) and biventricular pacing in 50 patients with LVEF ≤40% with persistent AF undergoing AV node ablation.390 In this study, both arms significantly improved LVEF at 9 months, with a small, but statistically significant superiority with HBP.69,336

6.10.3.3. Rhythm control

Atrial fibrillation can result in haemodynamic and clinical decompensation due to shortening of the diastolic filling time with rapid heart rates and dependence on atrial contraction for LV filling. Therefore, maintenance of sinus rhythm is highly desirable and a rhythm control strategy is preferred, particularly in the presence of symptoms.

Regarding long-term pharmacological treatment,336 antiarrhythmic drugs (AADs) have shown limited success in maintaining sinus rhythm over time both in the general population and in patients with cardiomyopathies,391–393 show high rates of withdrawal due to intolerance,394 and, most importantly, are associated with significant side effects, including proarrhythmia and extracardiac side effects, and, in some cases (sotalol and class IA drugs, such as quinidine and disopyramide), increased mortality.394 As a consequence, a degree of caution is recommended when using antiarrhythmic drugs in this population. Data on antiarrhythmic therapy for the specific management of AF in the context of genetic cardiomyopathies other than HCM are scarce. It is important to note the potential for proarrhythmia of class I antiarrhythmics, particularly in the presence of significant structural heart disease; these should therefore be used with caution. Antiarrhythmic drug–drug treatment has mostly been limited to amiodarone or sotalol, as there are no available data regarding other antiarrhythmics such as dofetilide or dronedarone. Importantly, sotalol should not be used in patients with HFrEF, significant LVH, prolonged QT, asthma, hypokalaemia, or creatinine clearance (CrCl)<30 ml/min. Likewise, dronedarone should be avoided in patients with recent decompensated heart failure or permanent AF as it has been shown to increase mortality.395,396

Catheter ablation of AF is a safe and superior alternative to AAD therapy for maintenance of sinus rhythm, reducing AF-related symptoms, and improving QoL, and can be considered an alternative to AAD therapy in practically any type and context of AF.336,397 In patients with AF and normal LVEF, catheter ablation has not been shown to reduce total mortality or stroke.398 In selected patients with HFrEF,399–401 ablation has shown a reduction in all-cause mortality and hospitalizations, and should be considered as a first-line option. In the general AF population, the Early Treatment of Atrial Fibrillation for Stroke Prevention Trial (EAST-AFNET 4) randomized 2789 patients with early AF and associated cardiovascular comorbidities to an early rhythm control strategy or usual care (28.6% with heart failure).402 The trial was stopped early after a median follow-up of 5.1 years for a lower occurrence of the primary outcome of death, stroke, or hospitalization for worsening heart failure or acute coronary syndrome in the patients in the early rhythm control group vs. those assigned to usual care. A pre-specified analysis evaluated the effects in patients with heart failure, showing the benefit of early rhythm control in this subgroup of patients,403 findings which corroborated those of the CABANA trial.400 In patients with AF and heart failure, several RCTs have demonstrated an improvement in outcomes with catheter ablation when compared with medical therapy.399–401,404–409 Some observational studies in patients with HFpEF have also suggested better results in terms of freedom from AF and all-cause mortality,410 but proper RCTs are warranted.

The role of catheter ablation in patients with cardiomyopathies has been reported in several registries, mainly in HCM patients.397,411–420 Overall, maintenance is achieved in up to two-thirds of patients, although repeat procedures or continuation of antiarrhythmic medications are often necessary.397,411,415–419 Patients with cardiomyopathies may have a higher risk of AF recurrence, particularly in the presence of atrial remodelling/dilatation.397

6.10.3.4. Comorbidities and risk factor management

Cardiovascular risk factors and comorbidities are also more frequent in patients with cardiomyopathies and AF. These include smoking, alcohol consumption, hypertension, diabetes mellitus type 2, hyperlipidaemia, renal impairment, chronic obstructive pulmonary disease, valvular and ischaemic heart disease, and anaemia.332,334 Furthermore, these patients have a larger body mass index and report less physical activity than those without AF.332,334 These risk factors and comorbidities are associated with the risk of AF and its complications and should therefore be appropriately identified and managed to prevent AF progression and the occurrence of adverse outcomes.336

Recommendation Table 11

Recommendations for management of atrial fibrillation and atrial flutter in patients with cardiomyopathy

graphic
graphic
graphic
graphic
Recommendation Table 11

Recommendations for management of atrial fibrillation and atrial flutter in patients with cardiomyopathy

graphic
graphic
graphic
graphic

6.10.4. Management of ventricular arrhythmias

Ventricular arrhythmias, particularly in the form of electrical storm and/or repetitive appropriate ICD interventions, contribute to a significantly increased risk of morbidity and mortality in patients with cardiomyopathies.299

The 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death provide detailed recommendations on acute and long-term management of ventricular arrhythmias in patients with cardiomyopathies.299 Limited data exist addressing ventricular arrhythmia management in patients with specific genetic cardiomyopathies. Nonetheless, some general concepts can be highlighted:

  • Any reversible cause and/or precipitating factor, such as electrolyte imbalances, ischaemia, hypoxaemia, or drugs, should be identified and corrected when possible.

  • Extensive efforts should be made in the attempt to understand the aetiology (i.e. underlying mechanism and substrate, and their relationship with the underlying cardiomyopathy) as this will influence the choice of treatment.

  • Acute termination of sustained ventricular arrhythmias can be achieved with electrical cardioversion, AADs, or pacing. The initial choice of treatment will depend on the haemodynamic tolerance, the underlying aetiology, and the patient profile.

  • In patients presenting with electrical storm, mild-to-moderate sedation is recommended to alleviate psychological distress and reduce sympathetic tone. If the electrical storm remains intractable despite antiarrhythmic therapies, deep sedation/intubation should be considered.

  • In case of incessant ventricular arrhythmias and electrical storm not responding to antiarrhythmic medication, catheter ablation is recommended. In refractory cases or whenever VT ablation is either not indicated or not immediately available, autonomic modulation (i.e. stellate ganglion block or cardiac sympathetic denervation, depending on the setting) and/or MCS may be considered.

  • In patients with cardiomyopathies and scar-related ventricular arrhythmias, the therapeutic arsenal for long-term prevention of recurrent ventricular arrhythmias includes antiarrhythmic medications (mostly limited to beta-blockers, sotalol, and amiodarone) and catheter ablation (particularly in the case of sustained monomorphic VT or in the case of polymorphic VT triggered by a premature ventricular complex of similar morphology). Additional strategies, performed by experienced centres, may be considered, depending on the characteristics of the patient and the ventricular arrhythmia, including acute neuromodulation strategies (stellate ganglion block and thoracic epidural anaesthesia), chronic neuromodulation strategies (cardiac sympathetic denervation), and stereotactic non-invasive VT ablation.514–520 Limited data are available at present concerning the long-term cardiac and extracardiac safety of stereotactic non-invasive VT ablation, as well as the dose–response relationship, therefore its usage should be limited to compassionate cases or within prospective clinical studies.

  • The acute as well as the chronic management of patients with cardiomyopathies and refractory ventricular arrhythmias, particularly in case of concomitant moderate-to-severe ventricular dysfunction, should involve an integrated evaluation by a heart team including cardiomyopathy specialists, electrophysiologists with specific experience in catheter ablation of ventricular arrhythmias and neuromodulation, anaesthesiologists, and cardiac surgeons.

6.10.5. Device therapy: implantable cardioverter defibrillator

Implantable cardioverter defibrillators are effective at correcting potentially lethal ventricular arrhythmias and preventing SCD, but are also associated with complications, particularly in young patients who will require several replacements during their lifetimes. Implantable cardioverter defibrillators reduce mortality in survivors of cardiac arrest and in patients who have experienced haemodynamically compromising sustained ventricular arrhythmias.521–523 An ICD is recommended in such patients when the intent is to increase survival; the decision to implant should consider the patient’s view and their QoL, as well as the absence of other diseases likely to cause death within the following year.

Arrhythmic risk calculators may be useful tools to predict the risk of SCD and, where available, they may provide a clinical benefit compared with a risk factor approach.524–526 The issue of the threshold for ICD implantation may be a reasonable concern as every cut-off point comes with a trade-off between unnecessary ICDs with their potential complications vs. the potential for unprotected SCD. The relative weight of these opposing undesirable events varies significantly from one person to another and should be part of the individualized decision-making process. Risk stratification strategies in each cardiomyopathy and the role of ICDs for primary prevention are discussed in Section 7.

The 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death provide detailed recommendations regarding optimal device programming and prevention/treatment of inappropriate therapy. The implantation of conditional devices is reasonable taking into account the expected need for CMR during follow-up. In children, simpler ICD devices (e.g. single chamber/single coil or subcutaneous) should be considered, bearing in mind specific issues of body size/shape and growth. The wearable cardioverter defibrillator has been shown to detect and treat ventricular arrhythmias successfully.527 However, data on its benefit for primary prevention other than the early phase of myocardial infarction (e.g. myocarditis, PPCM etc.) are scarce and no recommendation can be made at present.

Recommendation Table 12

Recommendations for implantable cardioverter defibrillator in patients with cardiomyopathy

graphic
graphic
graphic
graphic
Recommendation Table 12

Recommendations for implantable cardioverter defibrillator in patients with cardiomyopathy

graphic
graphic
graphic
graphic

6.10.6. Routine follow-up of patients with cardiomyopathy

In general, patients with cardiomyopathy require lifelong follow-up to detect changes in symptoms, risk of adverse events, ventricular function, and cardiac rhythm.

The frequency of monitoring is determined by the severity of disease, age, and symptoms. A clinical examination, including 12-lead ECG and TTE, should be performed every 1–2 years, or sooner should patients complain of new symptoms. Ambulatory electrocardiography is recommended every 1–2 years in most patients to detect asymptomatic atrial and ventricular arrhythmia, and is indicated whenever patients experience syncope or palpitations. Cardiac magnetic resonance evaluation should be considered every 2–5 years or more frequently in patients with progressive disease (see Section 6.7.3). Cardio-pulmonary exercise testing can provide objective evidence for worsening disease but need only be performed every 2–3 years unless there is a change in symptoms. Ergometry and treadmill exercise testing may also provide valuable functional information in patients unable to perform CPET.

Recommendation Table 13

Recommendations for routine follow-up of patients with cardiomyopathy

graphic
graphic
Recommendation Table 13

Recommendations for routine follow-up of patients with cardiomyopathy

graphic
graphic

6.11. Family screening and follow-up evaluation of relatives

All first-degree relatives of patients with cardiomyopathy should be offered clinical screening with ECG and cardiac imaging (echocardiogram [ECHO] and/or CMR). In families in whom a disease-causing genetic variant has been identified, cascade genetic testing should be offered (see Section 6.8.3). Individuals found not to carry the familial variant and who do not have a clinical phenotype can usually be discharged, with advice to seek re-assessment if they develop symptoms or when new clinically relevant data emerge in the family. Those relatives harbouring the familial genetic variant(s) should undergo regular clinical evaluation with ECG, multimodality cardiac imaging, and additional investigations (e.g. Holter monitoring) guided by age, family phenotype, and genotype (Figure 11). Similarly, if a genetic cause of the disease has not been identified, either because P/LP variants are absent in the proband or because genetic testing has not been performed, clinical follow-up of all first-degree relatives is recommended; in families without a known disease-causing variant, children should be offered ongoing clinical surveillance, due to age-related penetrance, and ongoing surveillance should also be offered to adult relatives dependent on family history and other factors. In families where there is only one affected individual and where no genetic variant has been identified, the frequency and duration of clinical follow-up may be reduced (see Figure 11).

Algorithm for the approach to family screening and follow-up of family members.
Figure 11

Algorithm for the approach to family screening and follow-up of family members.

P/LP, pathogenic/likely pathogenic, VUS, variant of unknown significance. aIf no additional affected relatives and no variant identified on genetic testing, consider earlier termination of clinical screening.

Generally, the frequency of the clinical cardiac evaluation in relatives will be based on the inheritance pattern, the risk of events in the affected individual(s), and the quality-adjusted life-year. It would also depend on age, type of cardiomyopathy, and family history (penetrance, phenotype expression, and risk of complications in affected relatives).

Disease-penetrance studies have demonstrated a similar sigmoid shape pattern of phenotypic expression throughout life in families with confirmed genetic cardiomyopathies. The penetrance during childhood is ∼5% during the first decade of life, increasing to 10–20% per decade from the second to the seventh decades, after which the slope flattens to 5–10% in the last decades, although up to 25% of diagnoses can be made in individuals older than 65 years in some populations.544 The slope in childhood and early adulthood can be steeper (20% per decade) and similar to that in middle age for HCM, where male sex, subtle ECG abnormalities, and particular genes are predictors of disease expression during follow-up.178

Penetrance in most cardiomyopathies is incomplete, reaching 70–90% by the age of 70 years in families with cardiomyopathy.178 With some exceptions using the current diagnostic criteria, the penetrance of the disease in women has been shown to be delayed (shifted) by 10 years compared with men.178,545–548

Cardiac screening in: (i) carriers of genetic P/LP variants associated with cardiomyopathies; or (ii) in those with demonstration of a familial disease should be offered from childhood to old age. The proposed frequency of screening is every 1–3 years with ECG and ECHO (plus additional tests where this is considered appropriate) before the age of 60 years, and then every 3–5 years thereafter.

These recommendations apply to families affected by cardiomyopathy. The penetrance of similar variants identified outside this context is likely to be much lower, and the benefits and harm of screening and surveillance remain under evaluation.549–551

Recommendation Table 14

Recommendations for family screening and follow-up evaluation of relatives

graphic
graphic
Recommendation Table 14

Recommendations for family screening and follow-up evaluation of relatives

graphic
graphic

6.11.1. Special considerations in family screening

If the comprehensive study of the index cardiomyopathy patient (including negative genetic testing) and first-degree relatives from informative families (i.e. with a large enough pedigree) leads to the conclusion that the cardiomyopathy presents in isolation (i.e. the index patient is the only individual affected), termination of periodic surveillance could be considered in first-degree relatives ≥50 years of age with normal cardiac investigations.

When the pattern of inheritance is likely to be, or is definitively, other than autosomal dominant, consideration for periodic evaluation of relatives should be individualized, e.g. (i) heterozygous carriers from clear recessive forms of cardiomyopathy could be discharged; (ii) heterozygous carriers of X-linked disease may delay cardiac evaluation, as phenotype may express later in life; and (iii) follow-up in families with more than one likely or definitively pathogenic variant (oligo-polygenicity) should be discussed in the cardiomyopathy team.

6.12. Psychological support in cardiomyopathy patients and family members

Adjusting to a diagnosis of an inherited cardiomyopathy can pose a psychosocial challenge. This includes coming to terms with a new diagnosis, exclusion from competitive sports, or living with the small risk of SCD.205 While studies show patients with inherited cardiomyopathies adjust well following an ICD, there is an important subgroup who do require additional support.552–556 The decision to have an ICD, and living with the device, can also pose psychological challenges, especially in those who are young or who have experienced multiple shocks and/or have poor baseline psychosocial functioning.553,554,557 The SCD of a young relative not only leads to profound grief, but one in two relatives report post-traumatic stress or prolonged grief on average 6 years after the death.558 Clinical psychological support for patients and their families affected by inherited cardiomyopathies is an important aspect of the multidisciplinary team’s care approach and should be available as required.559 Clinicians should be aware of the potential for poor psychological outcomes and should have a low threshold for referral.

Psychological challenges for patients and their family members are summarized in Table 16. While many patients and family members will benefit from psychosocial counselling provided by any number of healthcare professionals, it is important to highlight that for some, treatment by a trained professional such as a clinical psychologist is required.

Table 16

Psychological considerations

Patient groupPsychological considerations
New diagnosis
  • Stigma associated with cardiovascular disease and misconception that it only affects older people.

  • Fear of sudden cardiac death can shake confidence and create anxiety around exercise.

  • Fear of inheritance risk to other relatives, especially children.

  • Confidence and self-efficacy to manage their disease.

  • Direct experience with the disease will affect perceptions about prognosis.211

ICD
  • Most patients will adjust well following ICD insertion, although there might be an initial decline in health-related quality of life and psychological well-being, this often returns to normal.552,560,561

  • Up to 30% will develop anxiety and/or symptoms of post-traumatic stress and need additional support.562

  • Those who are young, who experience multiple ICD shocks, and/or have poor baseline psychological functioning are at greater risk of poor psychological outcomes.553–555,561,563

  • In young people, especially women, body image concerns can be a major consideration.554

  • Decision-making for those recommended to have an ICD should be patient-centred and include balanced discussion of benefits and risks and careful attention to questions and concerns.564

Exercise restrictions
  • Physical inactivity is a major determinant of poor health outcomes.

  • Can reduce health-related quality of life for those who become fearful of performing even low-intensity exercise.

  • Athletes who are recommended to reduce their activity levels can experience a profound grief and difficulty adjusting to this advice.565

  • Patient-centred discussions and careful attention to concerns is critical in helping to support people make drastic lifestyle changes.566–568

Family history of young SCD
  • Relatives who experience the SCD of a young relative have significant risk of poor psychological functioning, including post-traumatic stress and prolonged grief.558

  • Grief is a normal response to a loss. Prolonged grief occurs when the grieving process becomes ‘stuck’.569

  • Those who witness the death or discover the decedents body have a greater risk of psychological difficulties.558

  • Mothers of the decedent have greater anxiety.558

  • Psychological support for family members is an important and often unmet need following a young SCD.570,571

Children and adolescents
  • Diagnosis during childhood can raise anxiety especially among parents. Access to resources to support practical issues like information for schools is important.

  • Navigating transition from paediatric to adult care can be challenging for children and their families.

  • Decision-making regarding genetic testing of asymptomatic children can often benefit from the inclusion of a clinical psychologist to support adjustment to the result.

Symptomatic disease
  • Those managing symptoms will likely perceive a greater impact on their health-related quality of life. Factors influencing self-efficacy will impact on a patient’s ability to manage their disease, including medication adherence.572

  • Need for major intervention such as cardiac transplantation can raise significant psychological challenges and clinical psychological support is very important.573

Genetic testing
  • Despite potential adjustment issues, most patients who undertake genetic testing do not report distress.574

  • Genetic counselling should cover any psychosocial concerns or needs.204

  • Additional support to patients to convey the genetic risk information to at-risk family members should be provided as necessary.575

Patient groupPsychological considerations
New diagnosis
  • Stigma associated with cardiovascular disease and misconception that it only affects older people.

  • Fear of sudden cardiac death can shake confidence and create anxiety around exercise.

  • Fear of inheritance risk to other relatives, especially children.

  • Confidence and self-efficacy to manage their disease.

  • Direct experience with the disease will affect perceptions about prognosis.211

ICD
  • Most patients will adjust well following ICD insertion, although there might be an initial decline in health-related quality of life and psychological well-being, this often returns to normal.552,560,561

  • Up to 30% will develop anxiety and/or symptoms of post-traumatic stress and need additional support.562

  • Those who are young, who experience multiple ICD shocks, and/or have poor baseline psychological functioning are at greater risk of poor psychological outcomes.553–555,561,563

  • In young people, especially women, body image concerns can be a major consideration.554

  • Decision-making for those recommended to have an ICD should be patient-centred and include balanced discussion of benefits and risks and careful attention to questions and concerns.564

Exercise restrictions
  • Physical inactivity is a major determinant of poor health outcomes.

  • Can reduce health-related quality of life for those who become fearful of performing even low-intensity exercise.

  • Athletes who are recommended to reduce their activity levels can experience a profound grief and difficulty adjusting to this advice.565

  • Patient-centred discussions and careful attention to concerns is critical in helping to support people make drastic lifestyle changes.566–568

Family history of young SCD
  • Relatives who experience the SCD of a young relative have significant risk of poor psychological functioning, including post-traumatic stress and prolonged grief.558

  • Grief is a normal response to a loss. Prolonged grief occurs when the grieving process becomes ‘stuck’.569

  • Those who witness the death or discover the decedents body have a greater risk of psychological difficulties.558

  • Mothers of the decedent have greater anxiety.558

  • Psychological support for family members is an important and often unmet need following a young SCD.570,571

Children and adolescents
  • Diagnosis during childhood can raise anxiety especially among parents. Access to resources to support practical issues like information for schools is important.

  • Navigating transition from paediatric to adult care can be challenging for children and their families.

  • Decision-making regarding genetic testing of asymptomatic children can often benefit from the inclusion of a clinical psychologist to support adjustment to the result.

Symptomatic disease
  • Those managing symptoms will likely perceive a greater impact on their health-related quality of life. Factors influencing self-efficacy will impact on a patient’s ability to manage their disease, including medication adherence.572

  • Need for major intervention such as cardiac transplantation can raise significant psychological challenges and clinical psychological support is very important.573

Genetic testing
  • Despite potential adjustment issues, most patients who undertake genetic testing do not report distress.574

  • Genetic counselling should cover any psychosocial concerns or needs.204

  • Additional support to patients to convey the genetic risk information to at-risk family members should be provided as necessary.575

ICD, implantable cardioverter defibrillator; SCD, sudden cardiac death.

Table 16

Psychological considerations

Patient groupPsychological considerations
New diagnosis
  • Stigma associated with cardiovascular disease and misconception that it only affects older people.

  • Fear of sudden cardiac death can shake confidence and create anxiety around exercise.

  • Fear of inheritance risk to other relatives, especially children.

  • Confidence and self-efficacy to manage their disease.

  • Direct experience with the disease will affect perceptions about prognosis.211

ICD
  • Most patients will adjust well following ICD insertion, although there might be an initial decline in health-related quality of life and psychological well-being, this often returns to normal.552,560,561

  • Up to 30% will develop anxiety and/or symptoms of post-traumatic stress and need additional support.562

  • Those who are young, who experience multiple ICD shocks, and/or have poor baseline psychological functioning are at greater risk of poor psychological outcomes.553–555,561,563

  • In young people, especially women, body image concerns can be a major consideration.554

  • Decision-making for those recommended to have an ICD should be patient-centred and include balanced discussion of benefits and risks and careful attention to questions and concerns.564

Exercise restrictions
  • Physical inactivity is a major determinant of poor health outcomes.

  • Can reduce health-related quality of life for those who become fearful of performing even low-intensity exercise.

  • Athletes who are recommended to reduce their activity levels can experience a profound grief and difficulty adjusting to this advice.565

  • Patient-centred discussions and careful attention to concerns is critical in helping to support people make drastic lifestyle changes.566–568

Family history of young SCD
  • Relatives who experience the SCD of a young relative have significant risk of poor psychological functioning, including post-traumatic stress and prolonged grief.558

  • Grief is a normal response to a loss. Prolonged grief occurs when the grieving process becomes ‘stuck’.569

  • Those who witness the death or discover the decedents body have a greater risk of psychological difficulties.558

  • Mothers of the decedent have greater anxiety.558

  • Psychological support for family members is an important and often unmet need following a young SCD.570,571

Children and adolescents
  • Diagnosis during childhood can raise anxiety especially among parents. Access to resources to support practical issues like information for schools is important.

  • Navigating transition from paediatric to adult care can be challenging for children and their families.

  • Decision-making regarding genetic testing of asymptomatic children can often benefit from the inclusion of a clinical psychologist to support adjustment to the result.

Symptomatic disease
  • Those managing symptoms will likely perceive a greater impact on their health-related quality of life. Factors influencing self-efficacy will impact on a patient’s ability to manage their disease, including medication adherence.572

  • Need for major intervention such as cardiac transplantation can raise significant psychological challenges and clinical psychological support is very important.573

Genetic testing
  • Despite potential adjustment issues, most patients who undertake genetic testing do not report distress.574

  • Genetic counselling should cover any psychosocial concerns or needs.204

  • Additional support to patients to convey the genetic risk information to at-risk family members should be provided as necessary.575

Patient groupPsychological considerations
New diagnosis
  • Stigma associated with cardiovascular disease and misconception that it only affects older people.

  • Fear of sudden cardiac death can shake confidence and create anxiety around exercise.

  • Fear of inheritance risk to other relatives, especially children.

  • Confidence and self-efficacy to manage their disease.

  • Direct experience with the disease will affect perceptions about prognosis.211

ICD
  • Most patients will adjust well following ICD insertion, although there might be an initial decline in health-related quality of life and psychological well-being, this often returns to normal.552,560,561

  • Up to 30% will develop anxiety and/or symptoms of post-traumatic stress and need additional support.562

  • Those who are young, who experience multiple ICD shocks, and/or have poor baseline psychological functioning are at greater risk of poor psychological outcomes.553–555,561,563

  • In young people, especially women, body image concerns can be a major consideration.554

  • Decision-making for those recommended to have an ICD should be patient-centred and include balanced discussion of benefits and risks and careful attention to questions and concerns.564

Exercise restrictions
  • Physical inactivity is a major determinant of poor health outcomes.

  • Can reduce health-related quality of life for those who become fearful of performing even low-intensity exercise.

  • Athletes who are recommended to reduce their activity levels can experience a profound grief and difficulty adjusting to this advice.565

  • Patient-centred discussions and careful attention to concerns is critical in helping to support people make drastic lifestyle changes.566–568

Family history of young SCD
  • Relatives who experience the SCD of a young relative have significant risk of poor psychological functioning, including post-traumatic stress and prolonged grief.558

  • Grief is a normal response to a loss. Prolonged grief occurs when the grieving process becomes ‘stuck’.569

  • Those who witness the death or discover the decedents body have a greater risk of psychological difficulties.558

  • Mothers of the decedent have greater anxiety.558

  • Psychological support for family members is an important and often unmet need following a young SCD.570,571

Children and adolescents
  • Diagnosis during childhood can raise anxiety especially among parents. Access to resources to support practical issues like information for schools is important.

  • Navigating transition from paediatric to adult care can be challenging for children and their families.

  • Decision-making regarding genetic testing of asymptomatic children can often benefit from the inclusion of a clinical psychologist to support adjustment to the result.

Symptomatic disease
  • Those managing symptoms will likely perceive a greater impact on their health-related quality of life. Factors influencing self-efficacy will impact on a patient’s ability to manage their disease, including medication adherence.572

  • Need for major intervention such as cardiac transplantation can raise significant psychological challenges and clinical psychological support is very important.573

Genetic testing
  • Despite potential adjustment issues, most patients who undertake genetic testing do not report distress.574

  • Genetic counselling should cover any psychosocial concerns or needs.204

  • Additional support to patients to convey the genetic risk information to at-risk family members should be provided as necessary.575

ICD, implantable cardioverter defibrillator; SCD, sudden cardiac death.

Recommendation Table 15

Recommendations for psychological support in patients and family members with cardiomyopathies

graphic
graphic
Recommendation Table 15

Recommendations for psychological support in patients and family members with cardiomyopathies

graphic
graphic

6.13. The patient pathway

The systematic, multiparametric approach to diagnosis and evaluation of patients with suspected cardiomyopathy described in this section allows clinicians to establish the presence of a cardiomyopathy and identify its aetiology and guides the management of symptoms and prevention of disease-related complications. While many of the aspects of clinical care and the accompanying recommendations are common to all cardiomyopathy phenotypes, achieving an aetiological diagnosis is key to delivering disease-specific management; this is discussed in detail in the subsequent sections of this guideline (see Section 7).

7. Specific cardiomyopathy phenotypes

7.1. Hypertrophic cardiomyopathy

The 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy provide detailed recommendations on the assessment and management of patients with HCM.1 The aim in this guideline is to provide a focused update to the 2014 document, highlighting novel aspects and signposting the reader to the assessment and management of HCM in adults and children. Further details to support the recommendations are available in Supplementary data online, Table S1.

7.1.1. Diagnosis

7.1.1.1. Diagnostic criteria

Adults: in an adult, HCM is defined by an LV wall thickness ≥15 mm in any myocardial segment that is not explained solely by loading conditions. Lesser degrees of wall thickening (13–14 mm) require evaluation of other features including family history, genetic findings, and ECG abnormalities.

Children: the diagnosis of HCM requires an LV wall thickness more than 2 standard deviations greater than the predicted mean (z-score >2).578

Relatives: the clinical diagnosis of HCM in adult first-degree relatives of patients with unequivocal disease is based on the presence of LV wall thickness ≥13 mm. In child first-degree relatives with LV wall thickness z-scores of <2, the presence of associated morphological or ECG abnormalities should raise the suspicion but are not on their own diagnostic for HCM.

7.1.1.2. Diagnostic work-up

The initial work-up for HCM includes personal and family history, physical examination, electrocardiography, cardiac imaging, and first-line laboratory tests, as described in Section 6.

7.1.1.3. Echocardiography

As increased ventricular wall thickness can be found at any location (including the right ventricle), the presence, distribution, and severity of hypertrophy should be documented using a standardized protocol for cross-sectional imaging from several projections.579  Table 17 summarizes the key imaging features to assess in patients with suspected or confirmed HCM. Several imaging features can point to a specific diagnosis (Table 18 and Section 6).62

Table 17

Imaging evaluation in hypertrophic cardiomyopathy

Item to assessPrimary imaging modalityComments
LV wall thicknessECHO/CMR
  • All LV segments from base to apex examined in end-diastole, preferably in the 2D short-axis view, ensuring that the wall thickness is recorded at mitral, mid-LV, and apical levels.

  • CMR is superior in the detection of LV apical and anterolateral hypertrophy, aneurysms,580 and thrombi,581 and is more sensitive in the detection of subtle markers of disease in patients with sarcomeric protein gene variants (e.g. myocardial crypts, papillary muscle abnormalities).159,582,583

Systolic function (global and regional)ECHO/CMR
  • Ejection fraction is a suboptimal measure of LV systolic performance when hypertrophy is present.

  • Doppler myocardial velocities and deformation parameters (strain and strain rate) are typically reduced at the site of hypertrophy despite a normal EF and may be abnormal before the development of increased wall thickness in genetically affected patients.

Diastolic functionECHO
  • Routine examination should include mitral inflow assessment, tissue Doppler imaging, pulmonary vein flow velocities, pulmonary artery systolic pressure, and LA size/volume.

Mitral valveECHO
  • Assess presence and degree of SAM and mitral regurgitation. The presence of a central- or anteriorly directed jet of mitral regurgitation should raise suspicion of an intrinsic/primary mitral valve abnormality and prompt further assessment.

LVOTECHO
LA dimensionsECHO/CMR
  • Provides important prognostic information.365,525,584

  • Most common mechanisms of LA enlargement are SAM-related mitral regurgitation and elevated LV filling pressures.

Myocardial fibrosis/LGECMR
  • The distribution and severity of interstitial expansion can suggest specific diagnoses. Anderson–Fabry disease is characterized by a reduction in non-contrast T1 signal and the presence of posterolateral LGE.134,155 In cardiac amyloidosis, there is often global, subendocardial or segmental LGE and a highly specific pattern of myocardial and blood-pool gadolinium kinetics caused by similar myocardial and blood T1 signals.585,586

Item to assessPrimary imaging modalityComments
LV wall thicknessECHO/CMR
  • All LV segments from base to apex examined in end-diastole, preferably in the 2D short-axis view, ensuring that the wall thickness is recorded at mitral, mid-LV, and apical levels.

  • CMR is superior in the detection of LV apical and anterolateral hypertrophy, aneurysms,580 and thrombi,581 and is more sensitive in the detection of subtle markers of disease in patients with sarcomeric protein gene variants (e.g. myocardial crypts, papillary muscle abnormalities).159,582,583

Systolic function (global and regional)ECHO/CMR
  • Ejection fraction is a suboptimal measure of LV systolic performance when hypertrophy is present.

  • Doppler myocardial velocities and deformation parameters (strain and strain rate) are typically reduced at the site of hypertrophy despite a normal EF and may be abnormal before the development of increased wall thickness in genetically affected patients.

Diastolic functionECHO
  • Routine examination should include mitral inflow assessment, tissue Doppler imaging, pulmonary vein flow velocities, pulmonary artery systolic pressure, and LA size/volume.

Mitral valveECHO
  • Assess presence and degree of SAM and mitral regurgitation. The presence of a central- or anteriorly directed jet of mitral regurgitation should raise suspicion of an intrinsic/primary mitral valve abnormality and prompt further assessment.

LVOTECHO
LA dimensionsECHO/CMR
  • Provides important prognostic information.365,525,584

  • Most common mechanisms of LA enlargement are SAM-related mitral regurgitation and elevated LV filling pressures.

Myocardial fibrosis/LGECMR
  • The distribution and severity of interstitial expansion can suggest specific diagnoses. Anderson–Fabry disease is characterized by a reduction in non-contrast T1 signal and the presence of posterolateral LGE.134,155 In cardiac amyloidosis, there is often global, subendocardial or segmental LGE and a highly specific pattern of myocardial and blood-pool gadolinium kinetics caused by similar myocardial and blood T1 signals.585,586

2D, two-dimensional; CMR, cardiac magnetic resonance; ECHO, echocardiogram; EF, ejection fraction; LA, left atrium; LGE, late gadolinium enhancement; LV, left ventricular; LVOT, left ventricular outflow tract; SAM, systolic anterior motion; SCD, sudden cardiac death.

Table 17

Imaging evaluation in hypertrophic cardiomyopathy

Item to assessPrimary imaging modalityComments
LV wall thicknessECHO/CMR
  • All LV segments from base to apex examined in end-diastole, preferably in the 2D short-axis view, ensuring that the wall thickness is recorded at mitral, mid-LV, and apical levels.

  • CMR is superior in the detection of LV apical and anterolateral hypertrophy, aneurysms,580 and thrombi,581 and is more sensitive in the detection of subtle markers of disease in patients with sarcomeric protein gene variants (e.g. myocardial crypts, papillary muscle abnormalities).159,582,583

Systolic function (global and regional)ECHO/CMR
  • Ejection fraction is a suboptimal measure of LV systolic performance when hypertrophy is present.

  • Doppler myocardial velocities and deformation parameters (strain and strain rate) are typically reduced at the site of hypertrophy despite a normal EF and may be abnormal before the development of increased wall thickness in genetically affected patients.

Diastolic functionECHO
  • Routine examination should include mitral inflow assessment, tissue Doppler imaging, pulmonary vein flow velocities, pulmonary artery systolic pressure, and LA size/volume.

Mitral valveECHO
  • Assess presence and degree of SAM and mitral regurgitation. The presence of a central- or anteriorly directed jet of mitral regurgitation should raise suspicion of an intrinsic/primary mitral valve abnormality and prompt further assessment.

LVOTECHO
LA dimensionsECHO/CMR
  • Provides important prognostic information.365,525,584

  • Most common mechanisms of LA enlargement are SAM-related mitral regurgitation and elevated LV filling pressures.

Myocardial fibrosis/LGECMR
  • The distribution and severity of interstitial expansion can suggest specific diagnoses. Anderson–Fabry disease is characterized by a reduction in non-contrast T1 signal and the presence of posterolateral LGE.134,155 In cardiac amyloidosis, there is often global, subendocardial or segmental LGE and a highly specific pattern of myocardial and blood-pool gadolinium kinetics caused by similar myocardial and blood T1 signals.585,586

Item to assessPrimary imaging modalityComments
LV wall thicknessECHO/CMR
  • All LV segments from base to apex examined in end-diastole, preferably in the 2D short-axis view, ensuring that the wall thickness is recorded at mitral, mid-LV, and apical levels.

  • CMR is superior in the detection of LV apical and anterolateral hypertrophy, aneurysms,580 and thrombi,581 and is more sensitive in the detection of subtle markers of disease in patients with sarcomeric protein gene variants (e.g. myocardial crypts, papillary muscle abnormalities).159,582,583

Systolic function (global and regional)ECHO/CMR
  • Ejection fraction is a suboptimal measure of LV systolic performance when hypertrophy is present.

  • Doppler myocardial velocities and deformation parameters (strain and strain rate) are typically reduced at the site of hypertrophy despite a normal EF and may be abnormal before the development of increased wall thickness in genetically affected patients.

Diastolic functionECHO
  • Routine examination should include mitral inflow assessment, tissue Doppler imaging, pulmonary vein flow velocities, pulmonary artery systolic pressure, and LA size/volume.

Mitral valveECHO
  • Assess presence and degree of SAM and mitral regurgitation. The presence of a central- or anteriorly directed jet of mitral regurgitation should raise suspicion of an intrinsic/primary mitral valve abnormality and prompt further assessment.

LVOTECHO
LA dimensionsECHO/CMR
  • Provides important prognostic information.365,525,584

  • Most common mechanisms of LA enlargement are SAM-related mitral regurgitation and elevated LV filling pressures.

Myocardial fibrosis/LGECMR
  • The distribution and severity of interstitial expansion can suggest specific diagnoses. Anderson–Fabry disease is characterized by a reduction in non-contrast T1 signal and the presence of posterolateral LGE.134,155 In cardiac amyloidosis, there is often global, subendocardial or segmental LGE and a highly specific pattern of myocardial and blood-pool gadolinium kinetics caused by similar myocardial and blood T1 signals.585,586

2D, two-dimensional; CMR, cardiac magnetic resonance; ECHO, echocardiogram; EF, ejection fraction; LA, left atrium; LGE, late gadolinium enhancement; LV, left ventricular; LVOT, left ventricular outflow tract; SAM, systolic anterior motion; SCD, sudden cardiac death.

Table 18

Echocardiographic features that suggest specific aetiologies in hypertrophic cardiomyopathy

FindingSpecific diseases to be considered
Increased interatrial septum thicknessAmyloidosis
Increased AV valve thicknessAmyloidosis; Anderson–Fabry disease
Increased RV free wall thicknessAmyloidosis, myocarditis, Anderson–Fabry disease, Noonan syndrome, and related disorders
Mild-to-moderate pericardial effusionAmyloidosis, myocarditis/myopericarditis
Ground-glass appearance of ventricular myocardium on 2D echocardiographyAmyloidosis
Concentric LVHGlycogen storage disease, Anderson–Fabry disease, PRKAG2 variants, Friedreich ataxia
Extreme concentric LVH (wall thickness ≥30 mm)Danon disease, Pompe disease
Global LV hypokinesia (with or without LV dilatation)Mitochondrial disease, TTR-related amyloidosis, PRKAG2 variants, Danon disease, myocarditis, advanced sarcomeric HCM, Anderson–Fabry disease, Friedreich ataxia
RVOTONoonan syndrome and associated disorders
Apical sparing pattern on longitudinal strain imagingAmyloidosis
FindingSpecific diseases to be considered
Increased interatrial septum thicknessAmyloidosis
Increased AV valve thicknessAmyloidosis; Anderson–Fabry disease
Increased RV free wall thicknessAmyloidosis, myocarditis, Anderson–Fabry disease, Noonan syndrome, and related disorders
Mild-to-moderate pericardial effusionAmyloidosis, myocarditis/myopericarditis
Ground-glass appearance of ventricular myocardium on 2D echocardiographyAmyloidosis
Concentric LVHGlycogen storage disease, Anderson–Fabry disease, PRKAG2 variants, Friedreich ataxia
Extreme concentric LVH (wall thickness ≥30 mm)Danon disease, Pompe disease
Global LV hypokinesia (with or without LV dilatation)Mitochondrial disease, TTR-related amyloidosis, PRKAG2 variants, Danon disease, myocarditis, advanced sarcomeric HCM, Anderson–Fabry disease, Friedreich ataxia
RVOTONoonan syndrome and associated disorders
Apical sparing pattern on longitudinal strain imagingAmyloidosis

2D, two-dimensional; AV, atrioventricular; HCM, hypertrophic cardiomyopathy; LV, left ventricular; LVH, left ventricular hypertrophy; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; RV, right ventricular; RVOTO, right ventricular outflow tract obstruction; TTR, transthyretin.

Modified from Rapezzi et al.62

Table 18

Echocardiographic features that suggest specific aetiologies in hypertrophic cardiomyopathy

FindingSpecific diseases to be considered
Increased interatrial septum thicknessAmyloidosis
Increased AV valve thicknessAmyloidosis; Anderson–Fabry disease
Increased RV free wall thicknessAmyloidosis, myocarditis, Anderson–Fabry disease, Noonan syndrome, and related disorders
Mild-to-moderate pericardial effusionAmyloidosis, myocarditis/myopericarditis
Ground-glass appearance of ventricular myocardium on 2D echocardiographyAmyloidosis
Concentric LVHGlycogen storage disease, Anderson–Fabry disease, PRKAG2 variants, Friedreich ataxia
Extreme concentric LVH (wall thickness ≥30 mm)Danon disease, Pompe disease
Global LV hypokinesia (with or without LV dilatation)Mitochondrial disease, TTR-related amyloidosis, PRKAG2 variants, Danon disease, myocarditis, advanced sarcomeric HCM, Anderson–Fabry disease, Friedreich ataxia
RVOTONoonan syndrome and associated disorders
Apical sparing pattern on longitudinal strain imagingAmyloidosis
FindingSpecific diseases to be considered
Increased interatrial septum thicknessAmyloidosis
Increased AV valve thicknessAmyloidosis; Anderson–Fabry disease
Increased RV free wall thicknessAmyloidosis, myocarditis, Anderson–Fabry disease, Noonan syndrome, and related disorders
Mild-to-moderate pericardial effusionAmyloidosis, myocarditis/myopericarditis
Ground-glass appearance of ventricular myocardium on 2D echocardiographyAmyloidosis
Concentric LVHGlycogen storage disease, Anderson–Fabry disease, PRKAG2 variants, Friedreich ataxia
Extreme concentric LVH (wall thickness ≥30 mm)Danon disease, Pompe disease
Global LV hypokinesia (with or without LV dilatation)Mitochondrial disease, TTR-related amyloidosis, PRKAG2 variants, Danon disease, myocarditis, advanced sarcomeric HCM, Anderson–Fabry disease, Friedreich ataxia
RVOTONoonan syndrome and associated disorders
Apical sparing pattern on longitudinal strain imagingAmyloidosis

2D, two-dimensional; AV, atrioventricular; HCM, hypertrophic cardiomyopathy; LV, left ventricular; LVH, left ventricular hypertrophy; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; RV, right ventricular; RVOTO, right ventricular outflow tract obstruction; TTR, transthyretin.

Modified from Rapezzi et al.62

Identification of LVOTO is important in the management of symptoms and assessment of SCD risk (see Section 7.1.5). Two-dimensional and Doppler echocardiography during a Valsalva manoeuvre in the sitting and semi-supine positionand then on standing if no gradient is provokedis recommended in all patients (Figure 12).587,588 Exercise stress echocardiography is recommended in symptomatic patients if bedside manoeuvres fail to induce LVOTO ≥50 mmHg. Pharmacological provocation with dobutamine is not advised, as it is not physiological and can be poorly tolerated.

Protocol for the assessment and treatment of left ventricular outflow tract obstruction.
Figure 12

Protocol for the assessment and treatment of left ventricular outflow tract obstruction.

2D, two-dimensional; LVOTO, left ventricular outflow tract obstruction. aExercise echocardiography may be considered in individual patients when the presence of an left ventricular outflow tract gradient is relevant to lifestyle advice and decisions on medical treatment.

Recommendation Table 16

Recommendation for evaluation of left ventricular outflow tract obstruction

graphic
graphic
graphic
graphic
Recommendation Table 16

Recommendation for evaluation of left ventricular outflow tract obstruction

graphic
graphic
graphic
graphic
7.1.1.4. Cardiac magnetic resonance

Cardiac magnetic resonance is recommended in patients with HCM at their baseline assessment (general recommendations are described in Section 6.7.3 and Recommendation Table 5). CMR imaging can be particularly helpful in patients with suspected apical or lateral wall hypertrophy or LV apical aneurysm. Table 17 summarizes the main features to be assessed.

Late gadolinium enhancement is present in 65% of patients (range 33–84%), typically in a patchy mid-wall pattern in areas of hypertrophy and at the anterior and posterior RV insertion points.604 Late gadolinium enhancement is unusual in non-hypertrophied segments except in advanced stages of disease, when full-thickness LGE in association with wall thinning is common.604 Late gadolinium enhancement may be associated with increased myocardial stiffness and adverse LV remodelling and the extent of LGE is associated with a higher incidence of RWMAs. Late gadolinium enhancement varies substantially with the quantification method used but the 2-standard deviation technique is the only one validated against necropsy.605

Although CMR rarely distinguishes the causes of HCM by their magnetic properties alone, the distribution and severity of interstitial expansion can, in context, suggest specific diagnoses (see Section 6). The absence of fibrosis may be helpful in differentiating HCM from physiological adaptation in athletes, but LGE may be absent in people with HCM, particularly young people and those with mild disease.

Recommendation Table 17

Additional recommendation for cardiovascular magnetic resonance evaluation in hypertrophic cardiomyopathy

graphic
graphic
Recommendation Table 17

Additional recommendation for cardiovascular magnetic resonance evaluation in hypertrophic cardiomyopathy

graphic
graphic
7.1.1.5. Nuclear imaging

The major clinical contribution of nuclear imaging in HCM is the detection of TTR-related cardiac amyloidosis (see Section 7.7). Recommendations on the utility of bone scintigraphy and cardiac CT are described in Section 6.7.4.

7.1.2. Genetic testing and family screening

In about half of cases, HCM is inherited as a Mendelian genetic trait. In such cases, the inheritance is primarily autosomal dominant, i.e. with a 50% risk of transmission to offspring.608 Apparently sporadic cases can have a monogenic cause, either because of incomplete penetrance of a variant inherited from a parent or due to de novo variants that were not carried by the parents or, less commonly, due to autosomal recessive inheritance. In those who undergo genetic testing, ∼40–60% will have a single variant identified as the cause of their disease, although this is influenced by the cohort studied.124 The likelihood of finding a causal variant is highest in young patients with familial disease and lowest in older patients and individuals with non-classical features. Phenotype-based scores to predict genetic yield in HCM have been developed and may be used to prioritize genetic testing where resources are limited.609,610 Genes with definitive evidence for gene–disease association with HCM are summarized in Table 10. An important subgroup characterized by no identifiable monogenic variant, no family history of disease and often being older, more likely to be male and with a history of hypertension, and less risk of major cardiovascular events is likely to be underlied by complex aetiology.238,611,612

Less than 5% of adult patients, but up to 25% of children, with HCM, will have a causative variant in a gene that is known to mimic the HCM phenotype. Such genocopies can have clinically important differences such as altered inheritance risks, and management and therapy options. The aetiology of HCM in childhood is more heterogeneous than that seen in adult populations, and includes inborn errors of metabolism, malformation syndromes, and neuromuscular disorders.613–615 Most cases of HCM in childhood, however, are caused by variants in the cardiac sarcomere protein genes, inherited as autosomal dominant traits.616,617 The relative prevalence of different HCM aetiologies varies according to age: HCM related to inborn errors of metabolism and malformation syndromes is most commonly diagnosed in the first 2 years of life, whereas HCM due to neuromuscular disorders (e.g. Friedreich ataxia) most commonly presents in adolescence.613–615 Outside of infancy, sarcomere protein gene variants account for 55–75% of cases of childhood-onset HCM,616–619 and even in infancy, sarcomeric disease is present in up to 40% of cases.616,620 Although rarer, inborn errors of metabolism and malformation syndromes can also present for the first time in older children and adolescents (see Section 7.6).614

A thorough and comprehensive diagnostic work-up is essential in the diagnosis of childhood-onset HCM in order to confirm the diagnosis, identify the underlying aetiology, and guide treatment (see Section 6).

Recommendations for clinical screening, genetic counselling, and testing are described in Sections 6.8.3 and 6.11, respectively.

7.1.3. Assessment of symptoms

Most people with HCM are asymptomatic and have a normal lifespan, but some develop symptoms, often many years after the appearance of ECG or echocardiographic evidence of LVH. Assessment of symptoms in patients with cardiomyopathies is described in Section 6.4. Assessment of LVOTO, as outlined in Figure 12, should be part of the routine evaluation of all symptomatic patients.

7.1.4. Management of symptoms and complications

In the absence of many randomized trials,621–623 pharmacological therapy is mostly administered on an empirical basis to improve functional capacity and reduce symptoms. In symptomatic patients with LVOTO, the aim is to improve symptoms by using drugs, surgery, or alcohol septal ablation. Therapy in symptomatic patients without LVOTO focuses on management of arrhythmia, reduction of LV filling pressures, and treatment of angina. Patients with progressive LV systolic or diastolic dysfunction refractory to medical therapy may be candidates for cardiac transplantation (Figure 13).

Algorithm for the treatment of heart failure in hypertrophic cardiomyopathy.
Figure 13

Algorithm for the treatment of heart failure in hypertrophic cardiomyopathy.

ACEi, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; ARNI, angiotensin receptor neprilysin inhibitor; LVEF, left ventricular ejection fraction; LVOTO, left ventricular outflow tract obstruction; MCS, mechanical circulatory support; MRA, mineralocorticoid receptor antagonist; NYHA, New York Heart Association; SGLT2i, sodium–glucose co-transporter 2 inhibitor.

7.1.4.1. Management of left ventricular outflow tract obstruction

By convention, LVOTO is defined as a peak instantaneous Doppler LV outflow tract gradient of ≥30 mmHg, but the threshold for invasive treatment is usually considered to be ≥50 mmHg (the threshold at which theoretical models examining the relationship between the gradient and stroke volume predict that this becomes haemodynamically significant).587,624,625 Most patients with a maximum resting or provoked LV outflow tract gradient <50 mmHg should be managed in accordance with the recommendations for non-obstructive HCM but, in a very small number of selected cases with LV outflow tract gradients between 30 and 50 mmHg and no other obvious cause of symptoms, invasive gradient reduction may be considered, acknowledging that data covering this group are lacking. Most asymptomatic patients with LVOTO do not require treatment but, in a very small number of selected cases, pharmacological treatment to reduced LV pressures may be considered.626,627

General measures

All patients with LVOTO should avoid dehydration and excess alcohol consumption, and weight loss should be encouraged. Arterial and venous dilators, including nitrates and phosphodiesterase type 5 inhibitors, can exacerbate LVOTO and should be avoided if possible (see Section 12.2).626 New-onset or poorly controlled AF can exacerbate symptoms caused by LVOTO and should be managed by prompt restoration of sinus rhythm or ventricular rate control.628

Recommendation Table 18

Recommendations for treatment of left ventricular outflow tract obstruction (general measures)

graphic
graphic
Recommendation Table 18

Recommendations for treatment of left ventricular outflow tract obstruction (general measures)

graphic
graphic
Drug therapy

Figure 14 describes the management of LVOTO in patients with HCM. By consensus, patients with symptomatic LVOTO have been treated initially with non-vasodilating beta-blockers titrated to the maximum tolerated dose, but there are very few studies comparing individual beta-blockers. A recent small, randomized placebo-controlled trial showed reduction of resting and exertional LVOTO, and improvement in symptoms and QoL with metoprolol therapy.631

Flow chart on the management of left ventricular outflow tract obstruction.
Figure 14

Flow chart on the management of left ventricular outflow tract obstruction.

LVOTO, left ventricular outflow tract obstruction.

If beta-blockers alone are ineffective, disopyramide, titrated up to a maximum tolerated dose (usually 400–600 mg/day), may be added.632–634 This class IA AAD can abolish basal LV outflow pressure gradients and improve exercise tolerance and functional capacity with a low risk of proarrhythmic effects and without an increased risk of SCD.632,633 Dose-limiting anticholinergic side effects include dry eyes and mouth, urinary hesitancy or retention, and constipation.632,633,635 The QTc interval should be monitored during dose up-titration and the dose reduced if it exceeds 500 ms. Disopyramide should be avoided in patients with glaucoma, in men with prostatism, and in patients taking other drugs that prolong the QT interval, such as amiodarone and sotalol. Disopyramide may be used in combination with verapamil.633

Verapamil (starting dose 40 mg three times daily to maximum 480 mg daily) can be used when beta-blockers are contraindicated or ineffective but, based on limited data, should be used cautiously in patients with severe obstruction (≥100 mmHg) or elevated pulmonary artery systolic pressures, as it may provoke pulmonary oedema.636 Short-term oral administration may increase exercise capacity, improve symptoms, and normalize or improve LV diastolic filling without altering systolic function.637–640 Similar findings have been demonstrated for diltiazem (starting dose 60 mg three times daily to maximum 360 mg daily),641 and it should be considered in patients who are intolerant or have contraindications to beta-blockers and verapamil.

Low-dose loop or thiazide diuretics may be used cautiously to improve dyspnoea associated with LVOTO, but it is important to avoid hypovolaemia.

Cardiac myosin ATPase inhibitors. Mavacamten is a first-in-class cardiac myosin adenosine triphosphatase (ATPase) inhibitor that acts by reducing actin–myosin cross-bridge formation, thereby reducing contractility and improving myocardial energetics. In the recently published Clinical Study to Evaluate Mavacamten in Adults with Symptomatic Obstructive Hypertrophic Cardiomyopathy (EXPLORER-HCM) trial, mavacamten reduced the left ventricular outflow tract (LVOT) gradient and improved exercise capacity compared with placebo in patients with HCM and symptomatic LVOTO (NYHA II–III and EF >55%); 27% of patients on mavacamten had an LVOT gradient reduction to <30 mmHg and improved to NYHA class I.622 The drug was well tolerated and has a good safety profile; only a small subset of patients developed transient LV systolic dysfunction, which resolved after temporary discontinuation of the drug. A second study (A Study to Evaluate Mavacamten in Adults With Symptomatic Obstructive HCM Who Are Eligible for Septal Reduction Therapy [VALOR-HCM]) in adult patients with obstructive HCM referred for septal reduction therapy (SRT) due to intractable symptoms showed that mavacamten significantly reduced the proportion of patients meeting criteria for SRT at 16 and 32 weeks.642,643 Small CMR and ECHO substudies suggest that mavacamten may also lead to positive myocardial remodelling, with reduction in myocardial mass, LV wall thickness, and left atrial volume.644–646 Aficamten, a next-in-class cardiac myosin inhibitor, was also recently shown in a Phase II randomized placebo-controlled study (Randomized Evaluation of Dosing With CK-3773274 in Obstructive Outflow Disease in HCM [REDWOOD-HCM]) to significantly reduce LVOT gradients and NT-proBNP levels in adult patients with symptomatic obstructive HCM.647

In the absence of a direct head-to-head comparison, the Task Force was unable to recommend the use of cardiac myosin ATPase inhibitors as first-line medical therapy, but did consider the evidence sufficiently robust to support the recommendation that their use as second-line therapy should be considered when optimal medical therapy with beta-blockers, calcium antagonists, and/or disopyramide is ineffective or poorly tolerated. In the absence of evidence to the contrary, cardiac myosin ATPase inhibitors should not be used with disopyramide, but may be coadministered with beta-blockers or calcium antagonists. Up-titration of medication to a maximum dose of 15 mg should be monitored in accordance with licensed recommendations using echocardiography. In patients with contraindications or known sensitivity to beta-blockers, calcium antagonists, and disopyramide, cardiac myosin ATPase inhibitors may be considered as monotherapy.

Recommendation Table 19

Recommendations for medical treatment of left ventricular outflow tract obstruction

graphic
graphic
graphic
graphic
Recommendation Table 19

Recommendations for medical treatment of left ventricular outflow tract obstruction

graphic
graphic
graphic
graphic
Invasive treatment of left ventricular outflow tract (septal reduction therapy)

There are no data to support the use of invasive procedures to reduce LVOTO in asymptomatic patients, regardless of its severity. However, some retrospective data suggest that individuals with high LVOT gradients, even if minimally symptomatic, have a higher mortality than those without markedly elevated gradients.651 Delay in SRT may have an impact on long-term outcomes, particularly when >5 years from first detection of gradient, even when successful relief of symptoms and gradient is achieved. Earlier interventions may be associated with lower complication rates and better prognosis.652

Invasive treatment (SRT) to reduce LVOTO should be considered in patients with a LVOTO gradient ≥50 mmHg, severe symptoms (NYHA functional class III–IV), and/or exertional or unexplained recurrent syncope in spite of maximally tolerated drug therapy. Invasive therapy may also be considered in patients with mild symptoms (NYHA class II) refractory to medical therapy who have a resting or maximum provoked gradient of ≥50 mmHg (exercise or Valsalva) and moderate-to-severe systolic anterior motion-related mitral regurgitation, AF, or moderate-to-severe left atrial dilatation in expert centres with demonstrable low procedural complication rates.653

Surgery. The most commonly performed surgical procedure to treat LVOTO is ventricular septal myectomy, in which a rectangular trough that extends distally to beyond the point of the mitral leaflet–septal contact is created in the basal septum below the aortic valve.654 This abolishes or substantially reduces LV outflow tract gradients in over 90% of cases, reduces systolic anterior motion-related mitral regurgitation, and improves exercise capacity and symptoms. Long-term symptomatic benefit is achieved in >80% of patients with a long-term survival comparable to that of the general population.655–665 Pre-operative determinants of a good long-term outcome are age <50 years, left atrial size <46 mm, absence of AF, and male sex.663

The main surgical complications are AV nodal block, left bundle branch block (LBBB), ventricular septal defect, and aortic regurgitation, but these are uncommon (except LBBB) in experienced centres using intra-operative transoesophageal echocardiography guidance.662,666,667 When there is coexisting mid-cavity obstruction, the standard myectomy can be extended distally into the mid-ventricle around the base of the papillary muscles; however, data on the efficacy and long-term outcomes of this approach are limited.668

In patients with intrinsic/primary mitral valve disease or marked mitral leaflet elongation and/or moderate-to-severe mitral regurgitation, septal myectomy can be combined with mitral valve repair or replacement.669–675 In patients with AF, concomitant ablation using the Cox–Maze procedure can also be performed.676 In infants and very young children, the modified Konno procedure may be an alternative to myectomy when the aortic annulus is too small.677

Alcohol septal ablation (ASA). In experienced centres, selective injection of alcohol into a septal perforator artery to create a localized septal scar has outcomes similar to surgery in terms of gradient reduction, symptom improvement, and exercise capacity, including in younger adults.678–685 In many centres, ASA has become the primary SRT modality. The main non-fatal complication is AV block in 7–20% of patients, and the procedural mortality is lower than isolated myectomy.679–683,686,687

Due to the variability of the septal blood supply, myocardial contrast echocardiography is essential prior to alcohol injection. Injection of large volumes of alcohol in multiple septal branches—with the aim of gradient reduction—in the catheter laboratory is generally not recommended, as it can be associated with a high risk of complications and arrhythmic events.688

Alternative methods have been reported in small numbers of patients, including non-ASA techniques (coils,689,690 polyvinyl alcohol foam particles,691 cyanoacrylate692) and direct endocavitary and intramuscular ablation (radiofrequency, cryotherapy).693,694 These alternative methods have not been directly compared with other septal reduction therapies and long-term outcome/safety data are not available. Alcohol septal ablation and alternative methods should not be used in children with HCM outside experimental settings, due to a lack of medium- to long-term safety and efficacy data.

Surgery vs. alcohol septal ablation. Because of specific anatomic features of the LVOT and the mitral valve, some patients with HCM will be more suitable candidates for septal myectomy than ASA. Experienced multidisciplinary teams should assess all patients before intervention, as morbidity and mortality are highly dependent on the available level of expertise (see Section 9).687,695,696 A summary of the key points in pre-operative assessment is shown in Figure 15.

Pre-assessment checklist for patients being considered for invasive septal reduction therapies.
Figure 15

Pre-assessment checklist for patients being considered for invasive septal reduction therapies.

AF, atrial fibrillation; MV, mitral valve; RVOTO, right ventricular outflow tract obstruction; SAM, systolic anterior motion.

There are no randomized trials comparing surgery and ASA, but several meta-analyses have shown that both procedures improve functional status with a similar procedural mortality.697–703 Alcohol septal ablation is associated with a higher risk of AV block, requiring permanent pacemaker implantation, and larger residual LV outflow tract gradients.697–702 The risk of AV block following surgery and ASA is highest in patients with pre-existing conduction disease, and prophylactic permanent pacing before intervention has been advocated,704 although recent data suggest that the long-term outcome of patients after ASA with implanted permanent pacemaker is similar to those without pacemaker.705 Repeat ASA or myectomy procedure is reported in 7–20% of patients after ASA, which is higher than reported following surgical myectomy.702 Septal ablation may be less effective in patients with very severe hypertrophy (≥30 mm), but systematic data are limited.706 In general, the risk of ventricular septal defect following septal myectomy is very small and could be higher in patients with mild hypertrophy (≤16 mm) at the point of the mitral leaflet–septal contact. This risk is exceedingly rare with ASA, but alternatives such as dual-chamber pacing or mitral valve repair/replacement may also be considered in such cases.707

Recommendation Table 20

Recommendations for septal reduction therapy

graphic
graphic
graphic
graphic
Recommendation Table 20

Recommendations for septal reduction therapy

graphic
graphic
graphic
graphic

Dual-chamber pacing. Three small, randomized, placebo-controlled studies of dual-chamber pacing and several long-term observational studies have reported reductions in LV outflow tract gradients and variable improvement in symptoms and QoL, including one paediatric study.719–724 A Cochrane review concluded that the data on the benefits of pacing are based on physiological measures and lack information on clinically relevant endpoints.725

Recommendation Table 21

Recommendations for indications for cardiac pacing in patients with obstruction

graphic
graphic
Recommendation Table 21

Recommendations for indications for cardiac pacing in patients with obstruction

graphic
graphic

Left ventricular mid-cavity obstruction and apical aneurysms. Left ventricular mid-cavity obstruction occurs in ∼10% of patients with HCM.727,728 Patients with mid-cavity obstruction tend to be very symptomatic and, in a number of studies, have shown an increased risk of progressive heart failure and SCD.727–729 Approximately 25% of patients also have an LV apical aneurysm (see Section 7.1.5).580,727,728,730 Patients with LV mid-cavity obstruction should be treated with high-dose beta-blockers, verapamil, or diltiazem, but the response is often suboptimal. Limited experience, mostly from single centres, suggests that mid-ventricular obstruction can be relieved by transaortic myectomy, a transapical approach, or combined transaortic and transapical incisions, with good short-term outcomes but uncertain long-term survival.731,732

Left ventricular apical aneurysms by themselves rarely need treatment. A few patients develop monomorphic ventricular tachycardia related to adjacent apical scarring, which may be amenable to mapping and ablation (see Section 7.1.5).730,733 Rarely, thrombi are present within the aneurysm and should be treated with long-term oral anticoagulation.734,735 Anticoagulation may also be considered in patients with HCM and apical aneurysms in the absence of documented thrombi.736,737

7.1.4.2. Management of symptoms in patients without left ventricular outflow tract obstruction
Heart failure and chest pain

Management of heart failure in patients without LVOTO should follow the recommendations of the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure, summarized in Section 6.10.2. The aim of drug therapy is to reduce LV diastolic pressures and improve LV filling by slowing the heart rate with beta-blockers, verapamil, or diltiazem (ideally monitored by ambulatory ECG recording), and cautious use of loop diuretics. Beta-blockers or calcium antagonists should be considered in patients with exertional or prolonged episodes of angina-like pain even in the absence of resting or provocable LVOTO or obstructive CAD. In the absence of LVOTO, cautious use of oral nitrates may be considered. Ranolazine may also be considered to improve symptoms in patients with angina-like chest pain and no evidence for LVOTO.738,739

Recommendation Table 22

Recommendations for chest pain on exertion in patients without left ventricular outflow tract obstruction

graphic
graphic
Recommendation Table 22

Recommendations for chest pain on exertion in patients without left ventricular outflow tract obstruction

graphic
graphic
Cardiac resynchronization therapy

Regional heterogeneity of LV contraction and relaxation can be seen in patients with HCM, and LV dyssynchrony may be a marker of poor prognosis.745 Data on the impact of CRT on symptoms, LV function, and prognosis in patients with non-obstructive HCM remain limited, but new evidence has emerged since the 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy.746,747 There is one small study using a blinded crossover design of biventricular and sham pacing and a pre-specified analysis stratified by changes in LV end-diastolic volume (LVEDV) with exercise at baseline.748 Biventricular pacing was associated with significant increases in LVEDV and stroke volume in patients who had a reduction in exercise LVEDV pre-pacing (consistent with the relief of diastolic ventricular interaction). This translated into improvements in peak maximum oxygen consumption (VO2) (1.4 mL/kg/min) and QoL scores.748 Together, they suggest that symptomatic responses to CRT may occur in individual patients, but that these are not associated with consistent changes in LVEF or evidence for a reduction in progression to end-stage heart failure.

The 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy recommend that standard criteria for CRT are used in patients with HCM.724 The Task Force considered these of limited utility in HCM, as the unique pathology of this disease means that patients with contractile impairment rarely have an LVEF ≤35%. While acknowledging this as an area of unmet research need, the Task Force suggests a more pragmatic approach in which CRT might be considered in individual symptomatic patients with LV impairment (LVEF <50%) that meet current ESC ECG criteria (LBBB, QRS 130–149 ms). Cardiac resynchronization therapy might also be considered in patients with HCM and impaired systolic function who require permanent ventricular pacing.746 In keeping with the 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy, the Task Force did not include these as specific recommendations, given the limited evidence base.

7.1.5. Sudden cardiac death prevention in hypertrophic cardiomyopathy

Most contemporary series of adult patients with HCM report an annual incidence for cardiovascular death of 1–2%, with SCD, heart failure, and thrombo-embolism being the main causes of death.749 The most commonly recorded fatal arrhythmic event is spontaneous ventricular fibrillation (VF), but asystole, AV block, and pulseless electrical activity are described.532,750–754 In children with HCM, although initial studies, usually from small, highly selected cohorts, reported SCD rates of up to 10% per year,755–757 more recent, larger, population-based studies have shown SCD rates in the region of 1.2–1.5% per year.81,535,758 While much lower than previously thought, this is still >50% higher than reported in adult HCM populations. Sudden cardiac death appears to be very rare below the age of 6 years.81,759

Estimation of SCD risk is an integral part of clinical management. Clinical features that are associated with an increased SCD risk and that have been used in previous guidelines to estimate risk are shown in Table 19.

Table 19

Major clinical features associated with an increased risk of sudden cardiac death

Risk factorComment
Age
  • The effect of age on SCD has been examined in a number of studies86,525,584,760–764 and two have shown a significant association, with an increased risk of SCD in younger patients.525,584

  • Some risk factors appear to be more important in younger patients, most notably NSVT,765 severe LVH,766 and unexplained syncope.584

  • Sudden cardiac death is very rare below the age of 6 years,535,767 and there are some data to suggest a peak of SCD in childhood HCM between 9 and 15 years;757 however, the association between age at diagnosis and SCD risk in childhood HCM remains unclear.

NSVT
  • NSVT (defined as ≥3 consecutive ventricular beats at ≥120 b.p.m. lasting <30 s) occurs in 20–30% of patients during ambulatory ECG monitoring and is an independent predictor of SCD.81,525,535,590,764,765,768–773

  • There is no evidence that the frequency, duration, or rate of NSVT influences the risk of SCD.765,774

  • NSVT occurring during or immediately following exercise is very rare, but may be associated with a high risk of SCD.768

Maximum LV wall thickness
  • The severity and extent of LVH measured by TTE are associated with the risk of SCD.81,535,592,593,763,765,770–772,775–780

  • Several studies have shown the greatest risk of SCD in patients with a maximum wall thickness of ≥30 mm; however, there are few data in patients with extreme hypertrophy (≥35 mm).525,592,763,765,769,781–784

Family history of sudden cardiac death at a young age
  • While definitions vary,525,592,762,782 a family history of SCD is usually considered clinically significant when one or more first-degree relatives have died suddenly aged <40 years with or without a diagnosis of HCM, or when SCD has occurred in a first-degree relative at any age with an established diagnosis of HCM.

  • Family history of SCD does not appear to be an independent risk factor for SCD in childhood HCM.81,535 This may be due to a higher prevalence of de novo variants in childhood HCM, the inclusion of non-sarcomeric disease, and/or under-reporting of family history in paediatric cohorts.

Syncope
  • Syncope is common in patients with HCM but is challenging to assess, as it has multiple causes.785

  • Non-neurocardiogenic syncope for which there is no explanation after investigation is associated with an increased risk of SCD.81,525,535,584,590,755,761,768,769,781,786–788

  • Episodes within 6 months of evaluation may be more predictive of SCD.584

Left atrial diameter
  • Several studies have reported a positive association between LA size and SCD.81,525,535,584,772,789 There are no data on the association between SCD and LA area or volume. Measurement of LA size is also important in assessing the risk of AF (see Section 6.10.3).

LV outflow tract obstruction
  • A number of studies have reported a significant association between LVOTO and SCD risk.86,525,590,762,768,790 Several unanswered questions remain, including the prognostic importance of provocable LVOTO and the impact of treatment (medical or invasive) on SCD.

  • In childhood HCM, there are conflicting data on the association between LVOTO and SCD risk.81,535,772,777

Risk factorComment
Age
  • The effect of age on SCD has been examined in a number of studies86,525,584,760–764 and two have shown a significant association, with an increased risk of SCD in younger patients.525,584

  • Some risk factors appear to be more important in younger patients, most notably NSVT,765 severe LVH,766 and unexplained syncope.584

  • Sudden cardiac death is very rare below the age of 6 years,535,767 and there are some data to suggest a peak of SCD in childhood HCM between 9 and 15 years;757 however, the association between age at diagnosis and SCD risk in childhood HCM remains unclear.

NSVT
  • NSVT (defined as ≥3 consecutive ventricular beats at ≥120 b.p.m. lasting <30 s) occurs in 20–30% of patients during ambulatory ECG monitoring and is an independent predictor of SCD.81,525,535,590,764,765,768–773

  • There is no evidence that the frequency, duration, or rate of NSVT influences the risk of SCD.765,774

  • NSVT occurring during or immediately following exercise is very rare, but may be associated with a high risk of SCD.768

Maximum LV wall thickness
  • The severity and extent of LVH measured by TTE are associated with the risk of SCD.81,535,592,593,763,765,770–772,775–780

  • Several studies have shown the greatest risk of SCD in patients with a maximum wall thickness of ≥30 mm; however, there are few data in patients with extreme hypertrophy (≥35 mm).525,592,763,765,769,781–784

Family history of sudden cardiac death at a young age
  • While definitions vary,525,592,762,782 a family history of SCD is usually considered clinically significant when one or more first-degree relatives have died suddenly aged <40 years with or without a diagnosis of HCM, or when SCD has occurred in a first-degree relative at any age with an established diagnosis of HCM.

  • Family history of SCD does not appear to be an independent risk factor for SCD in childhood HCM.81,535 This may be due to a higher prevalence of de novo variants in childhood HCM, the inclusion of non-sarcomeric disease, and/or under-reporting of family history in paediatric cohorts.

Syncope
  • Syncope is common in patients with HCM but is challenging to assess, as it has multiple causes.785

  • Non-neurocardiogenic syncope for which there is no explanation after investigation is associated with an increased risk of SCD.81,525,535,584,590,755,761,768,769,781,786–788

  • Episodes within 6 months of evaluation may be more predictive of SCD.584

Left atrial diameter
  • Several studies have reported a positive association between LA size and SCD.81,525,535,584,772,789 There are no data on the association between SCD and LA area or volume. Measurement of LA size is also important in assessing the risk of AF (see Section 6.10.3).

LV outflow tract obstruction
  • A number of studies have reported a significant association between LVOTO and SCD risk.86,525,590,762,768,790 Several unanswered questions remain, including the prognostic importance of provocable LVOTO and the impact of treatment (medical or invasive) on SCD.

  • In childhood HCM, there are conflicting data on the association between LVOTO and SCD risk.81,535,772,777

AF, atrial fibrillation; b.p.m., beats per minute; ECG, electrocardiogram; HCM, hypertrophic cardiomyopathy; LA, left atrium; LV, left ventricular; LVH, left ventricular hypertrophy; LVOTO, left ventricular outflow tract obstruction; NSVT, non-sustained ventricular tachycardia; SCD, sudden cardiac death; TTE, transthoracic echocardiogram.

Table 19

Major clinical features associated with an increased risk of sudden cardiac death

Risk factorComment
Age
  • The effect of age on SCD has been examined in a number of studies86,525,584,760–764 and two have shown a significant association, with an increased risk of SCD in younger patients.525,584

  • Some risk factors appear to be more important in younger patients, most notably NSVT,765 severe LVH,766 and unexplained syncope.584

  • Sudden cardiac death is very rare below the age of 6 years,535,767 and there are some data to suggest a peak of SCD in childhood HCM between 9 and 15 years;757 however, the association between age at diagnosis and SCD risk in childhood HCM remains unclear.

NSVT
  • NSVT (defined as ≥3 consecutive ventricular beats at ≥120 b.p.m. lasting <30 s) occurs in 20–30% of patients during ambulatory ECG monitoring and is an independent predictor of SCD.81,525,535,590,764,765,768–773

  • There is no evidence that the frequency, duration, or rate of NSVT influences the risk of SCD.765,774

  • NSVT occurring during or immediately following exercise is very rare, but may be associated with a high risk of SCD.768

Maximum LV wall thickness
  • The severity and extent of LVH measured by TTE are associated with the risk of SCD.81,535,592,593,763,765,770–772,775–780

  • Several studies have shown the greatest risk of SCD in patients with a maximum wall thickness of ≥30 mm; however, there are few data in patients with extreme hypertrophy (≥35 mm).525,592,763,765,769,781–784

Family history of sudden cardiac death at a young age
  • While definitions vary,525,592,762,782 a family history of SCD is usually considered clinically significant when one or more first-degree relatives have died suddenly aged <40 years with or without a diagnosis of HCM, or when SCD has occurred in a first-degree relative at any age with an established diagnosis of HCM.

  • Family history of SCD does not appear to be an independent risk factor for SCD in childhood HCM.81,535 This may be due to a higher prevalence of de novo variants in childhood HCM, the inclusion of non-sarcomeric disease, and/or under-reporting of family history in paediatric cohorts.

Syncope
  • Syncope is common in patients with HCM but is challenging to assess, as it has multiple causes.785

  • Non-neurocardiogenic syncope for which there is no explanation after investigation is associated with an increased risk of SCD.81,525,535,584,590,755,761,768,769,781,786–788

  • Episodes within 6 months of evaluation may be more predictive of SCD.584

Left atrial diameter
  • Several studies have reported a positive association between LA size and SCD.81,525,535,584,772,789 There are no data on the association between SCD and LA area or volume. Measurement of LA size is also important in assessing the risk of AF (see Section 6.10.3).

LV outflow tract obstruction
  • A number of studies have reported a significant association between LVOTO and SCD risk.86,525,590,762,768,790 Several unanswered questions remain, including the prognostic importance of provocable LVOTO and the impact of treatment (medical or invasive) on SCD.

  • In childhood HCM, there are conflicting data on the association between LVOTO and SCD risk.81,535,772,777

Risk factorComment
Age
  • The effect of age on SCD has been examined in a number of studies86,525,584,760–764 and two have shown a significant association, with an increased risk of SCD in younger patients.525,584

  • Some risk factors appear to be more important in younger patients, most notably NSVT,765 severe LVH,766 and unexplained syncope.584

  • Sudden cardiac death is very rare below the age of 6 years,535,767 and there are some data to suggest a peak of SCD in childhood HCM between 9 and 15 years;757 however, the association between age at diagnosis and SCD risk in childhood HCM remains unclear.

NSVT
  • NSVT (defined as ≥3 consecutive ventricular beats at ≥120 b.p.m. lasting <30 s) occurs in 20–30% of patients during ambulatory ECG monitoring and is an independent predictor of SCD.81,525,535,590,764,765,768–773

  • There is no evidence that the frequency, duration, or rate of NSVT influences the risk of SCD.765,774

  • NSVT occurring during or immediately following exercise is very rare, but may be associated with a high risk of SCD.768

Maximum LV wall thickness
  • The severity and extent of LVH measured by TTE are associated with the risk of SCD.81,535,592,593,763,765,770–772,775–780

  • Several studies have shown the greatest risk of SCD in patients with a maximum wall thickness of ≥30 mm; however, there are few data in patients with extreme hypertrophy (≥35 mm).525,592,763,765,769,781–784

Family history of sudden cardiac death at a young age
  • While definitions vary,525,592,762,782 a family history of SCD is usually considered clinically significant when one or more first-degree relatives have died suddenly aged <40 years with or without a diagnosis of HCM, or when SCD has occurred in a first-degree relative at any age with an established diagnosis of HCM.

  • Family history of SCD does not appear to be an independent risk factor for SCD in childhood HCM.81,535 This may be due to a higher prevalence of de novo variants in childhood HCM, the inclusion of non-sarcomeric disease, and/or under-reporting of family history in paediatric cohorts.

Syncope
  • Syncope is common in patients with HCM but is challenging to assess, as it has multiple causes.785

  • Non-neurocardiogenic syncope for which there is no explanation after investigation is associated with an increased risk of SCD.81,525,535,584,590,755,761,768,769,781,786–788

  • Episodes within 6 months of evaluation may be more predictive of SCD.584

Left atrial diameter
  • Several studies have reported a positive association between LA size and SCD.81,525,535,584,772,789 There are no data on the association between SCD and LA area or volume. Measurement of LA size is also important in assessing the risk of AF (see Section 6.10.3).

LV outflow tract obstruction
  • A number of studies have reported a significant association between LVOTO and SCD risk.86,525,590,762,768,790 Several unanswered questions remain, including the prognostic importance of provocable LVOTO and the impact of treatment (medical or invasive) on SCD.

  • In childhood HCM, there are conflicting data on the association between LVOTO and SCD risk.81,535,772,777

AF, atrial fibrillation; b.p.m., beats per minute; ECG, electrocardiogram; HCM, hypertrophic cardiomyopathy; LA, left atrium; LV, left ventricular; LVH, left ventricular hypertrophy; LVOTO, left ventricular outflow tract obstruction; NSVT, non-sustained ventricular tachycardia; SCD, sudden cardiac death; TTE, transthoracic echocardiogram.

7.1.5.1. Left ventricular apical aneurysms

Left ventricular apical aneurysms are defined as a discrete thin-walled dyskinetic or akinetic segment of the most distal portion of the left ventricle and are often associated with a mid-cavity gradient. Their prevalence in unselected patients is uncertain but they were reported in 3% of individuals in the prospective Hypertrophic Cardiomyopathy Registry (HCMR).124 The first descriptions of LV apical aneurysms in HCM suggested an association with sustained monomorphic ventricular tachycardia (SMVT)730a relatively rare occurrence in HCMand a number of studies have suggested that they are a useful marker of SCD risk.580,728,736,737,791,792 Based on these data, LV aneurysms were included in the recent 2020 American Heart Association/American College of Cardiology (AHA/ACC) HCM guideline as a major independent SCD risk factor and were considered a reasonable sole indication for an ICD.793 In a review for this guideline, the data from two published studies and a meta-analysis were evaluated (see Supplementary data online, Table S2). All these studies were retrospective and the absolute number of events is too small to assess the independent predictive value of apical aneurysms. In two small series that described a selected subgroup of HCM patients with mid-ventricular obstruction, there was no increase in incidence of SCD events. In the only series that provides a detailed analysis of SCD events, the majority were appropriate ICD interventions for monomorphic VT, suggesting significant inclusion bias.737 Finally, a large proportion of individuals with events had other important risk markers including prior sustained ventricular arrhythmia. Based on the current data, the Task Force recommends that individualized ICD decisions should be based using well-established risk factors and not solely on the presence of an LV apical aneurysm.

7.1.5.2. Left ventricular systolic dysfunction

A small number of retrospective studies and two larger registries have examined the relation between prognosis in patients with HCM and LV systolic dysfunction (most frequently defined by a LVEF <50%) (see Supplementary data online, Table S3). All studies consistently show an increased rate of SCD events in patients with left ventricular systolic dysfunction (LVSD) ranging from 7 to 20% compared with that of patients with normal LV systolic function. However, the independent and additional value of LVSD compared with current risk stratification tools has not been investigated. There is only one multivariable model that investigates the independent relation of LVSD to the risk of SCD events but the covariables examined were limited (age, sex, and follow-up time).315 As with other recently proposed risk markers in HCM, the Task Force maintains its recommendation to first estimate SCD risk using the HCM-SCD Risk and HCM Risk-Kids tools, and then to use the presence of an LVEF <50% in shared decision-making about prophylactic ICD implantation, with full disclosure of the lack of robust data on its impact on prognosis.

7.1.5.3. Late gadolinium enhancement on cardiac magnetic resonance imaging

In the 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy, the extent of LGE on CMR was considered helpful in predicting cardiovascular mortality, but data at that time were felt insufficient to support the use of LGE in prediction of SCD risk. Since then, more studies have been published (see Supplementary data online, Table S1). In aggregate, the data show that LGE is common and, that when extensive (expressed as a percentage of LV mass), is associated with an increase in SCD risk and other events, particularly in the presence of other markers of disease severity including LV systolic impairment. A meta-analysis of nearly 3000 patients from several studies suggests that the presence of LGE is associated with a 2.32-fold increased risk of SCD/aborted SCD/appropriate ICD discharge, and a 2.1-fold increase in all-cause mortality.794 It has been suggested that the addition of LGE to the current AHA/ACC sudden death algorithm or the HCM-SCD risk model improves stratification of patients who are otherwise considered low or intermediate risk.793

As in 2014, a number of uncertainties persist. These include the inevitable confounders in the retrospective studies that bias towards high-risk patients or patients referred specifically for septal myectomy. There also remains some debate about the methods used to quantify LGE with the 2-standard deviation technique; the only one that is validated against necropsy.605 Retrospective CMR series also report relatively high event rates suggesting that they are not representative of the broad spectrum of disease. In HCMR, a prospective CMR study of 2755 patients, LGE was present in 50% of patients based on visual criteria and in 60% based on >6 SCD signal criteria, but only 2% of patients had LGE >15% of LV mass.124 In the most recent report from the registry, there have been 24 deaths from any cause after a mean follow-up of 33.5 ± 12.4 months (median: 36 months and range 1–64 months); the relation with LGE is not reported.795 There are very limited data on the role of CMR over and above validated risk algorithms in SCD risk prediction in children with HCM.796,797

On balance, the Task Force maintains the recommendation to first estimate SCD risk using the HCM-SCD Risk calculators. For patients who are in the low to intermediate risk category, the presence of extensive LGE (≥15%) may be used in shared decision-making with patients about prophylactic ICD implantation, acknowledging the lack of robust data on the impact of scar quantification on the personalized risk estimates generated by the HCM-SCD Risk calculators.

7.1.5.4. Abnormal exercise blood pressure response

Approximately one-third of adult patients with HCM have an abnormal systolic blood pressure response to exercise characterized by progressive hypotension or a failure to augment the systolic blood pressure that is caused by an inappropriate drop in systemic vascular resistance and a low cardiac output reserve.798,799 Various definitions for abnormal blood pressure response in patients with HCM have been reported;590,765,768,782 for the purposes of this guideline, an abnormal blood pressure response is defined as a failure to increase systolic pressure by at least 20 mmHg from rest to peak exercise or a fall of >20 mmHg from peak pressure.590,765,768,782,800

Abnormal exercise blood pressure response may be associated with a higher risk of SCD in adult patients aged ≤40 years, but it has a low positive predictive accuracy and its prognostic significance in patients >40 years of age is unknown, and recent data have suggested that, although it may be associated with increased overall mortality largely related to heart failure, it is not consistently associated with an increased risk of ventricular arrhythmia or SCD.800,801 There is no evidence to suggest that abnormal blood pressure response to exercise is associated with a higher risk of SCD in children with HCM.802 The Task Force, therefore, does not recommend the use of abnormal blood pressure response to exercise as an indication for primary prevention ICD implantation in patients with a low or intermediate risk category.

7.1.5.5. Sarcomeric variants

A small number of studies have explored the prognostic value of sarcomeric variants in HCM. Despite initial attempts to classify variants as ‘malignant’ or ‘benign’,803–807 no studies have shown an independent role for specific sarcomeric variants in SCD risk prediction. Variants initially classified as ‘malignant’ or ‘benign’ can have very different phenotypic expression, even in members of the same family,808–810 and, as variants are often found in individual families, evaluation of their prognostic implications is problematic. Similarly, while the presence of multiple sarcomeric variants in an individual has been suggested to be associated with a worse prognosis,608,811–813 other cohorts have not consistently reported this association.807,814–816 Recent studies have evaluated the potential prognostic role of the presence of any sarcomeric variant. The largest of these, comprising 2763 patients, showed a statistically significant impact on overall prognosis in those with vs. without a sarcomeric variant, but did not assess its association specifically with SCD.238 A smaller study of 512 probands and 114 relatives, of whom 327 had a disease-causing sarcomeric variant, suggested that the presence of a pathogenic variant was independently associated with all-cause, cardiovascular, and heart failure mortality as well as SCD/aborted SCD (HR 2.88; 95% CI, 1.23–6.71).817 Patients with a sarcomeric variant were younger and were more likely to have NSVT, syncope, and LVOTO and the association with SCD lost statistical significance (HR 2.44; 95% CI, 0.99–6.01; P = 0.052) after adjusting for ≥2 major clinical risk factors. The role of sarcomeric variants as a predictor of SCD independent of SCD risk-prediction models (e.g. HCM Risk-SCD and HCM Risk-Kids) remains to be demonstrated. Based on the available data, the Task Force does not recommend the use of the presence of sarcomeric variant(s) to guide decisions around ICD implantation for primary prevention in individuals with a low or intermediate SCD risk score.

7.1.5.6. Prevention of sudden cardiac death

There are no randomized, controlled data to support the use of AADs for the prevention of SCD in HCM. Amiodarone was associated with a lower incidence of SCD in one small observational study of patients with non-sustained ventricular tachycardia (NSVT) on Holter monitoring, but observational data suggest that amiodarone often fails to prevent SCD.818,819 Disopyramide does not appear to have a significant impact on the risk of SCD.632 However, beta-blockers and/or amiodarone are recommended in patients with an ICD who continue to have symptomatic ventricular arrhythmias, paroxysmal AF, or recurrent shocks despite optimal treatment and device re-programming.820

There are no randomized trials or statistically validated prospective prediction models that can be used to guide ICD implantation in patients with HCM. Recommendations are instead based on observational, retrospective cohort studies that have determined the relationship between clinical characteristics and prognosis. The 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy recommended a risk-prediction model—HCM Risk-SCD (https://qxmd.com/calculate/calculator_303/hcm-risk-scd)—that provides individualized, quantitative risk estimates using an enhanced phenotypic approach.525 This approach has since been validated in independent cohorts and a meta-analysis of available published data, relevant to the 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy performance, for SCD prevention has shown that pooled estimates are concordant with the observed SCD risk in patients designated as high or low risk.821–824 In children, risk stratification for SCD has traditionally been based on risk factors extrapolated from adults with HCM, but this approach does not identify the children most at risk of SCD. In 2019, the first validated paediatric-specific risk model for SCD was developed (HCM Risk-Kids; https://hcmriskkids.org), using a similar approach to HCM Risk-SCD,81,825 and has since been independently externally validated.535,797,826 A similar paediatric risk-prediction model (PRIMaCY Childhood HCM Sudden Cardiac Death Risk Prediction tool) has also been developed, using similar clinical parameters and with similar reported accuracy to HCM Risk-Kids (https://primacy.shinyapps.io/calculator/).535

In this update, the Task Force maintains the principle of risk estimation using the validated HCM Risk-SCD tool as the first step in sudden death prevention in patients aged 16 years or more, and recommends the use of a validated risk score (e.g. HCM Risk-Kids tool) for children and adolescents <16 years. This is in contrast to the 2020 AHA/ACC Guideline for the diagnosis and treatment of patients with hypertrophic cardiomyopathy,793 in which the tool is considered an aid to a shared decision-making process for ICD placement in patients with clinical risk markers. This approach by the AHA/ACC, in part, reflected concerns that reliance on a risk tool does not account for individual patient perception and acceptance of pre-determined thresholds for medical intervention, as well as the omission of clinical risk markers such as LV systolic impairment from the HCM Risk-SCD model.

The Task Force acknowledges the challenges associated with defining universal thresholds for acceptable risk, but feels that reliance on an unquantified estimate of risk does nothing to resolve this dilemma. Instead, the Task Force recommends more overt shared decision-making based on real-world data as well as individual preferences, beliefs, circumstances, and values. Gaps in evidence are acknowledged and should be shared with patients. Similarly, competing risks related to the disease (heart failure, stroke) and to age and comorbidity, as well as device-related complications, should be discussed.726,827,828 Critically, the Task Force calls for development of enhanced patient decision aids tailored specifically to receivers of care as well as more traditional decision-support tools for healthcare practitioners.

Figure 16 summarizes the recommendations for primary prevention ICD implantation in HCM in each risk category. These take into account not only the absolute statistical risk, but also the age and general health of the patient, socioeconomic factors, and the psychological impact of therapy. The recommendations are meant to be sufficiently flexible to account for scenarios that are not encompassed by the HCM Risk-SCD or HCM Risk-Kids models. These models should not be used in elite athletes or in individuals with metabolic/infiltrative diseases (e.g. Anderson–Fabry disease) and syndromes (e.g. Noonan syndrome). The models do not use exercise-induced LV outflow tract gradients and have not been validated before and after myectomy. The HCM Risk-SCD model has been validated in one study of adult patients following ASA,829 and a recent study has suggested that severe LVH and residual LVOTO are associated with an increased risk of SCD following ASA, with a modest C-statistic of 0.68.830

Flow chart for implantation of an implantable cardioverter defibrillator in patients with hypertrophic cardiomyopathy.
Figure 16

Flow chart for implantation of an implantable cardioverter defibrillator in patients with hypertrophic cardiomyopathy.

2D, two-dimensional; CMR, cardiac magnetic resonance; ECG, electrocardiogram; HCM, hypertrophic cardiomyopathy; ICD, implantable cardioverter defibrillator; LGE, late gadolinium enhancement; LV, left ventricular; LVEF, left ventricular ejection fraction; NSVT, non-sustained ventricular tachycardia; SCD, sudden cardiac death; VF, ventricular fibrillation; VT, ventricular tachycardia. aClinical risk factors: extensive LGE (>15%) on CMR; LVEF <50%.

Recommendation Table 23

Additional recommendations for prevention of sudden cardiac death in patients with hypertrophic cardiomyopathy

graphic
graphic
graphic
graphic
graphic
graphic
Recommendation Table 23

Additional recommendations for prevention of sudden cardiac death in patients with hypertrophic cardiomyopathy

graphic
graphic
graphic
graphic
graphic
graphic

7.2. Dilated cardiomyopathy

7.2.1. Diagnosis

7.2.1.1. Index case

Dilated cardiomyopathy is defined by the presence of LV dilatation and systolic dysfunction unexplained solely by abnormal loading conditions or CAD. Left ventricular dilatation is defined by LV end-diastolic dimensions or volumes >2 z-scores above population mean values corrected for body size, sex, and/or age. For adults this represents an LV end-diastolic diameter >58 mm in males and >52 in females and an LVEDV index of ≥75 mL/m2 in males and ≥62 mL/m2 in females by ECHO.9,845,846 Left ventricular global systolic dysfunction is defined by LVEF <50%.9

7.2.1.2. Relatives

Clinical testing in relatives often reveals mild non-diagnostic abnormalities that overlap with normal variation or mimic changes seen in other more common diseases such as hypertension and obesity. In this context, the presence of isolated LV dilatation with preserved systolic function or in the presence of a familial causative variant is sufficient for a diagnosis of DCM in a relative. Additional electrocardiographic or imaging abnormalities in the context of a family history of DCM are suggestive of disease and warrant close follow-up.9,75,817 In the absence of conclusive genetic information in a family, DCM is considered familial if: (i) one or more first- or second-degree relatives have DCM; or (ii) when an otherwise unexplained SCD has occurred in a first-degree relative at any age with an established diagnosis of DCM.

7.2.1.3. Diagnostic work-up

The key elements of the diagnostic work-up for all patients with DCM are described in Section 6 and include clinical and family history, laboratory tests, ECG, Holter monitoring, cardiac imaging, and genetic testing. Echocardiography is central for the diagnosis and CMR provides more detailed morphological and prognostic information. Additional laboratory tests, exercise testing, EMB, cardiac CT, and cardiac catheterization should also be considered, as detailed in Section 6.

7.2.1.4. Echocardiography

Comprehensive TTE is recommended for all DCM patients as it provides all the relevant information on the global and regional LV anatomy, function and haemodynamics, valvular heart disease, right heart function, pulmonary pressure, atrial geometry, and associated features.71 Advanced echocardiographic techniques (tissue Doppler and speckle tracking deformation imaging) can allow the early detection of subclinical myocardial dysfunction in specific situations (e.g. genetic DCM carriers, recipients of known cardiotoxic chemotherapy).71,74

Contrast agents may be considered for better endocardial delineation, to better depict the presence of hypertrabeculation, or to exclude intraventricular thrombus. Transoesophageal echocardiography is rarely necessary except for when atrial thrombi are present in patients with AF, or for assessing valvular function and guiding transcatheter therapy in patients with concomitant secondary mitral or tricuspid regurgitation.

7.2.1.5. Cardiac magnetic resonance

Cardiac magnetic resonance provides additional information on tissue characterization in patients with DCM, including the presence of myocardial oedema, which may suggest a myocarditic or inflammatory cause, and LGE, to determine the presence and extent of fibrosis, as well as its distribution, which may allow exclusion of myocardial infarction and also point towards specific aetiologies (e.g. subepicardial distribution in post-myocarditis forms, patchy in sarcoidosis, extensive inferolateral in dystrophinopathies, septal mid-wall in LMNA carriers, and ring-like in DSP and FLNC-truncating variant carriers) (see Section 7.3).71,847 Late gadolinium enhancement distribution and extent hold prognostic value both for arrhythmia and heart failure severity.137,848 Dedicated T2* sequences describe myocardial iron deposition, which is useful for the diagnosis of haemochromatosis.71

7.2.1.6. Nuclear medicine

There is a limited role for radionuclide imaging in DCM. Measurement of 18F-fluorodeoxyglucose (18F-FDG) uptake using PET, with focal or focal-on-diffuse FDG uptake patterns especially if there is concomitant abnormal 18F-FDG-PET uptake in extracardiac tissues, can be useful in suspected cardiac sarcoidosis.849

7.2.2. Genetic testing and family screening

The aetiology of DCM is highly heterogeneous and includes inherited (genetic/familial) and acquired causes.9,545,850,851 Direct causes of DCM include pathogenic gene variants, toxins, auto-immunity, infections, storage diseases, and tachyarrhythmias. Monogenic gene variants causing DCM are highly heterogeneous, implicating many genes and diverse pathways. Moreover, only 30–40% of DCM cases are attributable to pathogenic rare variants, with a substantial polygenic/common variant contribution in this population. Furthermore, disease modifiers can play a role in the acceleration of the DCM phenotype.7,9,850 This includes conditions that may aggravate or trigger DCM, including epigenetic factors and acquired modifiers, such as pregnancy, hypertension, excessive alcohol use, and other toxins.42–44 It is important to consider the interplay between genetic and acquired causes during the diagnostic work-up. Identification of an acquired cause does not exclude an underlying causative gene variant, whereas the latter may require an additional acquired cause and/or disease modifier to manifest. Within the genes that can cause DCM, there are genes robustly associated with classical DCM that have been recently curated,189 and also others classically associated with ARVC but that very commonly can present with LV dilatation and predominantly LV dysfunction. Moreover, genes described in the context of hypertrabeculation/LVNC (e.g. NKX2.5 and PRDM16), or that can cause DCM with or without skeletal involvement (such as DMD or EMD), should also be considered DCM-associated genes and examined, particularly if phenotype is concordant. The most common genetic and acquired causes of DCM are shown in Table 10 and Table 20. Detailed lists of causes of DCM have been previously published.9,852

Table 20

Non-genetic causes of dilated cardiomyopathy

Infection (post-myocarditis)
Viral (enteroviruses, adenoviruses, echoviruses, herpes viruses, parvovirus B19, HIV, SARS-CoV-2, etc.)
Bacterial (Lyme disease)
Mycobacterial
Fungal
Parasitic (Chagas disease)
Toxic and overload
Alcohol (ethanol)
Cocaine, amphetamines, ecstasy
Cobalt
Anabolic/androgenic steroids
Haemochromatosis and other causes of iron overload
Endocrinology
Hypo- and hyperthyroidism
Cushing/Addison disease
Phaeochromocytoma
Acromegaly
Diabetes mellitus
Nutritional deficiency
Selenium deficiency
Thiamine deficiency (Beri-Beri)
Zinc and copper deficiency
Carnitine deficiency
Electrolyte disturbance
Hypocalcaemia
Hypophosphataemia
Peripartum
Autoimmune diseases
Giant cell myocarditis
Inflammatory (biopsy-proven, non-infectious myocarditis)
Eosinophilic granulomatosis with polyangiitis
Systemic lupus erythematosus
Sarcoidosis
Rheumatoid arthritis
Coeliac disease
Primary biliary cirrhosis
Myasthenia gravis
Pemphigus pemphigoid
Crohn disease
Ulcerative colitis
Polymyositis/dermatomyositis
Reactive arthritis
Drugs
Antineoplastic drugsAnthracyclines; antimetabolites; alkylating agents; Taxol; hypomethylating agent; monoclonal antibodies; tyrosine kinase inhibitors; immunomodulating agents
Psychiatric drugsClozapine, olanzapine; chlorpromazine, risperidone, lithium; methylphenidate; tricyclic antidepressants
Other drugsAll-trans retinoic acid; antiretroviral agents; phenothiazines
Infection (post-myocarditis)
Viral (enteroviruses, adenoviruses, echoviruses, herpes viruses, parvovirus B19, HIV, SARS-CoV-2, etc.)
Bacterial (Lyme disease)
Mycobacterial
Fungal
Parasitic (Chagas disease)
Toxic and overload
Alcohol (ethanol)
Cocaine, amphetamines, ecstasy
Cobalt
Anabolic/androgenic steroids
Haemochromatosis and other causes of iron overload
Endocrinology
Hypo- and hyperthyroidism
Cushing/Addison disease
Phaeochromocytoma
Acromegaly
Diabetes mellitus
Nutritional deficiency
Selenium deficiency
Thiamine deficiency (Beri-Beri)
Zinc and copper deficiency
Carnitine deficiency
Electrolyte disturbance
Hypocalcaemia
Hypophosphataemia
Peripartum
Autoimmune diseases
Giant cell myocarditis
Inflammatory (biopsy-proven, non-infectious myocarditis)
Eosinophilic granulomatosis with polyangiitis
Systemic lupus erythematosus
Sarcoidosis
Rheumatoid arthritis
Coeliac disease
Primary biliary cirrhosis
Myasthenia gravis
Pemphigus pemphigoid
Crohn disease
Ulcerative colitis
Polymyositis/dermatomyositis
Reactive arthritis
Drugs
Antineoplastic drugsAnthracyclines; antimetabolites; alkylating agents; Taxol; hypomethylating agent; monoclonal antibodies; tyrosine kinase inhibitors; immunomodulating agents
Psychiatric drugsClozapine, olanzapine; chlorpromazine, risperidone, lithium; methylphenidate; tricyclic antidepressants
Other drugsAll-trans retinoic acid; antiretroviral agents; phenothiazines

HIV, human immunodeficiency virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

Table 20

Non-genetic causes of dilated cardiomyopathy

Infection (post-myocarditis)
Viral (enteroviruses, adenoviruses, echoviruses, herpes viruses, parvovirus B19, HIV, SARS-CoV-2, etc.)
Bacterial (Lyme disease)
Mycobacterial
Fungal
Parasitic (Chagas disease)
Toxic and overload
Alcohol (ethanol)
Cocaine, amphetamines, ecstasy
Cobalt
Anabolic/androgenic steroids
Haemochromatosis and other causes of iron overload
Endocrinology
Hypo- and hyperthyroidism
Cushing/Addison disease
Phaeochromocytoma
Acromegaly
Diabetes mellitus
Nutritional deficiency
Selenium deficiency
Thiamine deficiency (Beri-Beri)
Zinc and copper deficiency
Carnitine deficiency
Electrolyte disturbance
Hypocalcaemia
Hypophosphataemia
Peripartum
Autoimmune diseases
Giant cell myocarditis
Inflammatory (biopsy-proven, non-infectious myocarditis)
Eosinophilic granulomatosis with polyangiitis
Systemic lupus erythematosus
Sarcoidosis
Rheumatoid arthritis
Coeliac disease
Primary biliary cirrhosis
Myasthenia gravis
Pemphigus pemphigoid
Crohn disease
Ulcerative colitis
Polymyositis/dermatomyositis
Reactive arthritis
Drugs
Antineoplastic drugsAnthracyclines; antimetabolites; alkylating agents; Taxol; hypomethylating agent; monoclonal antibodies; tyrosine kinase inhibitors; immunomodulating agents
Psychiatric drugsClozapine, olanzapine; chlorpromazine, risperidone, lithium; methylphenidate; tricyclic antidepressants
Other drugsAll-trans retinoic acid; antiretroviral agents; phenothiazines
Infection (post-myocarditis)
Viral (enteroviruses, adenoviruses, echoviruses, herpes viruses, parvovirus B19, HIV, SARS-CoV-2, etc.)
Bacterial (Lyme disease)
Mycobacterial
Fungal
Parasitic (Chagas disease)
Toxic and overload
Alcohol (ethanol)
Cocaine, amphetamines, ecstasy
Cobalt
Anabolic/androgenic steroids
Haemochromatosis and other causes of iron overload
Endocrinology
Hypo- and hyperthyroidism
Cushing/Addison disease
Phaeochromocytoma
Acromegaly
Diabetes mellitus
Nutritional deficiency
Selenium deficiency
Thiamine deficiency (Beri-Beri)
Zinc and copper deficiency
Carnitine deficiency
Electrolyte disturbance
Hypocalcaemia
Hypophosphataemia
Peripartum
Autoimmune diseases
Giant cell myocarditis
Inflammatory (biopsy-proven, non-infectious myocarditis)
Eosinophilic granulomatosis with polyangiitis
Systemic lupus erythematosus
Sarcoidosis
Rheumatoid arthritis
Coeliac disease
Primary biliary cirrhosis
Myasthenia gravis
Pemphigus pemphigoid
Crohn disease
Ulcerative colitis
Polymyositis/dermatomyositis
Reactive arthritis
Drugs
Antineoplastic drugsAnthracyclines; antimetabolites; alkylating agents; Taxol; hypomethylating agent; monoclonal antibodies; tyrosine kinase inhibitors; immunomodulating agents
Psychiatric drugsClozapine, olanzapine; chlorpromazine, risperidone, lithium; methylphenidate; tricyclic antidepressants
Other drugsAll-trans retinoic acid; antiretroviral agents; phenothiazines

HIV, human immunodeficiency virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

7.2.2.1. Genetic testing

Causative gene variants occur in up to 40% of DCM patients in contemporary cohorts,185,186,853,854 and between 10 and 15% in chemotherapy-induced, alcoholic, or peripartum DCM.42–44 Although the prevalence of genetic variants is higher in familial DCM, causative genetic variants are also identified in over 20% of non-familial DCM cases.185,854,855 Finding a causative gene variant in a patient with DCM allows better prediction of the disease outcome and progression, may contribute to the indications for device implantation, informs genetic counselling, and allows familial screening for relatives. Moreover, genetic testing in DCM has long-term implications in terms of cost-effectiveness by identifying at-risk family members with positive genotypes, and thus reducing the number of family members requiring serial clinical follow-up.229 Genetic testing can therefore be beneficial in all patients with DCM, including children856,857 and those with alcohol-/chemotherapy-induced and peripartum DCM. Where resources are limited, scores designed to identify DCM patients with a high probability of a positive genotype (e.g. the Madrid DCM Genotype Score [https://madridDCMscore.com]) may be considered to prioritize genetic testing.858 Of note, age should not be a limiting factor when deciding which DCM patients should undergo genetic testing.185,858,859

Recommendations for clinical screening, genetic counselling, and testing are described in Sections 6.8.3 and 6.11. More detailed evaluation of conduction defects or arrhythmia, which may be an early presentation of certain genetic DCM subtypes, should be considered in the context of certain gene variants (e.g. LMNA, EMD, DES). Cardiac MRI should also be considered in relatives with normal cardiac function who carry causative genetic variants associated with increased risk of SCD (e.g. FLNC, DES, DSP, PLN, LMNA, TMEM43, RMB20). If there are no additional family members with DCM, other than the proband, periodic evaluation of first-degree relatives should follow the same intervals according to age (see Section 6.11), but termination of periodic surveillance in families in whom a genetic variant has not been identified could be considered in first-degree relatives ≥50 years of age with normal ECG and normal cardiac imaging tests.

7.2.3. Assessment of symptoms

Patients with DCM often develop symptoms of heart failure, although this can occur many years after the appearance of ECG or echocardiographic abnormalities. Assessment of symptoms in patients with cardiomyopathies is described in Section 6.10.1.

7.2.4. Management

The clinical management of heart failure and other manifestations of DCM has been described in the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure, the 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation, and the 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy.69,336,724 In these guidelines, recommendations are generally independent of the aetiology of heart failure, AF, and other clinical presentations. As such, although they summarize large and robust datasets and trials, the treatment recommendations must be regarded as generic and not specific to the different forms of genetic DCM. However, as large cohorts of genetic DCMs with uniform genetic features are relatively rare, adequately powered RCTs in cardiomyopathies are scarce. The Task Force therefore recommends applying the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure, which contain treatment guidelines for patients with signs and symptoms of heart failure, for symptom management of patients with DCM.69 Treatment recommendations for asymptomatic LV dysfunction or dilatation are scarce, which presents a challenge for genetic DCM, where a sizeable proportion of the patients are young with no or mild symptoms, and where asymptomatic patients are frequently discovered through cascade screening. Recommendations for the pharmacological management of heart failure symptoms in patients with cardiomyopathies are described in Section 6.10.2.

7.2.5. Sudden cardiac death prevention in dilated cardiomyopathy

Predicting SCD is a challenging aspect of the clinical care of patients with DCM. Implantable cardioverter defibrillators are effective at treating potentially lethal ventricular arrhythmias and preventing SCD, but are also associated with complications, particularly in young patients, who will require several replacements during their lifetimes (see Section 6.10.5).

7.2.5.1. Secondary prevention of sudden cardiac death

Implantable cardioverter defibrillators reduce mortality in survivors of cardiac arrest and in patients who have experienced sustained symptomatic ventricular arrhythmias with haemodynamic compromise.531 An ICD is recommended in such patients when the intent is to increase survival; the decision to implant should consider the patient’s view and their QoL, as well as the absence of other diseases likely to cause death within the following year.

7.2.5.2. Primary prevention of sudden cardiac death

Available RCTs examining the usefulness of ICDs to prevent SCD and improve survival have included only patients with LVEF ≤35%, with conflicting results. While a trial including both ischaemic and non-ischaemic symptomatic heart failure patients showed reduction in mortality,860 trials including only patients without CAD did not significantly improve the overall risk of mortality despite the fact that there was an absolute reduction in SCD with ICDs, and subgroup analysis suggested that there was a survival benefit in patients ≤70 years.861,862 Nevertheless, in a recent meta-analysis of studies that examined the effect of ICDs in DCM, a survival benefit was observed, although the effect was modest compared with LVEF ≤35% patients with CAD.863

Although LVEF ≤35% has been reported as an independent risk marker of all-cause and cardiac death in DCM, it has also shown only modest ability in identifying DCM patients with higher risk of SCD, suggesting that additional factors should be taken into consideration when deciding on ICD implantation in a disease with significant aetiological heterogeneity. Recent natural history studies suggest that phenotype plays a role a role in SCD risk, with patients harbouring disease-causing variants in PLN, DSP, LMNA, FLNC, TMEM43, and RBM20 having a substantially higher rate of major arrhythmic events than other causes of DCM regardless of LVEF.440,542,864–870 A recent retrospective study of 1161 individuals with DCM has shown that DCM patients with P/LP DCM-causing genetic variants have a worse clinical evolution and higher rate of major arrhythmic events than genotype-negative DCM patients and particularly in DCM patients with LVEF ≤35%.185 The study also observed a higher risk of major arrhythmic events in DCM patients affected by DCM-causing variants in certain genotypes regardless of LVEF. Genes associated with higher arrhythmic risk included genes coding for nuclear envelope (LMNA, EMD, TMEM43), desmosomal (DSP, DSG2, DSC2, PKP2), and certain cytoskeletal proteins.185 Together, these data suggest that DCM patients harbouring DCM-causing variants in high-risk genes (LMNA, EMD, TMEM43, DSP, RBM20, PLN, FLNC-truncating variants) should be considered as patients with a high-risk genetic background for SCD and primary prevention ICD implantation should be considered with LVEF thresholds higher than 35%, particularly in the presence of additional risk factors (e.g. NSVT, increased ventricular ectopic beats, male sex, significant LGE, specific gene variant). For some high-risk genotypes (e.g. LMNA [https://lmna-risk-vta.fr]541), gene-specific (or, in the case of the PLN p.Arg14del variant, variant-specific [https://plnriskcalculator.shinyapps.io/final_shiny])542 risk-prediction scores have been developed that consider genotype characteristics and additional phenotypic features. Where such scores are available, they should be used to guide primary prevention ICD implantation (Figure 17). As discussed in Section 7.1.5, the Task Force acknowledges the challenges associated with defining universal thresholds for acceptable risk across different cardiomyopathy phenotypes, but is of the opinion that a similar approach to that taken in risk stratification for HCM is reasonable. Although the 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death have suggested a higher threshold of 10% risk at 5 years to guide primary prevention ICD implantation in patients with DCM and LMNA variants,3 this Task Force recommends shared decision-making based on real-world data as well as individual preferences, beliefs, circumstances, and values. Gaps in evidence should be shared with patients, and competing risks related to the disease (heart failure, stroke), and to age and comorbidity, as well as device-related complications, should be discussed. In support of this, a recent study validating the LMNA-risk VTA calculator overestimated arrhythmic risk when using ≥7% predicted 5-year risk as threshold (specificity 26%, C-statistic 0.85), despite a high sensitivity.871

Implantation of implantable cardioverter defibrillators in patients with dilated cardiomyopathy or non-dilated left ventricular cardiomyopathy flowchart.
Figure 17

Implantation of implantable cardioverter defibrillators in patients with dilated cardiomyopathy or non-dilated left ventricular cardiomyopathy flowchart.

CMR, cardiac magnetic resonance; DCM, dilated cardiomyopathy; ICD, implantable cardioverter defibrillator; LGE, late gadolinium enhancement; LVEF, left ventricular ejection fraction; NDLVC, non-dilated left ventricular cardiomyopathy; VE, ventricular ectopic beats; VT, ventricular fibrillation. aSee Table 21. bStrength of recommendation depends on gene and context. cAdditional risk factors include syncope, LGE presence on CMR.

Importantly, there are also data to suggest that other genotypes (e.g. TTN truncating variants) are associated with recovery of LVEF with standard heart failure criteria from the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure.185,867,872

In patients without a high-risk genotype and LVEF >35%, the presence and extent of myocardial scarring determined by LGE on CMR imaging can be helpful in risk stratification in patients with DCM.873,874 Late gadolinium enhancement is observed in 25–35% of patients with DCM and its presence is a strong risk marker for all-cause mortality and ventricular arrhythmias, both in retrospective and prospective studies. A recent prospective study of 1020 DCM patients with a median follow-up of 5.2 years showed that myocardial scar had a strong and incremental prognostic value for predicting SCD, while LVEF ≤35% was not associated with SCD.138 In another study, a risk calculator was developed that among others, incorporated the presence of LGE on CMR imaging540, although this has not yet been externally validated. There are at least two ongoing trials of ICD therapy according to the presence of scar on CMR imaging, including DCM patients (NCT04558723 and NCT03993730), but the Task Force’s opinion is that the existing level of evidence can support using LGE to guide ICD implantation in subgroups of patients with DCM (Figure 17). Additional risk factors, such as syncope or the presence of NSVT and burden of ventricular ectopy (VE), may also help guide ICD implantation. There are no data currently to support a specific threshold for VE burden, and this will depend on the underlying genotype and other clinical factors.542,867,872 In patients with unexplained syncope, programmed electrical stimulation (PES) may provide additional information on the underlying cause.875 There are no definitive data supporting the routine use of PES for primary prevention risk stratification in patients with DCM, but this may be beneficial in patients with DCM and myotonic dystrophy with an independent indication to electrophysiological study to assess conduction disturbances,876 although the clinical value of this approach has not been consistently demonstrated.877

Recommendation Table 24

Recommendations for an implantable cardioverter defibrillator in patients with dilated cardiomyopathy

graphic
graphic
Recommendation Table 24

Recommendations for an implantable cardioverter defibrillator in patients with dilated cardiomyopathy

graphic
graphic

7.3. Non-dilated left ventricular cardiomyopathy

7.3.1. Diagnosis

7.3.1.1. Index case

The non-dilated LV cardiomyopathy phenotype is defined by the presence of non-ischaemic LV scarring or fatty replacement in the absence of LV dilatation, with or without global or regional wall motion abnormalities, or isolated global LV hypokinesia without scarring (as assessed by the presence of LGE on CMR) that is unexplained solely by abnormal loading conditions (hypertension, valve disease) or CAD. Global LV systolic dysfunction is defined by abnormal LVEF (i.e. <50%).9

Table 21

High-risk genotypes and associated predictors of sudden cardiac death

GeneAnnual SCD ratePredictors of SCD
LMNA185,186,438,541,865,878,8795–10%Estimated 5-year risk of life-threatening arrhythmia using LMNA risk score (https://lmna-risk-vta.fr)
FLNC-truncating variants866,867,8805–10%LGE on CMR
LVEF < 45%
TMEM43868,8815–10%Male
Female and any of the following: LVEF <45%, NSVT, LGE on CMR, >200 VE on 24h Holter ECG
PLN542,882,8833–5%Estimated 5-year risk of life-threatening arrhythmia using PLN risk score (https://plnriskcalculator.shinyapps.io/final_shiny)
LVEF < 45%
LGE on CMR
NSVT
DSP185,1863–5%LGE on CMR
LVEF < 45%
RBM208693–5%LGE on CMR
LVEF < 45%
GeneAnnual SCD ratePredictors of SCD
LMNA185,186,438,541,865,878,8795–10%Estimated 5-year risk of life-threatening arrhythmia using LMNA risk score (https://lmna-risk-vta.fr)
FLNC-truncating variants866,867,8805–10%LGE on CMR
LVEF < 45%
TMEM43868,8815–10%Male
Female and any of the following: LVEF <45%, NSVT, LGE on CMR, >200 VE on 24h Holter ECG
PLN542,882,8833–5%Estimated 5-year risk of life-threatening arrhythmia using PLN risk score (https://plnriskcalculator.shinyapps.io/final_shiny)
LVEF < 45%
LGE on CMR
NSVT
DSP185,1863–5%LGE on CMR
LVEF < 45%
RBM208693–5%LGE on CMR
LVEF < 45%

CMR, cardiac magnetic resonance; DSP, desmoplakin; ECG, electrocardiogram; FLNC, filamin C; LGE, late gadolinium enhancement; LMNA, lamin A/C; LVEF, left ventricular ejection fraction; NSVT, non-sustained ventricular tachycardia; PLN, phospholamban; RMB, RNA binding motif protein; SCD, sudden cardiac death; VE, ventricular ectopic beats.

Table 21

High-risk genotypes and associated predictors of sudden cardiac death

GeneAnnual SCD ratePredictors of SCD
LMNA185,186,438,541,865,878,8795–10%Estimated 5-year risk of life-threatening arrhythmia using LMNA risk score (https://lmna-risk-vta.fr)
FLNC-truncating variants866,867,8805–10%LGE on CMR
LVEF < 45%
TMEM43868,8815–10%Male
Female and any of the following: LVEF <45%, NSVT, LGE on CMR, >200 VE on 24h Holter ECG
PLN542,882,8833–5%Estimated 5-year risk of life-threatening arrhythmia using PLN risk score (https://plnriskcalculator.shinyapps.io/final_shiny)
LVEF < 45%
LGE on CMR
NSVT
DSP185,1863–5%LGE on CMR
LVEF < 45%
RBM208693–5%LGE on CMR
LVEF < 45%
GeneAnnual SCD ratePredictors of SCD
LMNA185,186,438,541,865,878,8795–10%Estimated 5-year risk of life-threatening arrhythmia using LMNA risk score (https://lmna-risk-vta.fr)
FLNC-truncating variants866,867,8805–10%LGE on CMR
LVEF < 45%
TMEM43868,8815–10%Male
Female and any of the following: LVEF <45%, NSVT, LGE on CMR, >200 VE on 24h Holter ECG
PLN542,882,8833–5%Estimated 5-year risk of life-threatening arrhythmia using PLN risk score (https://plnriskcalculator.shinyapps.io/final_shiny)
LVEF < 45%
LGE on CMR
NSVT
DSP185,1863–5%LGE on CMR
LVEF < 45%
RBM208693–5%LGE on CMR
LVEF < 45%

CMR, cardiac magnetic resonance; DSP, desmoplakin; ECG, electrocardiogram; FLNC, filamin C; LGE, late gadolinium enhancement; LMNA, lamin A/C; LVEF, left ventricular ejection fraction; NSVT, non-sustained ventricular tachycardia; PLN, phospholamban; RMB, RNA binding motif protein; SCD, sudden cardiac death; VE, ventricular ectopic beats.

7.3.1.2. Relatives

Clinical testing in relatives may reveal non-diagnostic abnormalities. In this context, the presence of LV systolic global or regional dysfunction, or additional electrocardiographic abnormalities (e.g. repolarization abnormalities, low QRS voltages, frequent ventricular extrasystoles [>500 per 24 h] or NSVT) in a first-degree relative of an individual with NDLVC (or a first-degree relative with autopsy-proven NDLVC) is highly suggestive of NDLVC and warrants close follow-up.

In the absence of conclusive genetic information in the family, NDLVC should be considered familial if one or more first- or second-degree relatives have NDLVC, or when SCD has occurred in a first-degree relative at any age with an established diagnosis of NDLVC. Familial disease should also be suspected if a first-degree relative has sudden death at <50 years of age and autopsy findings suggestive of the NDLVC phenotype.

7.3.1.3. Diagnostic work-up

The key elements of the diagnostic work-up for all patients with NDLVC are described in Section 6 and include clinical history, laboratory tests, Holter monitoring and cardiac imaging, and genetic testing. Echocardiography and CMR are both central to the diagnosis. Additional laboratory tests, exercise testing, EMB, and cardiac catheterization may also be considered (see Section 6).

7.3.1.4. Electrocardiographic features

Recommendations on resting and ambulatory ECG testing are described in Section 6.5 and are of particular importance in patients with NDLVC, as specific features can indicate the underlying genetic cause. Prolonged PR interval or AV block is frequent in neuromuscular causes of NDLVC and in sarcoidosis. Laminopathies are characterized by prolonged PR interval, AF, and ventricular ectopics (VEs), and frequently show low voltage in pre-cordial leads.887 Depolarization abnormalities such as low QRS voltage are also a common finding in NDLVC caused by DSP and PLN variants.542 Ambulatory ECG monitoring is useful in NDLVC patients to reveal supraventricular and ventricular arrhythmias or bradycardias due to AV conduction block and is recommended at least yearly, or when there is a change in clinical status. In some patients with NDLVC at high risk of developing conduction disease and/or arrhythmias (including laminopathies, neuromuscular disease, PLN, and FLNC-truncating variants), Holter monitoring may be considered more frequently.

Recommendation Table 25

Recommendation for resting and ambulatory electrocardiogram monitoring in patients with non-dilated left ventricular cardiomyopathy

graphic
graphic
Recommendation Table 25

Recommendation for resting and ambulatory electrocardiogram monitoring in patients with non-dilated left ventricular cardiomyopathy

graphic
graphic
7.3.1.5. Echocardiography

Comprehensive TTE is recommended for all patients with NDLVC as it provides all relevant information on the global and regional LV anatomy, function, and haemodynamics; valvular heart disease; right heart function; pulmonary pressure; atrial geometry; and other features.71,73 Advanced echocardiographic techniques (including deformation imaging using tissue Doppler and speckle tracking) can allow the early detection of subclinical myocardial dysfunction in specific situations (e.g. genetic NDLVC carriers).71,74

7.3.1.6. Cardiac magnetic resonance

Cardiac magnetic resonance with LGE is the foremost imaging modality in NDLVC as it provides confirmation of the presence of non-ischaemic myocardial fibrosis that is essential for the diagnosis in most cases. Cardiac magnetic resonance can also be useful to detect the presence of myocardial oedema, which may suggest an inflammatory or myocarditic aetiology, and to describe the extent and pattern of fibrosis distribution. This can provide clues to the underlying aetiology (e.g. subepicardial distribution in post-myocarditis forms, patchy in sarcoidosis, extensive inferolateral in dystrophinopathies, septal mid-wall in LMNA carriers, and ring-like in DSP and FLNC variant carriers),71 and may provide additional prognostic value both for arrhythmia and heart failure severity.137,848

7.3.1.7. Nuclear medicine

The role of radionuclide imaging in NDLVC is limited. Measurement of 18F-FDG uptake using PET, with focal or focal-on-diffuse FDG uptake patterns, especially if concomitant abnormal 18F-FDG-PET uptake in extracardiac tissues, can be useful in suspected cardiac sarcoidosis.849 Isolated cardiac uptake has also been described in patients with NDLVC caused by DSP variants.888

7.3.1.8. Endomyocardial biopsy

Endomyocardial biopsy (EMB) with immunohistochemical quantification of inflammatory cells remains the gold standard investigation for the identification of cardiac inflammation. It may confirm the diagnosis of autoimmune disease in patients with unexplained heart failure and suspected giant cell myocarditis, eosinophilic myocarditis, vasculitis, and sarcoidosis. In experienced centres, electroanatomical voltage mapping-guided EMB may improve the yield of diagnosis of NDLVC.889 The risks and benefits of EMB should be evaluated and this procedure should be reserved for specific situations where its results may affect diagnosis or treatment (see Section 6.7.5).

7.3.2. Genetic testing

The genes most commonly implicated in NDLVC are DSP, FLNC (truncating variants), DES, LMNA, or PLN, but there is substantial overlap with the genetic background of both DCM and ARVC (Table 10). Desmoplakin (DSP) variants, in particular, cause a unique form of cardiomyopathy with a high prevalence of LV fibrosis and myocardial inflammatory episodes.864

The identification of a P/LP gene variant in a patient with NDLVC allows better prediction of the disease outcome and progression, may contribute to the indications for device implantation, informs genetic counselling, and allows familial screening for relatives (see Section 6.8.3). Therefore, genetic testing is recommended in all patients with NDLVC.

Recommendations for clinical screening, genetic counselling, and testing are described in Sections 6.8.3 and 6.11. Evaluation of conduction disease, and atrial and ventricular arrhythmia, is of particular importance in patients with NDLVC, as these may often be early phenotypic features. There are very few data on the natural history of phenotype-negative variant carriers or on the clinical yield of familial cascade screening in NDLVC, but cross-sectional studies suggest age-related increases in penetrance.9 Precautionary long-term evaluation of first-degree relatives is therefore recommended.

7.3.3. Assessment of symptoms

Most patients with NDLVC are asymptomatic, but some develop symptoms related to arrhythmia or conduction disease (e.g. syncope, palpitation) or diastolic heart failure (e.g. dyspnoea). Sustained ventricular arrhythmia, cardiac arrest, or SCD can be the initial presentation in a proportion of patients. Assessment of symptoms in patients with cardiomyopathies is described in Section 6.10.1.

7.3.4. Management

The clinical management of heart failure and other manifestations of NDLVC (atrial tachyarrhythmia, conduction disease) has been described in the 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure, the 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation, the 2021 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy, and the 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death,69,299,336,724 and are discussed in Section 6.10.2.

7.3.5. Sudden cardiac death prevention in non-dilated left ventricular cardiomyopathy

The prediction and prevention of SCD is the cornerstone of the clinical care of patients with NDLVC.

7.3.5.1. Secondary prevention of sudden cardiac death

As in other cardiomyopathy subtypes, ICD implantation is recommended in survivors of cardiac arrest and in patients who have experienced sustained ventricular arrhythmias with haemodynamic compromise;531 the decision to implant should consider the patient’s view and their QoL, as well as the absence of other diseases likely to cause death within 1 year.

7.3.5.2. Primary prevention of sudden cardiac death

There are no available RCTs examining the usefulness of ICDs to prevent SCD in patients with mild or moderate LV dysfunction. Recommendations for ICD implantation in DCM individuals with LVEF <35% are discussed in Section 6.10.2 and also apply to patients with NDLVC and LVEF <35%. Most patients with NDLVC, however, have either normal or mildly impaired LV systolic function. Much of the data on the natural history and risk prediction in NDLVC are derived from cohorts that include either patients with DCM or with ARVC (see Sections 7.2 and 7.4), and data on patients with NDLVC are therefore necessarily very limited. However, the available data suggest that genotype is a major determinant of SCD risk, with patients harbouring variants in PLN, TMEM43, DES, DSP, LMNA, FLNC (truncating variants), and RBM20 having a substantially higher rate of major arrhythmic events than other causes regardless of LVEF.440,542,864–869 For some high-risk genotypes (e.g. LMNA [https://lmna-risk-vta.fr]541), gene-specific (or, in the case of the PLN p.Arg14del variant, variant-specific [https://plnriskcalculator.shinyapps.io/final_shiny])542 risk-prediction scores have been developed that consider genotype characteristics and additional phenotypic features. Where such scores are available, they should be used to guide primary prevention ICD implantation (Figure 12). As discussed in Sections 7.1.5 and 7.2.5, the Task Force acknowledges the challenges associated with defining universal thresholds for acceptable risk across different cardiomyopathy phenotypes, but is of the opinion that a similar approach to that taken in risk stratification for HCM is reasonable, although the 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death have suggested a higher threshold of 10% risk at 5 years to guide primary prevention ICD implantation in patients with NDLVC and LMNA variants.3 This Task Force recommends more overt shared decision-making based on real-world data as well as individual preferences, beliefs, circumstances, and values. Gaps in evidence are acknowledged and should be shared with patients, and competing risks related to the disease (heart failure, stroke) and to age and comorbidity, as well as device-related complications, should be discussed.

There are very few data to guide risk stratification in patients with NDLVC without a known causative gene variant, but on the basis of the existing literature, the Task Force suggests that it may be reasonable to consider primary prevention ICD implantation in patients with NSVT, a family history of SCD, or significant LGE. Additional risk factors, such as the burden of VE, may also help guide ICD implantation, but there are no data currently to support a specific threshold for VE burden, and this will depend on the underlying genotype and other clinical factors.542,867,872 In patients with unexplained syncope, PES may provide additional information on the underlying cause.875 There are no definitive data supporting the regular use of PES for primary prevention risk stratification in patients with NDLVC, but may be beneficial in patients with NDLVC and myotonic dystrophy with an independent indication to EP study to assess conduction disturbances,876 although the clinical value of this approach has not been consistently demonstrated.877 Given the overlap with DCM and available data, and in keeping with the 2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death, the Task Force agreed that the recommendations for primary prevention ICD implantation in NDLVC should be the same as those for DCM (Figure 17), but the level of evidence is necessarily lower.

Recommendation Table 26

Recommendations for an implantable cardioverter defibrillator in patients with non-dilated left ventricular cardiomyopathy

graphic
graphic
Recommendation Table 26

Recommendations for an implantable cardioverter defibrillator in patients with non-dilated left ventricular cardiomyopathy

graphic
graphic

7.4. Arrhythmogenic right ventricular cardiomyopathy

7.4.1. Diagnosis

7.4.1.1. Index case

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized structurally by a progressive myocardial atrophy with fibro-fatty replacement of the RV myocardium.890 Lesions can also be present in the LV myocardium; predominant LV disease can coexist in the same family. Arrhythmogenic right ventricular cardiomyopathy usually manifests in the second to fourth decade of life.891 Men are affected more frequently than women and an age-related penetrance has been demonstrated, with high clinical and genetic variability.

An ARVC diagnosis should be suspected in adolescents or young adults with palpitations, syncope, or aborted sudden death. Frequent VEs or VT of LBBB morphology are among the most common clinical presentation. The presence of right pre-cordial TWI (V1–V3) in routine ECG testing should also be suspected for ARVC.10,892 Less common ECG changes include low QRS voltages in the peripheral leads and terminal activation delay in the right pre-cordial leads.893 Right ventricular dilatation on 2D echocardiography is also a frequent reason for patient referral. Less common presentations are RV or biventricular heart failure that can mimic DCM or NDLVC.894 Patients with multiple variants are thought to develop a more severe phenotype, and patients with a DSP or DSG2 variant are more prone to develop heart failure.895,896

In children and young adults, syncope, palpitations, and ventricular arrhythmias are also the usual presenting symptoms.897 However, chest pain, dynamic ST-T wave changes on basal 12-lead ECG, and myocardial enzymes release in the setting of normal coronary arteries are often reported, requiring differential diagnosis with myocarditis or acute myocardial infarction.898

7.4.1.2. Relatives

Clinical testing in relatives often reveals non-diagnostic abnormalities. In this context, the presence of RV systolic global or regional dysfunction, or additional electrocardiographic abnormalities (e.g. repolarization abnormalities, prolonged terminal activation duration, low QRS voltages, frequent ventricular extrasystoles [>500 per 24 h], or NSVT) in a first-degree relative of an individual with ARVC (or a first-degree relative with autopsy-proven ARVC) is highly suggestive of ARVC and warrants close follow-up.

7.4.1.3. Diagnostic work-up

The key elements of the diagnostic work-up for all patients with ARVC are defined by the diagnostic criteria used for the identification of affected individuals. The revised Task Force criteria for the diagnosis of ARVC published by Marcus et al. in 2010 have been used for the diagnosis of ARVC for more than a decade.10 More recently, the Padua criteria have offered an updated iteration to include LV involvement but are yet to be externally validated.5 Key elements of the diagnostic work-up include ECG, Holter monitoring, cardiac imaging, genetic testing, and, in specific circumstances, EMB.4,10,892 Additional laboratory tests, exercise testing, and cardiac catheterization should also be considered, as detailed in Section 6.

7.4.1.4. Electrocardiography and Holter monitoring

Abnormalities of the repolarization and depolarization as well as arrhythmias are key to the diagnosis of ARVC.5 The diagnostic utility of late potentials on signal-averaged electrocardiogram (SAECG) has been challenged in patients with ARVC for showing poor sensitivity and specificity.5,899 It has been noted that epsilon waves are frequently overdiagnosed and that there is poor agreement even between experts regarding their presence.900 Furthermore, it has been demonstrated that they occur in the presence of severe structural disease and thus add little to the diagnosis.900,901 Therefore, epsilon waves and SAECG should be utilized for diagnostic purposes with caution.

Recommendation Table 27

Recommendation for resting and ambulatory electrocardiogram monitoring in patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic
Recommendation Table 27

Recommendation for resting and ambulatory electrocardiogram monitoring in patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic
7.4.1.5. Echocardiography and cardiac magnetic resonance

A comprehensive cardiac imaging assessment is recommended for all ARVC patients.71,73 Structural and functional alterations assessed by echocardiography and CMR are key to ARVC diagnosis.10 The key feature is the presence of wall motional abnormalities such as RV akinesia, dyskinesia, or bulging, and the determinant of the diagnostic performance is the level of RV dilatation or dysfunction (major and minor criteria). Cardiac magnetic resonance should be considered the first-line test for assessment of the RV functional structural abnormalities criterion as it has demonstrated superior sensitivity.10 Contrast-enhanced CMR is the only tool allowing the detection of LV involvement which remains otherwise underestimated by applying the 2010 Task Force Criteria. Tissue characterization by CMR or indirectly by electroanatomical voltage mapping may show signs of fibro-fatty replacement that can be present in either ventricle.889,903,904

7.4.1.6. Endomyocardial biopsy

Differential diagnosis in patients with suspected ARVC includes inflammatory processes affecting the right ventricle such as myocarditis and sarcoidosis. In some instances, especially when dealing with probands with a sporadic form, EMB may be helpful to rule out myocarditis and sarcoidosis.72,892,905 Endomyocardial biopsy can also be useful in selected patients in whom non-invasive assessment is inconclusive.4,72 Electroanatomic voltage mapping-guided EMB may be considered in selected cases, particularly in case of negative CMR.906

7.4.1.7. Nuclear medicine

Measurement of 18F-FDG uptake using PET, with focal or focal-on-diffuse FDG uptake patterns, can be useful in suspected cardiac sarcoidosis.849 However, it has been demonstrated that patients with ARVC can also show myocardial 18F-FDG-PET uptake.888,907 Therefore, there is a limited role for radionuclide imaging in ARVC unless there is concomitant abnormal 18F-FDG-PET uptake in extracardiac tissues, or other clinical features suggestive of cardiac sarcoidosis.904,908

7.4.1.8. Arrhythmogenic right ventricular cardiomyopathy phenocopies

In suspicion of ARVC, a systematic approach to investigation of phenocopies should be undertaken. Differential diagnosis in patients with suspected ARVC includes myocarditis, sarcoidosis, RV infarction, DCM, Chagas disease, pulmonary hypertension, and CHD with volume overload (such as Ebstein anomaly, atrial septal defect, and partial anomalous venous return, left-to-right shunt, and pericardial agenesis).909,910 Disease phenocopies also include non-structural diseases. In fact, one of the main diagnostic dilemmas is to distinguish ARVC from idiopathic RV outflow tract VT, since the latter is usually benign.4 The idiopathic nature of VT is supported by the absence of family history, a normal basal 12-lead ECG, a normal ventricular structure by cardiac imaging and electroanatomic mapping, a single VT morphology, and the non-inducibility at programmed ventricular stimulation.

In highly trained competitive athletes, differential diagnosis with physiological adaptation to training needs to be considered.911 Right ventricular enlargement, ECG abnormalities, and arrhythmias reflect the increased haemodynamic load during exercise. While global RV systolic dysfunction and/or RWMAs, such as bulgings or aneurysms, are more in keeping with ARVC, the absence of overt structural changes of the right ventricle, frequent VEs, or inverted T waves in pre-cordial leads all support a benign nature (so-called athlete’s heart).72,912,913

7.4.2. Genetic testing and family screening

The genes underlying ARVC mainly encode proteins of the cardiac desmosome: plakophilin-2 (PKP2), desmoplakin (DSP), desmoglein-2 (DSG2), desmocollin-2 (DSC2), and plakoglobin (JUP). In addition to desmosomal genes, P/LP variants have also been described in other genes, including DES,914  TMEM43,915 and PLN.190,882 Pathogenic or likely pathogenic variants can be identified in up to 60% of patients with a diagnosis of ARVC.230 Given the diagnostic importance of genetic testing in ARVC, it is important that genetic variants are frequently re-appraised in terms of their pathogenicity.916 The pattern of inheritance in the majority of ARVC families is autosomal dominant. The penetrance of the disease in genetic carriers is age, gender, and physical activity dependent.892,917

Recommendations for clinical screening, genetic counselling, and testing are described in Sections 6.8.3 and 6.11. Cardiac evaluation should be adapted to the particular risk of complications in the family. Evaluation every 1–2 years including ECG, ECHO, and Holter/ECG monitoring is generally recommended for relatives at risk of developing the disease. Cardiac magnetic resonance should be considered at the baseline evaluation.

7.4.3. Assessment of symptoms

Patients with ARVC commonly experience palpitations and can develop symptoms of heart failure, although this may occur many years after the appearance of the initial abnormalities. Assessment of symptoms in patients with cardiomyopathies is described in Section 6.10.1.

7.4.4. Management

The aim of the clinical management of ARVC relies on the improvement of symptoms, the reduction of the pace of disease progression, and the prevention of complications. Recommendations for the pharmacological management of atrial arrhythmias and heart failure symptoms in patients with cardiomyopathies are described in Sections 6.10.2 and 6.10.3.

7.4.4.1. Antiarrhythmic therapy

Beta-blockers constitute the first option to reduce arrhythmic burden via a reduction in adrenergic tone, particularly on exercise. Titration to the maximal tolerated dose has been associated with an improvement in survival from major ventricular arrhythmias in retrospective observational studies.918

Amiodarone is often used when other beta-blockers fail to control arrhythmias.917,919,920 It should, however, be used with caution for the long-term management of ventricular arrhythmias, especially in young patients. Sotalol has been used for many years, but evidence regarding its efficacy remain limited and conflicting.921,922 Flecainide should be considered when single agent treatment has failed to control arrhythmia-related symptoms in patients with ARVC or when autonomic side effects limit the use of beta-blockers.923,924 Experience with other antiarrhythmics (dofetilide, ranolazine) is limited to very small case series.919,923

A proportion of patients require invasive arrhythmic procedures and/or ICD implantation. Complex endocardial and/or epicardial approach guided by three-dimensional (3D) electroanatomical mapping can be recommended but with a high recurrence rate (30–50% in experienced centres).919,925–927 Sympathetic denervation has also been used.928 Such procedures do not confer adequate protection against SCD, but may be very useful in reducing the VT burden and the risk of electrical storm.917 Discontinuation of intense physical exercise has shown a potential to slow the pace of disease progression and reduce the ventricular arrhythmia burden.917,919

Recommendation Table 28

Recommendations for the antiarrhythmic management of patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic
Recommendation Table 28

Recommendations for the antiarrhythmic management of patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic

7.4.5. Sudden cardiac death prevention in arrhythmogenic right ventricular cardiomyopathy

Arrhythmogenic right ventricular cardiomyopathy is characterized by its high propensity for ventricular arrhythmias and SCD.919 Although estimated to be a rare disease, it has been consistently reported as one of the most common causes of SCD in registries around the world.935–937 Sudden cardiac death seems to be more prevalent in young athletic individuals affected by the disease.935,938

7.4.5.1. Secondary prevention of sudden cardiac death

Implantable cardioverter defibrillators reduce mortality in survivors of cardiac arrest and in patients who have experienced sustained symptomatic ventricular arrhythmias.531 An ICD is recommended in such patients when the intent is to increase survival; the decision to implant should consider the patient’s view and their QoL, as well as the absence of other diseases likely to cause death within the following year.

7.4.5.2. Primary prevention of sudden cardiac death

Most of the current evidence on the outcomes of patients with ARVC and their predictors is limited to observational retrospective cohort studies that are typically of small size.939 Thus, the number of clinical predictors that can be studied using multivariate models is very limited, and most studies cannot be compared with one another. A systematic review and meta-analysis (n = 18 studies) has shown that the average risk of ventricular arrhythmia ranges from 3.7 to 10.6% per year and that male sex, RV dysfunction, and prior non-sustained or sustained VT/VF consistently predict ventricular arrhythmias in populations with ARVC.939

The first comprehensive effort to offer an approach to risk stratification in the context of decision-making for ICD implantation was made in the 2015 International Task Force consensus statement (ITFC) on the treatment of ARVC/dysplasia, where recommendations were made according to the presence of risk factors that would characterize the risk level of each patient.919 A follow-up study (n = 365) offered a modification on the International Task Force approach that resulted in better discrimination.940 The 2017 AHA/ACC/HRS guideline for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death941 and the 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy4 have also offered alternative approaches to this issue. A risk-prediction model was developed from a multicentre collaboration (n = 528); it utilizes sex, age, recent syncope, NSVT, VE count, number of leads with TWI, and right ventricular ejection fraction (RVEF) as predictors to provide an individualized estimate of sustained ventricular arrhythmias in patients with ARVC (arvcrisk.com).539

A study (n = 617) comparing the previous approaches to risk stratify patients has revealed that the modified ITFC approach provides the highest net benefit, up to an estimated 5-year risk of 25%, whereas AHA and HRS perform best in patients with an estimated 5-year risk >25%.538 In the same study, an estimated 5-year risk of 12.5% seems to be the optimal threshold, beyond which the risk-prediction model has the best performance. An external comparison (n = 140) of the different ARVC risk levels showed that the highest net benefit was seen with a 10% cut-off, using the 2019 ARVC risk calculator.536 In the same study, the 10% cut-off was superior to the HRS and ITFC approaches.536 Another external validation study (n = 128) of the 2019 ARVC risk model showed that although discriminative ability is excellent (c-index 0.84), the model seems to significantly overestimate the risk of patients below the 50% 5-year risk threshold.537 Recently, a correction to the 2019 ARVC risk calculator was issued.539 Two large external validation studies of the updated 2019 ARVC risk calculator have been published, suggesting a good discriminative performance, but the latter study revealed overestimation of risk.524,526 This raises concerns regarding the accuracy of the model in offering an individualized prediction that can help inform patients during decision-making; however, it can remain informative due to its excellent discriminative performance. Furthermore, one study suggested that the updated 2019 ARVC risk calculator performs best in PKP2 patients, but its performance is more limited in gene-negative individuals.524

Therefore, a combination of these approaches is recommended to individualize risk quantification that can aid clinicians in balancing the risks and benefits of ICD implantation. The final decision should be made together with the patient, considering other competing risks and the patient’s risk tolerance. As discussed in Section 7.1.5, the Task Force acknowledges the challenges associated with defining universal thresholds for acceptable risk across different cardiomyopathy phenotypes, but is of the opinion that a similar approach to that taken in risk stratification for HCM, DCM, and NDLVC is reasonable. In this context, the Task Force recommends shared decision-making based on real-world data as well as individual preferences, beliefs, circumstances, and values. Gaps in evidence should be shared with patients, and competing risks related to the disease (heart failure, stroke) and to age and comorbidity, as well as device-related complications, should be discussed. The suggested approach is summarized in Figure 18.

Algorithm to approach implantable cardioverter defibrillator decision-making in patients with arrhythmogenic right ventricular cardiomyopathy.
Figure 18

Algorithm to approach implantable cardioverter defibrillator decision-making in patients with arrhythmogenic right ventricular cardiomyopathy.

ARVC, arrhythmogenic right ventricular cardiomyopathy; ICD, implantable cardioverter defibrillator; LVEF, left ventricular ejection fraction; NSVT, non-sustained ventricular tachycardia; PES, programmed electrical stimulation; RVEF, right ventricular ejection fraction; SMVT, sustained monomorphic ventricular tachycardia; VT, ventricular tachycardia. aClinicians should aim to control ventricular arrhythmia with pharmacological or invasive antiarrhythmic therapies in addition to offering an ICD. bHigh-risk features are defined as either cardiac syncope, NSVT, RVEF <40%, LVEF <45%, SMVT at PES or as per the updated 2019 ARVC risk calculator.539

Patients with ARVC are known to suffer from sustained VTs that can be well tolerated without leading to SCD. Using appropriate ICD interventions as surrogate for SCD outcome has been shown to overestimate SCD.942 Considering that, in most centres, a high ratio of ARVC patients will be implanted with an ICD, it is conceivable why this may hamper risk stratification for SCD in patients with ARVC. Efforts to address this have been made within several studies,522,523,943–947 where the outcome of interest is fast VT (>250 b.p.m.) rather than any sustained VT. The largest of these studies (n = 864) has led to the development of a separate score for the prediction of unstable VT/VF.945 Due to the lack of any external validation studies, there is insufficient information to support the applicability of this risk score outside of its development cohorts. Furthermore, a specific rate cut-off is also not well evidence-based and its performance to predict SCD remains unclear. Although it is likely that slower sustained VTs per se are not life-threatening, it remains unknown how frequently they would degenerate to faster VTs or VF. It is therefore reasonable to suggest that all patients at risk of any sustained ventricular arrhythmia should be offered primary prevention ICDs.

The role of PES in risk stratification of ARVC patients is not well defined, particularly in those who are asymptomatic.523,939 However, current practice suggests that inducibility of SMVT at PES might add value in patients with symptoms consistent with sustained ventricular arrhythmia and this is further supported in this guideline3

Recommendation Table 29

Recommendations for sudden cardiac death prevention in patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic
Recommendation Table 29

Recommendations for sudden cardiac death prevention in patients with arrhythmogenic right ventricular cardiomyopathy

graphic
graphic

7.5. Restrictive cardiomyopathy

7.5.1. Diagnosis

Patients with overt RCM manifest signs and symptoms typical of HFpEF.306 The systematic approach to diagnosis should include clinical examination, ECG, echocardiography, and CMR.951 Physical examination may show a prominent jugular venous pulse. In the advanced phases, the pulse volume is low, the stroke volume declines, and the heart rate may increase. Hepatomegaly, ascites, and peripheral oedema are common in decompensated patients. Echocardiography is the gold standard diagnostic tool; criteria for diagnosing and grading diastolic dysfunction have been previously described.951,952 Importantly, the degree of diastolic dysfunction in patients with RCM is often only truly restrictive in advanced stages and most patients show milder grades of diastolic impairment at diagnosis.951 Cardiac catheterization should be performed in cases where the diagnosis is in doubt and to aid in the assessment for and timing of cardiac transplantation.953 Cardiac MRI distinguishes RCM from constrictive pericarditis, provides information on the presence and extent of myocardial fibrosis, and contributes to distinguishing metabolic from inflammatory diseases.951,954 Endomyocardial biopsy is a precision diagnostic tool in restrictive cardio-desminopathies;955 iron myocardial overload, both intramyocyte in haemochromatosis956 and mitochondrial in Friedreich ataxia cardiomyopathy;957 cystinosis;958 generalized arterial calcification of infancy;955,959 and lysosomal storage diseases (LSDs).960,961 Deep phenotyping in probands should go beyond cardiac traits and explore extracardiac manifestations in syndromic diseases and in RCM associated with neuromuscular disorders (see Section 6).962

7.5.2. Genetic testing

When inherited, RCM most commonly presents as an autosomal dominant disorder and, less commonly, autosomal recessive or sporadic. Genes associated with RCM encode sarcomeric structural and regulatory proteins and cytoskeletal intermediate filaments (Table 10). Although all major sarcomeric genes may cause RCM,963 the most common disease gene is TNNI3, which encodes the thin filament troponin I.964 Other less commonly involved genes include TNNT2, ACTC1, MYH7, MYBPC3, TTN, TPM1, MYPN, MYL3, and MYL2. Restrictive cardiomyopathy can be associated with intramyocyte accumulation of unfolded defective proteins, a feature that is increasingly demonstrated in carriers of defects in DES, FLNC, and BAG3. These diseases have significant implications for prognosis and timely decision-making, both in children and adults. Restrictive cardiomyopathy may also occur in individuals with a family history of HCM289 or DCM.965 The observation of different cardiomyopathy phenotypes within families suggests a variable response to the variant, and implicates factors beyond the specific variant in the determination of ultimate clinical manifestation of disease.966 Hereditary infiltrative diseases can also cause RCM, the most common of which is amyloidosis caused by pathogenic variants in the TTR gene, although this is usually in the presence of LVH (see Section 7.7).

7.5.3. Assessment of symptoms

Patients with RCM often develop symptoms of heart failure, although this can occur some years after the appearance of the initial abnormalities. Assessment of symptoms in patients with cardiomyopathies is described in Section 6.10.1.

7.5.4. Management

The administration of heart failure medications and device implantation, including ventricular assist device as a bridge-to-candidacy is guided by symptoms and heart failure phenotype and severity,967 and is described in Section 6.10.2. Precision diagnosis (phenotype and cause) is key to timely planning of heart transplantation as it guarantees the exclusion of all genetic and acquired phenocopies that may be amenable to alternative treatment. Prevention of heart transplantation in all RCM patients with alternative treatments is a major goal for all adult and paediatric RCM.

Precise diagnosis is also essential for genetic phenocopies with available target treatments: ERT for Anderson–Fabry disease or glycogenosis such as Pompe disease; therapeutic phlebotomy for haemochromatosis; immunosuppressive therapeutics for sarcoidosis; new biological drugs for systemic diseases (e.g. autoimmune diseases with cardiac involvement that can reverse or stabilize by treating the disease itself); and removal of the toxic causes (see Figure 19 and Supplementary data online, Table S4). Precision diagnosis today is essential due to the increasing availability of disease-specific treatments and diagnostic tools to exclude geno/phenocopies.

Spectrum of restrictive heart diseases.
Figure 19

Spectrum of restrictive heart diseases.

AFD, Anderson–Fabry disease; LVH, left ventricular hypertrophy; PRKAG2, Protein kinase AMP-activated non-catalytic subunit gamma 2; RCM, restrictive cardiomyopathy. For a more detailed spectrum of restrictive heart disease, please refer to Supplementary data online, Table S4.

Restrictive cardiomyopathy is associated with the worst prognosis of all the cardiomyopathy phenotypes. Survival data are limited to small windows of observation. The prognosis of RCM largely depends on the restrictive physiology, regardless of the underlying cause.968–971 More than 50% of children with RCM are at risk of death (including SCD) or transplantation shortly after diagnosis; clinical features putatively associated with increased risk of death or transplantation include: heart failure symptoms; reduced LV systolic function; increased left atrial size; syncope; ischaemia; and impaired LV diastolic function on echocardiography.286,969,972,973 Up to 75% of surviving patients demonstrate heart failure, and the outcome is either death or heart transplantation within a few years of diagnosis.968,969 Elevated pulmonary vascular resistance (PVR) is present in up to 40% of children with RCM, and can rise quickly even in the absence of other clinical changes, which has an impact on suitability for and timing of cardiac transplantation.953 Cardiac catheterization with an assessment of PVR is therefore recommended in all children at diagnosis and every 6 to 12 months.953 In adult patients with genetic RCM, the main cause of death is heart failure (more than 40%), with a 5-year survival rate of ∼50% in cohorts that include patients with HCM and restrictive physiology.616

Recommendation Table 30

Recommendations for the management of patients with restrictive cardiomyopathy

graphic
graphic
Recommendation Table 30

Recommendations for the management of patients with restrictive cardiomyopathy

graphic
graphic

7.6. Syndromic and metabolic cardiomyopathies

It is beyond the scope of these guidelines to provide a detailed review and recommendations on specific cardiomyopathy genocopies and phenocopies. Instead, the Task Force refers the reader to detailed position statements and consensus documents published on behalf of the ESC Working Group on Myocardial and Pericardial Diseases (e.g. on Anderson–Fabry Disease and amyloidosis ).370,375,974 This section highlights only the key diagnostic and management issues. Table 22 summarizes the clinical features and management of syndromic and metabolic cardiomyopathies.

Table 22

Clinical features and management of syndromic and metabolic cardiomyopathies

Clinical red flagsDiagnosisSpecific causeMultidisciplinary teamManagement
Abnormal facial features
Cryptorchidism
Pulmonary valve stenosis
Congenital heart disease
Extreme right-axis deviation at ECG
Lymphangectasis
Bleeding diathesis
Café au lait spots
Lentigines
Growth retardation
Sensorineural deafness
NGS panel testing for RASopathyNoonan syndrome
Costello syndrome
Cardiofaciocutaneous syndrome
Noonan syndrome with multiple lentigines
Cardiologist
Geneticist
Endocrinologist
Paediatrician
Dermatologist
Radiologist
Beta-blockers/CCBs
Selective management of RVOTO/pulmonary valvuloplasty
SCD risk stratification
Short PR interval
End-stage, hypokynetic HCM
AV block (Kearns–Sayre syndrome)
Lactic acidosis
Sensorineural deafness
Neutropenia (Barth syndrome)
Diabetes
Stroke-like lesions at brain MRI
NGS panel for mtDNA and nuclear DNA
Skeletal muscle biopsy/endomyocardial biopsy
MELAS syndrome
MERRF syndrome
Leigh syndrome
Other mitochondrial disease
Beta-oxidation disorders
Cardiologist
Neurologist
Endocrinologist
Paediatrician
Metabolism expert
Radiologist
Avoiding drugs or situational stressors
Beta-oxidation disorders: nutritional management, avoidance of fasting, aggressive treatment during increased metabolic stress
Carnitine supplementation (selected cases)
Hepatomegaly
Increased aminotransferase enzymes
Delayed motor milestones
Hypotonia
Short PR interval
ECG criteria for extreme LVH
Screening: GAA activity in DBS or leucocytes/Glc4 dosing
Diagnostic confirmation: acid alpha-glucosidase assay performed on skin fibroblasts (preferred method) or muscle biopsy
Type II glycogen storage disease (Pompe disease)Cardiologist
General Paediatrician/neonatologist
Gastroenterologist
Neuromuscular disease specialist
Enzyme replacement therapy
Short PR interval
Massive LVH
Skeletal myopathy
Increased serum CK enzyme
Intellectual disability
X-linked inheritance
NGS or target testing for LAMP-2 variantsDanon diseaseCardiologist
Neuromuscular disease specialist
Pneumologist
Advanced heart failure specialist
No treatment
Short PR interval
Early-onset atrial fibrillation
AV block
Increased serum CK enzyme
Autosomal dominant inheritance pattern
NGS or target testing for PRKAG2PRKAG2 syndromeCardiologist
Neuromuscular disease expert
No treatment
Progressive limb ataxia
Diabetes mellitus
Pes cavus
Reduced native T1 at CMR imaging
NGS testing for bi-allelic expansion of GAA repeats in the FXN geneFriedreich ataxiaCardiologist
Neurologist
Endocrinologist
Orthopaedic surgeon
Neuromuscular disease expert
No specific treatment
Bilateral carpal tunnel syndrome
Lumbar spinal stenosis
Autonomic dysfunction
Peripheral neuropathy
Relative apical sparing pattern
Ejection fraction/strain ratio >5
Pseudonecrosis Q waves
Low ECG voltages
OR
Positive serum or urine monoclonal chain at immunofixation
DPD/HMDP Tc99 scintigraphy
Free light chain/serum and urine immunofixation
Endomyocardial biopsy
Cardiac amyloidosis (AL or ATTR) (see Section 7.7)Cardiologist
Neurologist
Nephrologist
Haematologist (AL amyloidosis)
Ophthalmologist
Tafamidis
Patisirana
Inotersena
(ATTR-CA)
OR
Specific chemotherapy
(AL amyloidosis)
Gastrointestinal symptoms
Angiokeratoma
Cornea verticillata
Chronic kidney disease
Proteinuria
Sensorineural hypoacusia
Stroke/TIA
Neuropathic pain
X-linked inheritance pattern
Short PR interval
Low native T1 at cardiac CMR
Screening in males: lyso-Gb3 dosing
Screening in females/diagnostic confirmation: genetic testing for GLA variants
Anderson–Fabry diseaseCardiologist
Nephrologist
Neurologist
Ophthalmologist
Audiologist
Gastroenterologist
Dermatologist
Enzyme replacement therapy (agalsidase alfa/beta)
Migalastat
Skeletal myopathy
Posterolateral pseudonecrosis pattern
Posterolateral or inferolateral akinesia
Genetic testing for dystrophinopathiesDMDNeurologist
Cardiologist
Pneumologist
Neuromuscular disease expert
Steroids (prednisone or deflazacort)
Skeletal myopathy
AV block
Premature atrial fibrillation
Malignant ventricular arrhythmias
NGS testingLMNA cardiomyopathy
Emery–Dreifuss muscular dystrophy
Cardiologist
Neurologist
SCD risk prevention
Pacing if indicated
Bilateral hilar lymphadenopathy
Pulmonary infiltrates
Uveitis
Gastrointestinal involvement
High-degree AV block
Frequent VEs
Thinned basal interventricular septum
Extended LGE at CMR imaging
18F-FDG-PET
Endomyocardial biopsy
Lung biopsy
SarcoidosisCardiologist
Pneumologist
Neurologist
Gastroenterologist
Steroids
Steroid-sparing immunosuppressant drugs
Previous transfusions
Chronic liver disease
Skin pigmentation
Diabetes
Hypogonadotropic hypogonadism
Elevated ferritin
AV block
Iron status
Complete blood count
Increased T2* values at CMR imaging
Genetic test for HFE, HJV, hepcidin receptor, ferroportin, HAMP gene
Peripheral blood smear
Haemoglobin electrophoresis
Genetic testing for hereditary haemoglobinopathies
Iron overload cardiomyopathyCardiologist
Haematologist
Endocrinologist
Paediatrician
Gastroenterologist
Iron-chelating drugs
Phlebotomy
Clinical red flagsDiagnosisSpecific causeMultidisciplinary teamManagement
Abnormal facial features
Cryptorchidism
Pulmonary valve stenosis
Congenital heart disease
Extreme right-axis deviation at ECG
Lymphangectasis
Bleeding diathesis
Café au lait spots
Lentigines
Growth retardation
Sensorineural deafness
NGS panel testing for RASopathyNoonan syndrome
Costello syndrome
Cardiofaciocutaneous syndrome
Noonan syndrome with multiple lentigines
Cardiologist
Geneticist
Endocrinologist
Paediatrician
Dermatologist
Radiologist
Beta-blockers/CCBs
Selective management of RVOTO/pulmonary valvuloplasty
SCD risk stratification
Short PR interval
End-stage, hypokynetic HCM
AV block (Kearns–Sayre syndrome)
Lactic acidosis
Sensorineural deafness
Neutropenia (Barth syndrome)
Diabetes
Stroke-like lesions at brain MRI
NGS panel for mtDNA and nuclear DNA
Skeletal muscle biopsy/endomyocardial biopsy
MELAS syndrome
MERRF syndrome
Leigh syndrome
Other mitochondrial disease
Beta-oxidation disorders
Cardiologist
Neurologist
Endocrinologist
Paediatrician
Metabolism expert
Radiologist
Avoiding drugs or situational stressors
Beta-oxidation disorders: nutritional management, avoidance of fasting, aggressive treatment during increased metabolic stress
Carnitine supplementation (selected cases)
Hepatomegaly
Increased aminotransferase enzymes
Delayed motor milestones
Hypotonia
Short PR interval
ECG criteria for extreme LVH
Screening: GAA activity in DBS or leucocytes/Glc4 dosing
Diagnostic confirmation: acid alpha-glucosidase assay performed on skin fibroblasts (preferred method) or muscle biopsy
Type II glycogen storage disease (Pompe disease)Cardiologist
General Paediatrician/neonatologist
Gastroenterologist
Neuromuscular disease specialist
Enzyme replacement therapy
Short PR interval
Massive LVH
Skeletal myopathy
Increased serum CK enzyme
Intellectual disability
X-linked inheritance
NGS or target testing for LAMP-2 variantsDanon diseaseCardiologist
Neuromuscular disease specialist
Pneumologist
Advanced heart failure specialist
No treatment
Short PR interval
Early-onset atrial fibrillation
AV block
Increased serum CK enzyme
Autosomal dominant inheritance pattern
NGS or target testing for PRKAG2PRKAG2 syndromeCardiologist
Neuromuscular disease expert
No treatment
Progressive limb ataxia
Diabetes mellitus
Pes cavus
Reduced native T1 at CMR imaging
NGS testing for bi-allelic expansion of GAA repeats in the FXN geneFriedreich ataxiaCardiologist
Neurologist
Endocrinologist
Orthopaedic surgeon
Neuromuscular disease expert
No specific treatment
Bilateral carpal tunnel syndrome
Lumbar spinal stenosis
Autonomic dysfunction
Peripheral neuropathy
Relative apical sparing pattern
Ejection fraction/strain ratio >5
Pseudonecrosis Q waves
Low ECG voltages
OR
Positive serum or urine monoclonal chain at immunofixation
DPD/HMDP Tc99 scintigraphy
Free light chain/serum and urine immunofixation
Endomyocardial biopsy
Cardiac amyloidosis (AL or ATTR) (see Section 7.7)Cardiologist
Neurologist
Nephrologist
Haematologist (AL amyloidosis)
Ophthalmologist
Tafamidis
Patisirana
Inotersena
(ATTR-CA)
OR
Specific chemotherapy
(AL amyloidosis)
Gastrointestinal symptoms
Angiokeratoma
Cornea verticillata
Chronic kidney disease
Proteinuria
Sensorineural hypoacusia
Stroke/TIA
Neuropathic pain
X-linked inheritance pattern
Short PR interval
Low native T1 at cardiac CMR
Screening in males: lyso-Gb3 dosing
Screening in females/diagnostic confirmation: genetic testing for GLA variants
Anderson–Fabry diseaseCardiologist
Nephrologist
Neurologist
Ophthalmologist
Audiologist
Gastroenterologist
Dermatologist
Enzyme replacement therapy (agalsidase alfa/beta)
Migalastat
Skeletal myopathy
Posterolateral pseudonecrosis pattern
Posterolateral or inferolateral akinesia
Genetic testing for dystrophinopathiesDMDNeurologist
Cardiologist
Pneumologist
Neuromuscular disease expert
Steroids (prednisone or deflazacort)
Skeletal myopathy
AV block
Premature atrial fibrillation
Malignant ventricular arrhythmias
NGS testingLMNA cardiomyopathy
Emery–Dreifuss muscular dystrophy
Cardiologist
Neurologist
SCD risk prevention
Pacing if indicated
Bilateral hilar lymphadenopathy
Pulmonary infiltrates
Uveitis
Gastrointestinal involvement
High-degree AV block
Frequent VEs
Thinned basal interventricular septum
Extended LGE at CMR imaging
18F-FDG-PET
Endomyocardial biopsy
Lung biopsy
SarcoidosisCardiologist
Pneumologist
Neurologist
Gastroenterologist
Steroids
Steroid-sparing immunosuppressant drugs
Previous transfusions
Chronic liver disease
Skin pigmentation
Diabetes
Hypogonadotropic hypogonadism
Elevated ferritin
AV block
Iron status
Complete blood count
Increased T2* values at CMR imaging
Genetic test for HFE, HJV, hepcidin receptor, ferroportin, HAMP gene
Peripheral blood smear
Haemoglobin electrophoresis
Genetic testing for hereditary haemoglobinopathies
Iron overload cardiomyopathyCardiologist
Haematologist
Endocrinologist
Paediatrician
Gastroenterologist
Iron-chelating drugs
Phlebotomy

18F-FDG-PET, 18F-fluorodeoxyglucose positron emission tomography; AL, amyloid light chain; ATTR, transthyretin amyloidosis; ATTR-CA: transthyretin cardiac amyloidosis; AV, atrioventricular; CCB, calcium channel blocker; CK, creatinine kinase; CMR, cardiac magnetic resonance; DBS, deep brain stimulation; DMD, Duchenne muscular dystrophy; DPD, 3,3-diphosphono-1,2-propanodicarboxylic acid; ECG, electrocardiogram; Gb3, globotriaosylceramide; HCM, hypertrophic cardiomyopathy; HMDP, hydroxymethylene diphosphonate; LGE, late gadolinium enhancement; LMNA, lamin A/C; LVH, left ventricular hypertrophy; MELAS, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (syndrome); MERRF, mitochondrial epilepsy with ragged-red fibres; MRI, magnetic resonance; mtDNA, mitochondrial DNA; NGS, next-generation sequencing; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; RVOTO, right ventricular outflow tract obstruction; SCD, sudden cardiac death; TIA, transient ischaemic attack; VE, ventricular ectopic beats.

Patisiran and inotersen approved for treatment of familial polyneuropathy with/without cardiomyopathy.

Table 22

Clinical features and management of syndromic and metabolic cardiomyopathies

Clinical red flagsDiagnosisSpecific causeMultidisciplinary teamManagement
Abnormal facial features
Cryptorchidism
Pulmonary valve stenosis
Congenital heart disease
Extreme right-axis deviation at ECG
Lymphangectasis
Bleeding diathesis
Café au lait spots
Lentigines
Growth retardation
Sensorineural deafness
NGS panel testing for RASopathyNoonan syndrome
Costello syndrome
Cardiofaciocutaneous syndrome
Noonan syndrome with multiple lentigines
Cardiologist
Geneticist
Endocrinologist
Paediatrician
Dermatologist
Radiologist
Beta-blockers/CCBs
Selective management of RVOTO/pulmonary valvuloplasty
SCD risk stratification
Short PR interval
End-stage, hypokynetic HCM
AV block (Kearns–Sayre syndrome)
Lactic acidosis
Sensorineural deafness
Neutropenia (Barth syndrome)
Diabetes
Stroke-like lesions at brain MRI
NGS panel for mtDNA and nuclear DNA
Skeletal muscle biopsy/endomyocardial biopsy
MELAS syndrome
MERRF syndrome
Leigh syndrome
Other mitochondrial disease
Beta-oxidation disorders
Cardiologist
Neurologist
Endocrinologist
Paediatrician
Metabolism expert
Radiologist
Avoiding drugs or situational stressors
Beta-oxidation disorders: nutritional management, avoidance of fasting, aggressive treatment during increased metabolic stress
Carnitine supplementation (selected cases)
Hepatomegaly
Increased aminotransferase enzymes
Delayed motor milestones
Hypotonia
Short PR interval
ECG criteria for extreme LVH
Screening: GAA activity in DBS or leucocytes/Glc4 dosing
Diagnostic confirmation: acid alpha-glucosidase assay performed on skin fibroblasts (preferred method) or muscle biopsy
Type II glycogen storage disease (Pompe disease)Cardiologist
General Paediatrician/neonatologist
Gastroenterologist
Neuromuscular disease specialist
Enzyme replacement therapy
Short PR interval
Massive LVH
Skeletal myopathy
Increased serum CK enzyme
Intellectual disability
X-linked inheritance
NGS or target testing for LAMP-2 variantsDanon diseaseCardiologist
Neuromuscular disease specialist
Pneumologist
Advanced heart failure specialist
No treatment
Short PR interval
Early-onset atrial fibrillation
AV block
Increased serum CK enzyme
Autosomal dominant inheritance pattern
NGS or target testing for PRKAG2PRKAG2 syndromeCardiologist
Neuromuscular disease expert
No treatment
Progressive limb ataxia
Diabetes mellitus
Pes cavus
Reduced native T1 at CMR imaging
NGS testing for bi-allelic expansion of GAA repeats in the FXN geneFriedreich ataxiaCardiologist
Neurologist
Endocrinologist
Orthopaedic surgeon
Neuromuscular disease expert
No specific treatment
Bilateral carpal tunnel syndrome
Lumbar spinal stenosis
Autonomic dysfunction
Peripheral neuropathy
Relative apical sparing pattern
Ejection fraction/strain ratio >5
Pseudonecrosis Q waves
Low ECG voltages
OR
Positive serum or urine monoclonal chain at immunofixation
DPD/HMDP Tc99 scintigraphy
Free light chain/serum and urine immunofixation
Endomyocardial biopsy
Cardiac amyloidosis (AL or ATTR) (see Section 7.7)Cardiologist
Neurologist
Nephrologist
Haematologist (AL amyloidosis)
Ophthalmologist
Tafamidis
Patisirana
Inotersena
(ATTR-CA)
OR
Specific chemotherapy
(AL amyloidosis)
Gastrointestinal symptoms
Angiokeratoma
Cornea verticillata
Chronic kidney disease
Proteinuria
Sensorineural hypoacusia
Stroke/TIA
Neuropathic pain
X-linked inheritance pattern
Short PR interval
Low native T1 at cardiac CMR
Screening in males: lyso-Gb3 dosing
Screening in females/diagnostic confirmation: genetic testing for GLA variants
Anderson–Fabry diseaseCardiologist
Nephrologist
Neurologist
Ophthalmologist
Audiologist
Gastroenterologist
Dermatologist
Enzyme replacement therapy (agalsidase alfa/beta)
Migalastat
Skeletal myopathy
Posterolateral pseudonecrosis pattern
Posterolateral or inferolateral akinesia
Genetic testing for dystrophinopathiesDMDNeurologist
Cardiologist
Pneumologist
Neuromuscular disease expert
Steroids (prednisone or deflazacort)
Skeletal myopathy
AV block
Premature atrial fibrillation
Malignant ventricular arrhythmias
NGS testingLMNA cardiomyopathy
Emery–Dreifuss muscular dystrophy
Cardiologist
Neurologist
SCD risk prevention
Pacing if indicated
Bilateral hilar lymphadenopathy
Pulmonary infiltrates
Uveitis
Gastrointestinal involvement
High-degree AV block
Frequent VEs
Thinned basal interventricular septum
Extended LGE at CMR imaging
18F-FDG-PET
Endomyocardial biopsy
Lung biopsy
SarcoidosisCardiologist
Pneumologist
Neurologist
Gastroenterologist
Steroids
Steroid-sparing immunosuppressant drugs
Previous transfusions
Chronic liver disease
Skin pigmentation
Diabetes
Hypogonadotropic hypogonadism
Elevated ferritin
AV block
Iron status
Complete blood count
Increased T2* values at CMR imaging
Genetic test for HFE, HJV, hepcidin receptor, ferroportin, HAMP gene
Peripheral blood smear
Haemoglobin electrophoresis
Genetic testing for hereditary haemoglobinopathies
Iron overload cardiomyopathyCardiologist
Haematologist
Endocrinologist
Paediatrician
Gastroenterologist
Iron-chelating drugs
Phlebotomy
Clinical red flagsDiagnosisSpecific causeMultidisciplinary teamManagement
Abnormal facial features
Cryptorchidism
Pulmonary valve stenosis
Congenital heart disease
Extreme right-axis deviation at ECG
Lymphangectasis
Bleeding diathesis
Café au lait spots
Lentigines
Growth retardation
Sensorineural deafness
NGS panel testing for RASopathyNoonan syndrome
Costello syndrome
Cardiofaciocutaneous syndrome
Noonan syndrome with multiple lentigines
Cardiologist
Geneticist
Endocrinologist
Paediatrician
Dermatologist
Radiologist
Beta-blockers/CCBs
Selective management of RVOTO/pulmonary valvuloplasty
SCD risk stratification
Short PR interval
End-stage, hypokynetic HCM
AV block (Kearns–Sayre syndrome)
Lactic acidosis
Sensorineural deafness
Neutropenia (Barth syndrome)
Diabetes
Stroke-like lesions at brain MRI
NGS panel for mtDNA and nuclear DNA
Skeletal muscle biopsy/endomyocardial biopsy
MELAS syndrome
MERRF syndrome
Leigh syndrome
Other mitochondrial disease
Beta-oxidation disorders
Cardiologist
Neurologist
Endocrinologist
Paediatrician
Metabolism expert
Radiologist
Avoiding drugs or situational stressors
Beta-oxidation disorders: nutritional management, avoidance of fasting, aggressive treatment during increased metabolic stress
Carnitine supplementation (selected cases)
Hepatomegaly
Increased aminotransferase enzymes
Delayed motor milestones
Hypotonia
Short PR interval
ECG criteria for extreme LVH
Screening: GAA activity in DBS or leucocytes/Glc4 dosing
Diagnostic confirmation: acid alpha-glucosidase assay performed on skin fibroblasts (preferred method) or muscle biopsy
Type II glycogen storage disease (Pompe disease)Cardiologist
General Paediatrician/neonatologist
Gastroenterologist
Neuromuscular disease specialist
Enzyme replacement therapy
Short PR interval
Massive LVH
Skeletal myopathy
Increased serum CK enzyme
Intellectual disability
X-linked inheritance
NGS or target testing for LAMP-2 variantsDanon diseaseCardiologist
Neuromuscular disease specialist
Pneumologist
Advanced heart failure specialist
No treatment
Short PR interval
Early-onset atrial fibrillation
AV block
Increased serum CK enzyme
Autosomal dominant inheritance pattern
NGS or target testing for PRKAG2PRKAG2 syndromeCardiologist
Neuromuscular disease expert
No treatment
Progressive limb ataxia
Diabetes mellitus
Pes cavus
Reduced native T1 at CMR imaging
NGS testing for bi-allelic expansion of GAA repeats in the FXN geneFriedreich ataxiaCardiologist
Neurologist
Endocrinologist
Orthopaedic surgeon
Neuromuscular disease expert
No specific treatment
Bilateral carpal tunnel syndrome
Lumbar spinal stenosis
Autonomic dysfunction
Peripheral neuropathy
Relative apical sparing pattern
Ejection fraction/strain ratio >5
Pseudonecrosis Q waves
Low ECG voltages
OR
Positive serum or urine monoclonal chain at immunofixation
DPD/HMDP Tc99 scintigraphy
Free light chain/serum and urine immunofixation
Endomyocardial biopsy
Cardiac amyloidosis (AL or ATTR) (see Section 7.7)Cardiologist
Neurologist
Nephrologist
Haematologist (AL amyloidosis)
Ophthalmologist
Tafamidis
Patisirana
Inotersena
(ATTR-CA)
OR
Specific chemotherapy
(AL amyloidosis)
Gastrointestinal symptoms
Angiokeratoma
Cornea verticillata
Chronic kidney disease
Proteinuria
Sensorineural hypoacusia
Stroke/TIA
Neuropathic pain
X-linked inheritance pattern
Short PR interval
Low native T1 at cardiac CMR
Screening in males: lyso-Gb3 dosing
Screening in females/diagnostic confirmation: genetic testing for GLA variants
Anderson–Fabry diseaseCardiologist
Nephrologist
Neurologist
Ophthalmologist
Audiologist
Gastroenterologist
Dermatologist
Enzyme replacement therapy (agalsidase alfa/beta)
Migalastat
Skeletal myopathy
Posterolateral pseudonecrosis pattern
Posterolateral or inferolateral akinesia
Genetic testing for dystrophinopathiesDMDNeurologist
Cardiologist
Pneumologist
Neuromuscular disease expert
Steroids (prednisone or deflazacort)
Skeletal myopathy
AV block
Premature atrial fibrillation
Malignant ventricular arrhythmias
NGS testingLMNA cardiomyopathy
Emery–Dreifuss muscular dystrophy
Cardiologist
Neurologist
SCD risk prevention
Pacing if indicated
Bilateral hilar lymphadenopathy
Pulmonary infiltrates
Uveitis
Gastrointestinal involvement
High-degree AV block
Frequent VEs
Thinned basal interventricular septum
Extended LGE at CMR imaging
18F-FDG-PET
Endomyocardial biopsy
Lung biopsy
SarcoidosisCardiologist
Pneumologist
Neurologist
Gastroenterologist
Steroids
Steroid-sparing immunosuppressant drugs
Previous transfusions
Chronic liver disease
Skin pigmentation
Diabetes
Hypogonadotropic hypogonadism
Elevated ferritin
AV block
Iron status
Complete blood count
Increased T2* values at CMR imaging
Genetic test for HFE, HJV, hepcidin receptor, ferroportin, HAMP gene
Peripheral blood smear
Haemoglobin electrophoresis
Genetic testing for hereditary haemoglobinopathies
Iron overload cardiomyopathyCardiologist
Haematologist
Endocrinologist
Paediatrician
Gastroenterologist
Iron-chelating drugs
Phlebotomy

18F-FDG-PET, 18F-fluorodeoxyglucose positron emission tomography; AL, amyloid light chain; ATTR, transthyretin amyloidosis; ATTR-CA: transthyretin cardiac amyloidosis; AV, atrioventricular; CCB, calcium channel blocker; CK, creatinine kinase; CMR, cardiac magnetic resonance; DBS, deep brain stimulation; DMD, Duchenne muscular dystrophy; DPD, 3,3-diphosphono-1,2-propanodicarboxylic acid; ECG, electrocardiogram; Gb3, globotriaosylceramide; HCM, hypertrophic cardiomyopathy; HMDP, hydroxymethylene diphosphonate; LGE, late gadolinium enhancement; LMNA, lamin A/C; LVH, left ventricular hypertrophy; MELAS, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (syndrome); MERRF, mitochondrial epilepsy with ragged-red fibres; MRI, magnetic resonance; mtDNA, mitochondrial DNA; NGS, next-generation sequencing; PRKAG2, protein kinase AMP-activated non-catalytic subunit gamma 2; RVOTO, right ventricular outflow tract obstruction; SCD, sudden cardiac death; TIA, transient ischaemic attack; VE, ventricular ectopic beats.

Patisiran and inotersen approved for treatment of familial polyneuropathy with/without cardiomyopathy.

7.6.1. Anderson–Fabry disease

7.6.1.1. Definition

Anderson–Fabry disease is an inborn error of metabolism where a deficient or absent enzyme, alpha-galactosidase A (α-Gal A), due to a pathogenic genetic variant in the GLA gene, causes the storage of some degradation cell products, mainly globotriaosylceramide (Gb3) in a patient’s lysosomes.975 This storage causes cell dysfunction in its own right and activates cellular hypertrophy pathways, common to other causes of HCM, as well as inflammation and immune activation.976 It is a multisystem disorder affecting particularly the heart, kidney, and brain. 975 It is inherited in an X-linked manner; males are therefore always affected, while females’ organ involvement usually develops later in life but can become similar to males due to the lyonization phenomena.977,978

Two Anderson–Fabry phenotypes can be distinguished, depending on the gender, lyonization phenomena, and pathogenic genetic variant:976,979

  • A severe clinical phenotype, known as ‘classic’ Anderson–Fabry characterized by absent or severely reduced (<1% of mean normal) α-Gal A activity, marked Gb3 accumulation, and childhood or adolescent onset of symptoms followed by progressive multiorgan failure, is most often seen in males (but not exclusively) without residual enzyme activity.

  • A non-classical’ Anderson–Fabry phenotype or later-onset phenotype with incomplete systemic involvement, which is seen in both males and females, with some level of residual enzyme activity, and in most cases manifesting as isolated cardiac involvement.

7.6.1.2. Diagnosis, clinical work-up, and differential diagnosis

Anderson–Fabry disease should be suspected in patients with LVH and additional cardiac and extracardiac red flags (see Table 23) sought (Figure 20). The diagnosis is established by assessment of α-GalA activity and lyso-Gb3 measurement in male patients; in females, genetic testing is usually required to confirm the diagnosis. Severe LVH (>15 mm) is unlikely to be seen in patients <20 years of age.980 In children and adolescents, diagnosis is made by family history or based on other extracardiac symptoms, but overt LVH is usually not present.981

Anderson–Fabry disease diagnostic algorithm.
Figure 20

Anderson–Fabry disease diagnostic algorithm.

α-Gal A, alpha-galactosidase A; AFD, Anderson–Fabry disease; CMR, cardiac magnetic resonance; Gb3, globotriaosylceramide; HCM, hypertrophic cardiomyopathy; LVH, left ventricular hypertrophy; lyso Gb3, globotriaosylsphingosine; P/LP, pathogenic/likely pathogenic; VUS, variant of unknown significance. aSee Table 23. bGenetic analysis must include the study of possible large deletions or a copy number variation not detected by the Sanger method. cThe finding of increased plasma and/or urinary Gb3, or plasma lyso Gb3 and its analogues in the evaluation of male or female patients with a VUS and normal (in female patients) or lowered α-Gal A activity provides additional diagnostic information, but the role of biomarkers in such patients still requires validation. dLow native T1 values reinforce or generate suspicion of Fabry disease. Normal native T1 values do not exclude Fabry disease, as they are rarely observed in untreated patients with mild LVH (mostly females), or in advanced disease due to pseudonormalization. eAn endomyocardial biopsy is recommended, but could be done in other affected organs such as the kidneys and skin. It should be evaluated by expert pathologists and always include electron microscopy studies to detect lamellar bodies and intracellular inclusions. Of note, some drugs may produce drug-induced phospholipidosis with an intracellular accumulation of phospholipids in different organs that can mimic zebra bodies on electron microscopy.982,983

Table 23

Anderson–Fabry disease red flags

Extracardiac red flags
No male-to-male transmission in pedigree
Renal involvement (dialysis, renal transplantation) or LVH in relatives
Neuropathic pain
Angiokeratomas
Albuminuria
Cornea verticillata
Hypohidrosis, heat/cold and exercise intolerance
Gastrointestinal symptoms (nausea, vomiting, non-specific abdominal pain, constipation, diarrhoea)
Hearing loss (either progressive or sudden), tinnitus, vertigo
Cardiac red flags
ECGShort PQ interval in young patients
Atrioventricular blocks in adult patients
Bradycardia
Chronotropic incompetence
LVH
EchocardiogramLVH with normal systolic function
Hypertrophy of papillary muscles
Mitral and aortic valve thickening with mild-to-moderate regurgitation
Reduced global longitudinal strain
CMRBasal-inferolateral late gadolinium enhancement
Low native T1 (caution with ‘pseudonormalization’ in areas affected by fibrosis)
High focal/global T2
LaboratoryElevated high-sensitivity troponin
Elevated NT-proBNP
Extracardiac red flags
No male-to-male transmission in pedigree
Renal involvement (dialysis, renal transplantation) or LVH in relatives
Neuropathic pain
Angiokeratomas
Albuminuria
Cornea verticillata
Hypohidrosis, heat/cold and exercise intolerance
Gastrointestinal symptoms (nausea, vomiting, non-specific abdominal pain, constipation, diarrhoea)
Hearing loss (either progressive or sudden), tinnitus, vertigo
Cardiac red flags
ECGShort PQ interval in young patients
Atrioventricular blocks in adult patients
Bradycardia
Chronotropic incompetence
LVH
EchocardiogramLVH with normal systolic function
Hypertrophy of papillary muscles
Mitral and aortic valve thickening with mild-to-moderate regurgitation
Reduced global longitudinal strain
CMRBasal-inferolateral late gadolinium enhancement
Low native T1 (caution with ‘pseudonormalization’ in areas affected by fibrosis)
High focal/global T2
LaboratoryElevated high-sensitivity troponin
Elevated NT-proBNP

CMR, cardiac magnetic resonance; ECG, electrocardiogram; LVH, left ventricular hypertrophy; NT-proBNP, N-terminal pro-brain natriuretic peptide.

Table 23

Anderson–Fabry disease red flags

Extracardiac red flags
No male-to-male transmission in pedigree
Renal involvement (dialysis, renal transplantation) or LVH in relatives
Neuropathic pain
Angiokeratomas
Albuminuria
Cornea verticillata
Hypohidrosis, heat/cold and exercise intolerance
Gastrointestinal symptoms (nausea, vomiting, non-specific abdominal pain, constipation, diarrhoea)
Hearing loss (either progressive or sudden), tinnitus, vertigo
Cardiac red flags
ECGShort PQ interval in young patients
Atrioventricular blocks in adult patients
Bradycardia
Chronotropic incompetence
LVH
EchocardiogramLVH with normal systolic function
Hypertrophy of papillary muscles
Mitral and aortic valve thickening with mild-to-moderate regurgitation
Reduced global longitudinal strain
CMRBasal-inferolateral late gadolinium enhancement
Low native T1 (caution with ‘pseudonormalization’ in areas affected by fibrosis)
High focal/global T2
LaboratoryElevated high-sensitivity troponin
Elevated NT-proBNP
Extracardiac red flags
No male-to-male transmission in pedigree
Renal involvement (dialysis, renal transplantation) or LVH in relatives
Neuropathic pain
Angiokeratomas
Albuminuria
Cornea verticillata
Hypohidrosis, heat/cold and exercise intolerance
Gastrointestinal symptoms (nausea, vomiting, non-specific abdominal pain, constipation, diarrhoea)
Hearing loss (either progressive or sudden), tinnitus, vertigo
Cardiac red flags
ECGShort PQ interval in young patients
Atrioventricular blocks in adult patients
Bradycardia
Chronotropic incompetence
LVH
EchocardiogramLVH with normal systolic function
Hypertrophy of papillary muscles
Mitral and aortic valve thickening with mild-to-moderate regurgitation
Reduced global longitudinal strain
CMRBasal-inferolateral late gadolinium enhancement
Low native T1 (caution with ‘pseudonormalization’ in areas affected by fibrosis)
High focal/global T2
LaboratoryElevated high-sensitivity troponin
Elevated NT-proBNP

CMR, cardiac magnetic resonance; ECG, electrocardiogram; LVH, left ventricular hypertrophy; NT-proBNP, N-terminal pro-brain natriuretic peptide.

7.6.1.3. Clinical course, outcome, and risk stratification

Cardiovascular involvement usually manifests as LVH, myocardial fibrosis, inflammation, heart failure, and arrhythmias, which limit QoL and are the most common cause of death. Clinical monitoring is essential to assess disease progression and requires a multidisciplinary approach.980

7.6.1.4. Management

Specific treatment strategies, including enzyme replacement or pharmacological chaperone, have limited efficacy in advanced cases with irreversible organ damage, so early initiation appears to be important. Enzyme replacement therapy is indicated in all symptomatic patients with classical disease, including children, at the earliest signs of organ involvement.974 Therapeutic strategies currently in development include second-generation ERTs, substrate-reduction therapies, and gene and mRNA therapies.980

7.6.2. RASopathies

7.6.2.1. Definition

The RASopathies constitute a group of multisystemic syndromes caused by variants in the RAS-mitogen-activated kinase (RAS-MAPK) cascade,984–986 including Noonan syndrome,987–989 Noonan syndrome with multiple lentigines;990,991 Costello syndrome,992,993 and cardiofaciocutaneous syndrome.994–996

7.6.2.2. Diagnosis, clinical work-up, and differential diagnosis

The suspicion of an underlying RASopathy should be raised in infant- and childhood-onset HCM with coexisting CHD262,263,991,997–1000 or extracardiac abnormalities (see Table 22). Gene testing is recommended for diagnosis when phenotypic features are present. Compared with sarcomeric HCM, RASopathy-associated HCM (RAS-HCM) shows earlier age at diagnosis,261,999 increased prevalence and severity of left or biventricular obstruction,258,262,1001 and higher rates of early hospitalizations for heart failure or need for interventional procedures or surgery.258 Pulmonary stenosis is the most commonly associated CHD, with a prevalence ranging between 25% and 70%, and unfavourable outcomes for pulmonary valvuloplasty.256,1002–1004

7.6.2.3. Clinical course, management, and sudden death risk stratification

Data from the North American Pediatric Cardiomyopathy Registry1005 cohort show poorer survival rates among patients with RAS-HCM compared with non-syndromic HCM, particularly in patients who have been diagnosed before 1 year of age. Disease-specific risk factors for SCD are currently an area of debate, and may include the degree of LV hypertrophy, prolonged QTc interval, ECG risk score for HCM,771 and the HCM Risk-Kids score >6%.81,826

7.6.2.4. Management

Non-vasodilating beta-blockers should be titrated to maximum tolerated dose in patients with RAS-HCM, particularly in cases of severe biventricular obstruction.248,1002,1006–1008 Calcium channel blockers may be considered as a second-line option in patients >6 months of age when beta-blocker therapy is ineffective or not tolerated.267,639 Surgical myectomy and orthotopic heart transplantation may be considered in high-volume centres after multidisciplinary assessment by the heart team.265,266,1009–1011 Pulmonary valvuloplasty may be considered in children and infants with severe RV outflow tract obstruction (RVOTO).1012–1015

7.6.3. Friedreich ataxia

7.6.3.1. Definition

Friedreich ataxia is an autosomal recessive disorder caused by a homozygous GAA triplet repeat expansion in the frataxin (FTX) gene,1016–1019 leading to HCM, progressive neuromuscular symptoms, and extracardiac manifestations, including diabetes mellitus.1016,1020,1021

7.6.3.2. Diagnosis, clinical work-up, and differential diagnosis

Although several diagnostic criteria have been proposed to suspect Friedreich ataxia,1022,1023 genetic testing with identification of bi-co-allelic GAA expansion in the first intron of the FTX gene or compound heterozygosis is required for diagnosis.1024,1025

Cardiovascular involvement usually manifests as hypertrophic non-obstructive cardiomyopathy, with hypokinetic end-stage disease progression and impaired perfusion reserve,1026 leading to advanced heart failure and death.248,1005,1027–1029 There appears to be no specific relationship between the extent of neurological involvement and cardiac phenotype.248,1005,1027–1029,1005,1027–1030,1030 Mitochondrial iron storage is the pathologic hallmark of the disease.1031

7.6.3.3. Clinical course, management, and risk stratification

Supraventricular arrhythmias, particularly AF, are commonly detected.1027 Despite the lack of long-term follow-up longitudinal studies, the risk of ventricular arrhythmias and SCD seems low compared with sarcomeric HCM.1027,1032,1033 The Mitochondrial Protection with Idebenone in Cardiac or Neurological Outcome (MICONOS) study group1034 has proposed a staging of cardiac involvement based on LVEF and end-diastolic wall thickness. The extent of TWI at ECG, LVEF, LV end-diastolic posterior wall thickness, fibrosis on CMR, and hs-TnT have been proposed as negative prognostic factors.1035

7.6.3.4. Management

No specific treatment is currently available for Friedreich ataxia. Treatment with idebenone, a coenzyme Q10 analogue, showed the potential to improve LV mass and cardiac outcomes in open-label studies;1036 nevertheless, four RCTs1037–1040 showed no significant benefit on cardiac or neurologic outcomes.

7.6.4. Glycogen storage disorders

7.6.4.1. Definition

Glycogen storage disorders (GSDs) represent a heterogeneous group of metabolic diseases, including infantile-onset Pompe disease (GSD, type IIa), Danon disease (GSD, type IIb), and PRKAG2 disease.272

7.6.4.2. Diagnosis, clinical work-up, and differential diagnosis

Despite wide clinical heterogeneity, a presentation within the first few months of life, hypotonia, failure to thrive, generalized muscle weakness, and severe non-obstructive HCM with concentric pattern followed by hypokinetic end-stage cardiomyopathy, usually within the first year of life, are typical of GSD IIa.259,268,1041,1042 Short PR interval and increased ECG voltages may represent useful diagnostic clues for GSDs.1042,1043 PRKAG2 syndrome should be suspected in the setting of autosomal dominant transmission and association with conduction system disease including ventricular pre-excitation, sick sinus syndrome, AF, AV block, intraventricular conduction delays or sinoatrial blocks.1043–1047 An X-linked pattern of inheritance is typical of Danon disease (GSD IIb). Skeletal myopathy, in association with learning disability, retinal involvement and ventricular pre-excitation, has been detected in males affected by Danon disease, while the cardiac phenotype can be isolated in affected females.1048–1052

7.6.4.3. Clinical course, management, and risk stratification

In the absence of therapeutic intervention, Pompe disease has a poor prognosis, mainly due to end-stage heart failure.268,1041 Recently, data from a large multicentre European registry have shown that Danon disease runs a malignant phenotype, but there are insufficient data to identify candidate risk factors for sudden death.1049 Sudden cardiac death occurs in almost 10% of patients with PRKAG2 syndrome, mainly as a consequence of advanced AV block, supraventricular tachycardia degenerated to VF, or massive hypertrophy.1044,1053,1054

7.6.4.4. Management

Enzyme replacement therapy is recommended in patients with GSD IIa.269,274,275,1055,1056 To date, there are no approved aetiological therapies for PRKAG2 syndrome and Danon disease. Heart failure therapy, antiarrhythmic therapy, and indications for the implantation of devices are included in Section 6.10.

7.7. Amyloidosis

It is beyond the scope of this document to provide specific recommendations for the assessment and management of cardiac amyloidosis. Instead, the Task Force refers the reader to the 2021 position statement of the ESC Working Group on Myocardial and Pericardial Diseases on Diagnosis and Treatment of Cardiac Amyloidosis.375 This section highlights only the key diagnostic and management issues.

7.7.1. Definition

Cardiac amyloidosis is characterized by the extracellular deposition of misfolded proteins in the ventricular myocardium with the pathognomonic histological property of green birefringence when viewed under cross-polarized light after staining with Congo Red.375

Although once considered a rare disease, data obtained in the last decade suggest that cardiac amyloidosis is underappreciated as a cause of common cardiac diseases or syndromes such as HFpEF, aortic stenosis, or unexplained LVH, particularly in the elderly.1057–1059 Although nine different types of cardiac amyloidosis have been described, most cases correspond to monoclonal immunoglobulin light chain amyloidosis (AL) or transthyretin amyloidosis (ATTR), either in its hereditary (ATTRv) or acquired (ATTRwt) form.375 The ATTRwt form, which is associated with ageing, is currently considered the most frequent form of cardiac amyloidosis worldwide.

7.7.2. Diagnosis, clinical work-up, and differential diagnosis

Cardiac amyloidosis should be suspected in patients with increased LV wall thickness in the presence of cardiac or extracardiac red flags and/or in specific clinical situations, as detailed in Figure 21, particularly in patients >65 years of age.375

Screening for cardiac amyloidosis.
Figure 21

Screening for cardiac amyloidosis.

ATTR, transthyretin amyloidosis; AV, atrioventricular; ECG, electrocardiogram; ECV, extracellular volume; LGE, late gadolinium enhancement; MGUS, monoclonal gammopathy of undetermined significance.

Cardiac amyloidosis can be diagnosed using both invasive and non-invasive diagnostic criteria.375 Invasive diagnostic criteria apply to all forms of cardiac amyloidosis, whereas non-invasive criteria are accepted only for ATTR. Invasive criteria include demonstration of amyloid fibrils within cardiac tissue or, alternatively, demonstration of amyloid deposits in an extracardiac biopsy accompanied either by characteristic features of cardiac amyloidosis on echocardiography or CMR.375 Non-invasive criteria include typical echocardiographic/CMR findings combined with planar and single-photon emission computed tomography (SPECT) grade 2 or 3 myocardial radiotracer uptake in 99mtechnetium-pyrophosphate (99mTc-PYP) or 3,3-diphosphono-1,2-propanodicarboxylic acid (DPD) or hydroxymethylene diphosphonate (HMDP) scintigraphy and exclusion of a clonal dyscrasia by all the following tests: serum free light chain assay, serum and urine protein electrophoresis with immunofixation.168 Tomographic scintigraphy should be considered in order to reduce the number of misclassifications.1060 False negative scans may rarely occur in certain ATTRv genotypes; false positives may be due to AL, recent myocardial infarction, or long-term chloroquine use.370 Therefore, planar and SPECT scintigraphy coupled with assessment for monoclonal proteins followed by CMR and/or cardiac/extracardiac biopsy if necessary allows appropriate diagnosis in patients with suggestive signs/symptoms, as described in Figure 22.375 However, the DPD/PYP/HMDP scan cannot distinguish between wild-type and mutated ATTR, and therefore TTR genetic testing is required. Of note, TTR genetic testing is recommended in all transthyretin amyloid cardiomyopathy (ATTR-CM) patients regardless of age, as 5% of ATTR-CM patients ≥70 years (and 10% among females) have ATTRv.375,1061

Diagnosis of cardiac amyloidosis.
Figure 22

Diagnosis of cardiac amyloidosis.

AL, amyloid light chain; ATTR, transthyretin amyloidosis; ATTRv, variant transthyretin amyloidosis; ATTRwt, wild-type transthyretin amyloidosis; CMR, cardiac magnetic resonance; DPD, 3,3-diphosphono-1,2-propanodicarboxylic acid; ECG, electrocardiogram; ECHO, echocardiogram; HMDP, hydroxymethylene diphosphonate; PYP, pyrophosphate; TTR, transthyretin.

7.7.3. Clinical course and risk stratification

Cardiac amyloidosis is a progressive disease with poor outcomes if left untreated. Amyloid light chain cardiac amyloidosis is associated with more rapid progression of heart failure and worse prognosis than ATTR.1058,1062,1063 Fortunately, the prognosis of AL amyloidosis has significantly improved with the introduction of very effective therapies capable of dramatically reducing the production of the cardiotoxic light chains.1064 Prognosis in ATTR depends on the variant, degree of cardiac involvement, and neurologic phenotype.1065–1068 Several multiparametric biomarker-based staging systems have been developed for AL1069,1070 and ATTR cardiac amyloidosis1066–1068 (see Supplementary data online, Table S5).

7.7.4. Management

The treatment of cardiac amyloidosis includes treating and preventing complications and stopping or delaying amyloid deposition by specific treatment.375,1071 There is no evidence to support the use of standard heart failure therapy, which often is not well tolerated, apart from diuretics (see Section 6.10.2).1072,1073

The natural history of cardiac amyloidosis associates electrical conduction disease of the heart with symptomatic bradycardia and advanced AV block.375,1074,1075 The clinical threshold for pacemaker indication should be low, as the disease progresses and implantation of the device allows rate response to exercise and medication adjustment.375,1074 The role of ICD in cardiac amyloidosis for SCD prevention is not clearly known, but available data do not support their use in primary prevention.1076,1077

7.7.4.1. Specific therapies

Therapy for AL cardiac amyloidosis is based on treatment of the underlying haematological problem with chemotherapy or autologous stem-cell transplant.1064

Transthyretin stabilization and reduction of its production are the basis of TTR cardiac amyloidosis treatment. Tafamidis reduced all-cause mortality and cardiovascular hospitalizations in ATTR, with the largest effect achieved in patients at NYHA functional class I and II.1078 Additional studies are being conducted with other stabilizing agents and other molecules that reduce TTR production.1078a

8. Other recommendations

8.1. Sports

8.1.1. Cardiovascular benefits of exercise

Regular physical activity and systematic exercise confer several cardiovascular, psychological, and QoL benefits. Through curbing risk factors for atherosclerosis, such as obesity and insulin resistance,1079 hypertension,1080 and hyperlipidaemia,1081 regular physical activity is associated with an up to 50% risk reduction in an adverse event from CAD in middle-aged and older individuals.1082,1083 Individuals who exercise regularly live 5–7 years longer than their sedentary counterparts,1084 and have a lower risk of cerebrovascular accidents1085 and certain malignancies.1085–1087 These benefits that can be derived later in life also apply to individuals with established cardiovascular disease. For a definition of exercise intensity levels, please refer to Supplementary data online, Table S6.

8.1.2. Exercise-related sudden cardiac death and historical exercise recommendations for patients with cardiomyopathy

Rigorous exercise may trigger myocardial infarction and fatal arrhythmias among individuals with an underlying cardiovascular disease.1088–1091 Superimposed on the pathological substrate of the disease entity itself, exercise may induce sudden cardiac arrest through mechanical shearing forces within the coronary arteries, effects of high concentrations of circulating catecholamines, increased cardiac loading conditions, raised core temperature, electrolyte shifts, and acid-base disturbance.

Cardiomyopathies are the leading cause of exercise-related SCD in young people in the Western world.40,1092–1095 The established link between exercise and SCD from cardiomyopathy, and the finding that, in some cardiomyopathy phenotypes, exercise may accelerate progression of the underlying cardiomyopathic disease process, has historically resulted in restrictive exercise recommendations in all affected patients regardless of pathology, disease severity, symptomatic status, general risk profile, or prior therapeutic interventions, including an ICD.1096–1098 As a result, individuals with cardiomyopathy often confine themselves to a relatively sedentary lifestyle through fear of potential SCD and accrue risk factors for atherosclerotic CAD, which confer a worse prognosis.1099–1102,1096,1097

8.1.3. Exercise recommendations in hypertrophic cardiomyopathy

Recent pre-clinical1103 and clinical data suggest that moderate exercise may be beneficial and safe in patients with HCM.1098–1102 Information on a safe dose of vigorous exercise is still limited, but the heterogeneous morphology and pathophysiology of HCM means that some individuals are capable of participating in vigorous exercise, including high-intensity competitive sports.760 Most athletes capable of exercising intensively have mild LV hypertrophy, normal-sized or enlarged LV, normal diastolic function, and no evidence of LVOTO.1104,1105 Currently available data indicate that participation in vigorous exercise and competitive sport may be considered in a select group of predominantly adult patients who have mild morphology and a low-risk profile.1106–1108 However, studies examining the effect of vigorous exercise or moderate-to-high-intensity competitive sport on the natural history of HCM were not designed or powered adequately to address the question and there are potential issues of selection bias. Nevertheless, based on emerging evidence, the Task Force agreed to adopt a comparatively liberal approach, advocating that, after appropriate selection, some individuals with a low-risk profile may participate in high-intensity exercise and competitive sport after comprehensive expert evaluation and shared discussion, which highlights the unpredictable nature of exercise-related SCD in HCM. Sporting disciplines in which syncope may result in fatal accidental injury or danger to others are not recommended.

Genotype-positive/phenotype-negative patients may engage in all competitive sport; however, annual assessment is recommended to check for developing phenotypic features of disease.1109

8.1.4. Exercise recommendations in arrhythmogenic right ventricular cardiomyopathy

Arrhythmogenic right ventricular cardiomyopathy is a recognized cause of exercise-related SCD in young asymptomatic individuals,40,890 postulated to result from ventricular stretch leading to myocyte detachment with subsequent inflammation and fibro-fatty replacement of the ventricular myocardium. Fatal arrhythmias may occur during the inflammatory process or because of myocardial scar. In addition, there are data to suggest that high-intensity exercise is associated with acceleration of disease phenotype in individuals with ARVC, including those who are genotype positive/phenotype negative, and particularly those with PKP2 variants.181,1110–1114 Furthermore, exercise restriction has been shown to improve clinical outcomes in patients with ARVC.40,1111,1115–1117 Based on these data, the Task Force recommends against intensive exercise or competitive sports in individuals with ARVC as part of a shared decision-making process. The evidence on the impact of exercise in genotype-positive/phenotype-negative individuals is more limited. In these cases, the Task Force recommends a cautious approach in the context of shared decision-making when discussing competitive sports participation. Mild-to-moderate physical activity for up to 150 min per week is considered safe and is recommended in able phenotype-negative individuals.1118

8.1.5. Exercise recommendations in dilated cardiomyopathy and non-dilated left ventricular cardiomyopathy

There is evidence that moderate exercise in optimally treated patients with DCM improves functional capacity, ventricular function, and QoL;1119 however, intensive exercise and competitive sports may also trigger fatal arrhythmias in DCM and NDLVC.1093,1120–1122

In general, symptomatic individuals with DCM and NDLVC should abstain from most competitive and leisure sports, or recreational exercise associated with moderate or high exercise intensity. A select group of asymptomatic individuals with DCM and NDLVC who have mildly impaired LV function without exercise-induced arrhythmias or significant myocardial fibrosis may participate in most competitive sports.

Although the natural history of most pathogenic variants capable of causing DCM and NDLVC is unknown, it would be reasonable to permit intensive exercise and competitive sports in most individuals with pathogenic variants in the absence of overt features of DCM or NDLVC. Special consideration, however, should be given to individuals with pathogenic variants in genes that are associated with an increased risk of life-threatening arrhythmias, such as lamin A/C181,1123 or TMEM43 variants, for which there is emerging evidence that exercise may have an adverse effect on cardiac function and risk of potentially fatal arrhythmias. The impact of vigorous exercise in patients with pathogenic variants in other high-risk genes, such as filamin C variants1112 exhibiting DCM or NDLVC phenotypes, is not fully understood; however, extrapolating our understanding of the effect of exercise on some ARVC and DCM phenotypes necessitates a cautious approach.

Recommendation Table 31

Exercise recommendations for patients with cardiomyopathy

graphic
graphic
graphic
graphic
Recommendation Table 31

Exercise recommendations for patients with cardiomyopathy

graphic
graphic
graphic
graphic

8.2. Reproductive issues

Pregnancy and the post-partum period constitute periods of increased risk of cardiovascular complications in women with cardiomyopathy.1127–1130 Cardiomyopathy can also be first diagnosed in pregnancy or arise during pregnancy as PPCM.1131

The risk associated with pregnancy in a patient with a cardiomyopathy is estimated using the modified World Health Organization (mWHO) classification.1130 Pregnancy is contraindicated in women with WHO class IV, including patients with EF <30% or NYHA class III–IV or previous PPCM with persisting impairment of the LV function.

8.2.1. Contraception, in vitro fertilization, and hormonal treatment

Counselling on safe and effective contraception is indicated in all women of fertile age. Ethinyloestradiol-containing contraceptives have the greatest risk of thrombosis1132 and are not advised in women with a high risk of thrombo-embolic disease. Progestin-only contraceptives are an alternative, as they have little or no effect on coagulation factors, blood pressure, and lipid levels. Levonorgestrel-based long-acting reversible contraception implants or intrauterine devices are the safest and most effective contraceptives and have few side effects affecting cardiomyopathies.

Medically assisted procreation adds risks beyond those of pregnancy alone; superovulation is pro-thrombotic and can be complicated by ovarian hyperstimulation syndrome, with marked fluid shifts and an even greater risk of thrombosis. Hormonal stimulation should be carefully considered in women who have WHO class III disease (VT or HCM) or who are anticoagulated.

8.2.2. Pregnancy management

8.2.2.1. Pre-pregnancy

Patients with a known cardiomyopathy and at risk of developing cardiomyopathy should receive pre-pregnancy counselling by a multidisciplinary team: the pregnancy heart team. The individual risk of the woman by pregnancy should be discussed using the WHO classification, in addition to discussing the likelihood of transmission of the disease to the offspring and how to reduce the transgenerational risk of transmitting the disorder.

For individual risk estimation, at a minimum, an ECG, echocardiography, and exercise test should be performed. Several aspects must be discussed with the woman, including long-term prognosis, drug therapy, estimated maternal risk and outcome, and plans for pregnancy care and delivery.

8.2.2.2. Pregnancy

In women with mWHO class II–III, III, and IV (including women with HCM, VTs, and EF <35%), management during pregnancy and around delivery should be conducted in an expert centre by a multidisciplinary team: the pregnancy heart team, including cardiologists with expertise in cardiomyopathies and arrhythmias; obstetricians; and anaesthetists. Depending on the individual case, other specialists may be included (geneticist, cardio-thoracic surgeon, paediatric cardiologist, foetal medicine specialist, neonatologist, etc.). A delivery plan should be created that includes the details of induction; the management of labour and delivery; and post-partum surveillance.

8.2.2.3. Timing and mode of delivery

The timing and mode of delivery should be personalized according to the type of cardiomyopathy, ventricular function, NYHA class, arrhythmic risk, and thrombo-embolic risk. Vaginal delivery is associated with less blood loss and lower risk of infection, venous thrombosis, and embolism than caesarean section and should be advised for most women. Caesarean section should be considered for obstetric indications, patients with severe outflow tract obstruction, or in cases of severe acute/intractable heart failure, or in cases at high risk of threatening arrhythmia and for patients presenting in labour on oral anticoagulants.1130 During delivery, patients with cardiomyopathy should be circulatory and heart rhythm monitored on an individualized basis.

8.2.2.4. Post-partum

The post-partum period is associated with significant haemodynamic changes and fluid shifts, particularly in the first 24–48 h after delivery, which may precipitate heart failure. Haemodynamic monitoring should therefore be continued for at least 24–48 h in patients at risk. Most drugs enter the milk and could thus contraindicate breastfeeding (see Section 8.2.2.5).

8.2.2.5. Pharmacological treatment: general aspects

Pharmacological treatment in pregnant women should be the same as in non-pregnant patients, with an avoidance of drugs contraindicated in pregnancy, such as ACE-Is, ARBs, and renin inhibitors.1130 The first trimester is associated with the greatest teratogenic risk. Pharmacologic therapy is advised to begin as late as possible in pregnancy and at the lowest effective dose. Drug exposure later in pregnancy may confer adverse effects on foetal growth and development. It is recommended to check drug and safety data before initiation of a new drug in pregnancy; see Table 7 in the 2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy.1130 From this list, antiarrhythmics can be summarized as follows:

  • Well tolerated: sotalol, oral verapamil.

  • While the benefits and risks should be evaluated in each case, the following drugs can often be continued if there is a clear indication for use during pregnancy: bisoprolol, carvedilol, digoxin, diltiazem (possible teratogenic effects), disopyramide (uterine contractions), flecainide, lidocaine, metoprolol, nadolol, propranolol, verapamil, quinidine.

  • Insufficient data: ivabradine, mexiletine, propafenone, vernakalant.

  • Contraindicated: amiodarone, atenolol, dronedarone, ACE-Is, ARBs, renin inhibitors, and spironolactone.1130

Ongoing beta-blocker treatment in cardiomyopathies should be continued during pregnancy, with close monitoring of foetal growth. After delivery, it is advised to heart rhythm monitor the infant for 48 h. The use of beta-blockers and anticoagulation during pregnancy is described in the 2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy.1130

Vitamin K antagonist use in the first trimester results in embryopathy (limb defects, nasal hypoplasia) in 0.6–10% of cases.1133,1134 In contrast, unfractionated heparin (UFH) and low-molecular-weight heparin (LMWH) do not cross the placenta; therefore, substitution of VKA with UFH or LMWH in weeks 6–12 almost eliminates the risk of embryopathy. This risk is also dose dependent (0.45–0.9% with low-dose warfarin). Vaginal delivery while the mother is on VKAs is contraindicated because of the risk of foetal intracranial bleeding. Haemorrhagic complications in the mother occur with all regimens, but the incidence is lower with VKA than with LMWH/UFH throughout pregnancy.1130

VKA should be continued until pregnancy is achieved. Continuation of VKAs throughout pregnancy should be considered when the dose is low (see Table 7 in the 2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy1130). The target international normalized ratio (INR) should be chosen according to current guidelines, with INR monitoring weekly or every 2 weeks. Self-monitoring of INR in suitable patients is recommended. Alternatively, depending on the indication, a switch to LMWH from weeks 6–12 under strict monitoring may be considered in patients with a low dose requirement. When a higher dose of VKAs is required, discontinuation of VKAs between weeks 6 and 12 and replacement with adjusted-dose i.v. UFH or LMWH twice daily with dose adjustment according to peak anti-Xa (for LMWH) levels should be considered.

In case of delivery in anticoagulated women (not including mechanical valves) with a planned caesarean section, therapeutic LMWH dosing can be simply omitted for 24 h prior to surgery. If delivery has to be performed earlier, anti-Xa activity can guide the timing of the procedure.

Antiarrhythmic therapy in pregnancy other than medication. Implantation of an ICD and catheter ablation should ideally be considered prior to pregnancy in patients with a high risk of ventricular arrhythmias to avoid implantations and interventions during pregnancy.1135 If an ICD is indicated in pregnancy, ICD implantation should be performed beyond 8 weeks of gestation with radiation protection1136 and the indication should be weighed against the limited experience available. In pregnant patients with existing ICD, routine ICD interrogation and advice are recommended prior to delivery.

8.2.2.6. Specific cardiomyopathies

Most women with HCM tolerate pregnancy well.1137 Complications during pregnancy most often occur in women who have symptoms, arrhythmias, or impaired LV function before pregnancy. Left ventricular outflow tract gradients may increase slightly during pregnancy and high gradients before pregnancy are associated with more complications.1137 Women should be assessed according to WHO risk class, indicating at trimester for low-risk patients (class II) and monthly or bi-monthly for higher-risk patients (class III). Therapeutic anticoagulation with LMWH or VKAs according to the stage of pregnancy is recommended for patients with AF. Cardioversion in pregnancy should be considered for poorly tolerated persistent AF. Hypovolaemia is poorly tolerated. Caesarean section should be considered in patients with severe LVOTO, pre-term labour while on oral anticoagulants, or severe heart failure.1130 Epidural and spinal anaesthesia must be applied cautiously, especially with severe LVOTO, due to potential hypovolaemia, and single-shot spinal anaesthesia should be avoided.

Pregnancy in ARVC seems to be relatively tolerable, as shown in several studies, with no excess mortality and no clear negative long-term outcome.1138–1141 Previous VTs represent a WHO risk class III, indicating bi-monthly or monthly follow-up at an expert centre.

Women with DCM are at risk of further deterioration of LV function in pregnancy. Data suggest that pregnancy might not be associated with long-term adverse disease progression or event-free survival in LMNA genotype-positive women.1142 Predictors of maternal mortality are NYHA class III/IV and EF <40%. Highly adverse risk factors include EF <20%, severe mitral regurgitation, RV failure, AF, and/or hypotension.1143

8.2.2.7. Peripartum cardiomyopathy

Genetic studies in patients with PPCM have revealed genetic similarity between PPCM and DCM. Specifically, an overrepresentation of truncating variants has been demonstrated in TTN, FLNC, BAG3, and DSP, with TTN truncating variants most commonly involved (found in ∼10% of patients).44,45 It has been suggested that approaches to genetic testing in PPCM should mirror those taken in DCM.45 Medications used to treat heart failure during pregnancy require special considerations as discussed above. In the presence of persistant cardiac dysfunction, medication should be continued. Use of bromocriptine as disease-specific therapy in patients with PPCM as an addition to standard heart failure therapy has shown promising results in two clinical trials.1144,1145 In severe cases of PPCM, temporary MCS has been used successfully and should be considered in patients with haemodynamic instability despite inotropic support.1146 In patients with PPCM, thresholds for early ICD implantations should be higher than in other conditions because of a high rate of spontaneous recovery after delivery.1147

Recommendation Table 32

Recommendations for reproductive issues in patients with cardiomyopathy

graphic
graphic
Recommendation Table 32

Recommendations for reproductive issues in patients with cardiomyopathy

graphic
graphic

8.3. Recommendations for non-cardiac surgery

Cardiomyopathies, in general, are associated with an increased incidence of peri-operative heart failure and arrhythmias, although the significant variability in the phenotypic expression of cardiomyopathies must be considered. Special attention should be given to the clinical status, LVEF, volume overload, and increased levels of natriuretic peptides. In the period after non-cardiac surgery (NCS), fluids given during the operation may be mobilized, causing hypervolaemia and pulmonary congestion. Careful attention to fluid balance is therefore essential.1148,1149 Obstructive HCM deserves specific consideration due to its peculiar pathophysiology, with adequate intra-operative vigilance, avoiding factors and medication that may increase LVOTO and prompt pharmacological treatment and intravascular fluid therapy if needed (see Supplementary data online, Table S7).1150,1151

Natriuretic peptide concentrations are quantitative plasma biomarkers for the presence and severity of haemodynamic cardiac stress and heart failure, and elevated NT-proBNP concentrations may facilitate detection of heart failure, optimal intra-operative monitoring, and initiation or optimization of heart failure therapy after surgery.1152 Moreover, in cardiomyopathy patients elevated NT-proBNP values are strong predictors of overall prognosis.1153–1156

Patients with a first-degree relative with a genetic cardiomyopathy should be evaluated with an ECG and an echocardiographic examination to rule out the presence of the disease, irrespective of age (see Section 6.11). There are no specific data on risks of NCS in phenotype-negative family members; however, they are at risk of developing the disease, which may be subclinical at the time of the NCS.1157 Data in children with HCM undergoing general anaesthesia for cardiac and non-cardiac procedures show that, in a specialist setting with multidisciplinary involvement, peri-operative morbidity and mortality are extremely low.1158

Recommendation Table 33

Recommendations for non-cardiac surgery in patients with cardiomyopathy

graphic
graphic
Recommendation Table 33

Recommendations for non-cardiac surgery in patients with cardiomyopathy

graphic
graphic

9. Requirements for specialized cardiomyopathy units

As genomic tests and information are incorporated into strategies for the routine diagnosis and management of cardiomyopathies and the estimation of disease risk, cardiologists need to familiarize themselves with the general principles underlying the interpretation of test results and must be able to convey the implications to patients. They also need to be able to make informed judgments about which tests are appropriate for different patients and clinical situations. The risk of SCD and the possibility that family members could inherit the condition makes multidisciplinary expertise, including genetic counselling, psychological care, and patient support associations, a critical aspect of care.1166 As a result, there is a growing need for clinicians to develop their understanding of the basic principles of clinical genetics and the diverse clinical manifestations of individual genetic disorders.54,964,1166,1167

Cardiomyopathies have a highly heterogeneous clinical presentation and an evolution that sometimes is difficult to predict. Disease phenotype can be the result of various acquired factors or genetic backgrounds. Mixed phenotypes or two conditions within the same patient or among a family can coexist. Genetic diagnosis raises common logistical and ethical problems in its execution, as well as in the interpretation and communication of the results.1166 Diagnostic process, the management of symptoms, and risk stratification often require comprehensive evaluation of the patient and their family, with the participation of multidisciplinary teams. On the other hand, interventional procedures (septal ablation, myectomies, etc.) require an expertise that only centres that treat many patients can achieve. Specialization in this area also requires permanent updating to accurately characterize the disease prognosis, ensure the choice of the best therapeutic option in each case, and guarantee the implementation of that choice by a team with experience in the field.

These characteristics imply that the adequate management of these diseases requires specific tools, extensive experience, and a multidisciplinary basic-clinical approach that are difficult to achieve.

The cardiomyopathy unit is usually integrated into a general cardiogenetic (or inherited cardiac conditions) unit, where other professionals involved in hereditary cardiac and vascular conditions, such as channelopathies, genetic aortopathies, familial dyslipidaemias, and a number of genetic metabolic and syndromic diseases with cardiac involvement, are co-ordinated. They represent an organizational model aimed at providing comprehensive cardiovascular and genetic assessment and personalized management in patients with inherited cardiovascular diseases. Specialized multidisciplinary clinics have long been advocated as the ideal model for the management of patients and families with inherited cardiac conditions.4,53,559,1166 Such a model of care supports the holistic care of patients and their at-risk family members, taking a patient-centred approach and valuing clinical, genetic, and psychosocial outcomes. The benefit of a specialized clinic for management of HCM has been previously reported, with patients showing better adjustment and less worry than those who did not attend.53,224 Besides expertise in the field of inherited cardiac conditions, the presence of a multidisciplinary team, access to good technical resources, participation in dedicated research projects, availability of genetic counselling, and family screening are all pre-requisites for organizing a cardiogenetic clinic. The ability to provide education and training for medical professionals and collaboration with patients’ associations is of utmost importance.

Supplementary data online, Table S8 synthesizes the requirements and skills and recommendations for professional education/training needed for a cardiogenetic clinic as proposed by international expert associations.

10. Living with cardiomyopathy: advice for patients

Most people with cardiomyopathy lead normal and productive lives, but a small number experience significant symptoms and are at risk of disease-related complications. Regardless of the severity of their disease, it is important that individuals receive support and accurate advice from cardiomyopathy specialists and other healthcare professionals, and that they are encouraged to understand and manage the disease themselves (see the Supplemental Data online, Table S9, for a description of the patient education process). Table 24 summarizes some of the key issues that should be discussed with patients, relatives, and carers. When appropriate (e.g. when considering pregnancy, see Section 8.2), patients should be referred to other specialist services.

Table 24

General guidance for daily activity for patients with cardiomyopathies

TopicGeneral guidance
Exercise
  • See earlier section on exercise recommendations.

Diet, alcohol use, and weight
  • Patients should be encouraged to maintain a recommended body mass index.

  • Avoid dehydration, excess alcohol intake, and drugs consumption.

Smoking
  • There are no data that show an interaction between tobacco smoking and cardiomyopathy, but patients should be provided with general advice on the health risks associated with smoking, including pro-arrhythmic and pro-inflammatory effects and, when available, information on smoking cessation.1168–1171

Reproductive issues
  • Patients should be given the opportunity to discuss their concerns about reproductive issues. Anxiety and depression following a diagnosis are frequent, and some patients may express guilt or fear about their genetic diagnosis and the risk of transmission to offspring.

Sexual activity
  • Patients should be counselled on the potential effect of their medication on sexual performance.

  • Most people with cardiomyopathy will be able to undertake normal sexual activity. Individualized advice should be provided regarding its safety and the possible impact of sexual activity on the risk of disease progression, ventricular arrhythmias, and/or ICD shocks.

Medication
  • Patients should be provided with information about their medication, including potential side and teratogenic effects and interactions with prescribed medications, over-the-counter remedies, and other complementary therapies.

Vaccination
  • In the absence of contraindications, patients should be advised to have regular recommended vaccinations (e.g. yearly influenza and SARS-CoV-2 vaccination).

Driving
  • Most patients should be eligible for an ordinary driving licence and can continue driving unless they experience distracting or disabling symptoms.

  • Advice on driving licences for heavy goods or passenger-carrying vehicles should be in line with local legislation.

  • For further information on driving with ICDs, see local rules.

Occupation
  • Most people with cardiomyopathy will be able to accomplish their normal jobs. The implications of heavily manual jobs that involve strenuous activity should be discussed with the appropriate specialist.

  • For some occupations, such as pilots, military personnel, and emergency services personnel, there are strict guidelines or rules on eligibility.

  • The social and financial implications of a diagnosis of cardiomyopathy should be included in the counselling of relatives before clinical or genetic screening.

Holidays and travel insurance
  • Most asymptomatic or mildly symptomatic patients can fly safely. For further information on flying with ICD, see ‘Fitness to fly for passengers with cardiovascular disease’.1172

  • Insurance companies may charge more for travel insurance. In some countries, patient support organizations can provide advice about obtaining reasonable insurance.

Life insurance
  • The diagnosis of cardiomyopathy will result in difficulty obtaining life insurance or mortgages. Advice on the rules that apply in different countries should be provided to patients at diagnosis.

Pregnancy and childbirthSee Section 8.2
Education/schooling
  • Teachers and other carers should be provided with advice and written information relevant to the care of children with cardiomyopathy.

  • In the absence of symptoms and risk factors, children should be allowed to perform low- to moderate-level aerobic physical activity, in accordance with advice from their paediatric cardiologist. Advice on high-intensity exercise in children should be guided by cardiomyopathy phenotype and the presence of symptoms and risk factors within a specialist paediatric cardiomyopathy setting.

  • Provision should be made for children with learning difficulties and other special needs.

  • Parents, teachers, and staff at sports facilities should be trained in CPR and in the use of AEDs.

TopicGeneral guidance
Exercise
  • See earlier section on exercise recommendations.

Diet, alcohol use, and weight
  • Patients should be encouraged to maintain a recommended body mass index.

  • Avoid dehydration, excess alcohol intake, and drugs consumption.

Smoking
  • There are no data that show an interaction between tobacco smoking and cardiomyopathy, but patients should be provided with general advice on the health risks associated with smoking, including pro-arrhythmic and pro-inflammatory effects and, when available, information on smoking cessation.1168–1171

Reproductive issues
  • Patients should be given the opportunity to discuss their concerns about reproductive issues. Anxiety and depression following a diagnosis are frequent, and some patients may express guilt or fear about their genetic diagnosis and the risk of transmission to offspring.

Sexual activity
  • Patients should be counselled on the potential effect of their medication on sexual performance.

  • Most people with cardiomyopathy will be able to undertake normal sexual activity. Individualized advice should be provided regarding its safety and the possible impact of sexual activity on the risk of disease progression, ventricular arrhythmias, and/or ICD shocks.

Medication
  • Patients should be provided with information about their medication, including potential side and teratogenic effects and interactions with prescribed medications, over-the-counter remedies, and other complementary therapies.

Vaccination
  • In the absence of contraindications, patients should be advised to have regular recommended vaccinations (e.g. yearly influenza and SARS-CoV-2 vaccination).

Driving
  • Most patients should be eligible for an ordinary driving licence and can continue driving unless they experience distracting or disabling symptoms.

  • Advice on driving licences for heavy goods or passenger-carrying vehicles should be in line with local legislation.

  • For further information on driving with ICDs, see local rules.

Occupation
  • Most people with cardiomyopathy will be able to accomplish their normal jobs. The implications of heavily manual jobs that involve strenuous activity should be discussed with the appropriate specialist.

  • For some occupations, such as pilots, military personnel, and emergency services personnel, there are strict guidelines or rules on eligibility.

  • The social and financial implications of a diagnosis of cardiomyopathy should be included in the counselling of relatives before clinical or genetic screening.

Holidays and travel insurance
  • Most asymptomatic or mildly symptomatic patients can fly safely. For further information on flying with ICD, see ‘Fitness to fly for passengers with cardiovascular disease’.1172

  • Insurance companies may charge more for travel insurance. In some countries, patient support organizations can provide advice about obtaining reasonable insurance.

Life insurance
  • The diagnosis of cardiomyopathy will result in difficulty obtaining life insurance or mortgages. Advice on the rules that apply in different countries should be provided to patients at diagnosis.

Pregnancy and childbirthSee Section 8.2
Education/schooling
  • Teachers and other carers should be provided with advice and written information relevant to the care of children with cardiomyopathy.

  • In the absence of symptoms and risk factors, children should be allowed to perform low- to moderate-level aerobic physical activity, in accordance with advice from their paediatric cardiologist. Advice on high-intensity exercise in children should be guided by cardiomyopathy phenotype and the presence of symptoms and risk factors within a specialist paediatric cardiomyopathy setting.

  • Provision should be made for children with learning difficulties and other special needs.

  • Parents, teachers, and staff at sports facilities should be trained in CPR and in the use of AEDs.

AED, automated external defibrillator; CPR, cardio-pulmonary resuscitation; ICD, implantable cardioverter defibrillator; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

Table 24

General guidance for daily activity for patients with cardiomyopathies

TopicGeneral guidance
Exercise
  • See earlier section on exercise recommendations.

Diet, alcohol use, and weight
  • Patients should be encouraged to maintain a recommended body mass index.

  • Avoid dehydration, excess alcohol intake, and drugs consumption.

Smoking
  • There are no data that show an interaction between tobacco smoking and cardiomyopathy, but patients should be provided with general advice on the health risks associated with smoking, including pro-arrhythmic and pro-inflammatory effects and, when available, information on smoking cessation.1168–1171

Reproductive issues
  • Patients should be given the opportunity to discuss their concerns about reproductive issues. Anxiety and depression following a diagnosis are frequent, and some patients may express guilt or fear about their genetic diagnosis and the risk of transmission to offspring.

Sexual activity
  • Patients should be counselled on the potential effect of their medication on sexual performance.

  • Most people with cardiomyopathy will be able to undertake normal sexual activity. Individualized advice should be provided regarding its safety and the possible impact of sexual activity on the risk of disease progression, ventricular arrhythmias, and/or ICD shocks.

Medication
  • Patients should be provided with information about their medication, including potential side and teratogenic effects and interactions with prescribed medications, over-the-counter remedies, and other complementary therapies.

Vaccination
  • In the absence of contraindications, patients should be advised to have regular recommended vaccinations (e.g. yearly influenza and SARS-CoV-2 vaccination).

Driving
  • Most patients should be eligible for an ordinary driving licence and can continue driving unless they experience distracting or disabling symptoms.

  • Advice on driving licences for heavy goods or passenger-carrying vehicles should be in line with local legislation.

  • For further information on driving with ICDs, see local rules.

Occupation
  • Most people with cardiomyopathy will be able to accomplish their normal jobs. The implications of heavily manual jobs that involve strenuous activity should be discussed with the appropriate specialist.

  • For some occupations, such as pilots, military personnel, and emergency services personnel, there are strict guidelines or rules on eligibility.

  • The social and financial implications of a diagnosis of cardiomyopathy should be included in the counselling of relatives before clinical or genetic screening.

Holidays and travel insurance
  • Most asymptomatic or mildly symptomatic patients can fly safely. For further information on flying with ICD, see ‘Fitness to fly for passengers with cardiovascular disease’.1172

  • Insurance companies may charge more for travel insurance. In some countries, patient support organizations can provide advice about obtaining reasonable insurance.

Life insurance
  • The diagnosis of cardiomyopathy will result in difficulty obtaining life insurance or mortgages. Advice on the rules that apply in different countries should be provided to patients at diagnosis.

Pregnancy and childbirthSee Section 8.2
Education/schooling
  • Teachers and other carers should be provided with advice and written information relevant to the care of children with cardiomyopathy.

  • In the absence of symptoms and risk factors, children should be allowed to perform low- to moderate-level aerobic physical activity, in accordance with advice from their paediatric cardiologist. Advice on high-intensity exercise in children should be guided by cardiomyopathy phenotype and the presence of symptoms and risk factors within a specialist paediatric cardiomyopathy setting.

  • Provision should be made for children with learning difficulties and other special needs.

  • Parents, teachers, and staff at sports facilities should be trained in CPR and in the use of AEDs.

TopicGeneral guidance
Exercise
  • See earlier section on exercise recommendations.

Diet, alcohol use, and weight
  • Patients should be encouraged to maintain a recommended body mass index.

  • Avoid dehydration, excess alcohol intake, and drugs consumption.

Smoking
  • There are no data that show an interaction between tobacco smoking and cardiomyopathy, but patients should be provided with general advice on the health risks associated with smoking, including pro-arrhythmic and pro-inflammatory effects and, when available, information on smoking cessation.1168–1171

Reproductive issues
  • Patients should be given the opportunity to discuss their concerns about reproductive issues. Anxiety and depression following a diagnosis are frequent, and some patients may express guilt or fear about their genetic diagnosis and the risk of transmission to offspring.

Sexual activity
  • Patients should be counselled on the potential effect of their medication on sexual performance.

  • Most people with cardiomyopathy will be able to undertake normal sexual activity. Individualized advice should be provided regarding its safety and the possible impact of sexual activity on the risk of disease progression, ventricular arrhythmias, and/or ICD shocks.

Medication
  • Patients should be provided with information about their medication, including potential side and teratogenic effects and interactions with prescribed medications, over-the-counter remedies, and other complementary therapies.

Vaccination
  • In the absence of contraindications, patients should be advised to have regular recommended vaccinations (e.g. yearly influenza and SARS-CoV-2 vaccination).

Driving
  • Most patients should be eligible for an ordinary driving licence and can continue driving unless they experience distracting or disabling symptoms.

  • Advice on driving licences for heavy goods or passenger-carrying vehicles should be in line with local legislation.

  • For further information on driving with ICDs, see local rules.

Occupation
  • Most people with cardiomyopathy will be able to accomplish their normal jobs. The implications of heavily manual jobs that involve strenuous activity should be discussed with the appropriate specialist.

  • For some occupations, such as pilots, military personnel, and emergency services personnel, there are strict guidelines or rules on eligibility.

  • The social and financial implications of a diagnosis of cardiomyopathy should be included in the counselling of relatives before clinical or genetic screening.

Holidays and travel insurance
  • Most asymptomatic or mildly symptomatic patients can fly safely. For further information on flying with ICD, see ‘Fitness to fly for passengers with cardiovascular disease’.1172

  • Insurance companies may charge more for travel insurance. In some countries, patient support organizations can provide advice about obtaining reasonable insurance.

Life insurance
  • The diagnosis of cardiomyopathy will result in difficulty obtaining life insurance or mortgages. Advice on the rules that apply in different countries should be provided to patients at diagnosis.

Pregnancy and childbirthSee Section 8.2
Education/schooling
  • Teachers and other carers should be provided with advice and written information relevant to the care of children with cardiomyopathy.

  • In the absence of symptoms and risk factors, children should be allowed to perform low- to moderate-level aerobic physical activity, in accordance with advice from their paediatric cardiologist. Advice on high-intensity exercise in children should be guided by cardiomyopathy phenotype and the presence of symptoms and risk factors within a specialist paediatric cardiomyopathy setting.

  • Provision should be made for children with learning difficulties and other special needs.

  • Parents, teachers, and staff at sports facilities should be trained in CPR and in the use of AEDs.

AED, automated external defibrillator; CPR, cardio-pulmonary resuscitation; ICD, implantable cardioverter defibrillator; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

11. Sex differences in cardiomyopathies

Sex differences in phenotypic expression and outcomes are well documented across cardiovascular medicine. Differences in clinical presentation, progression, and outcome in cardiomyopathies between females and males can be attributable to genetic and hormonal differences, but also to variations in management, access to healthcare, or response to specific therapies.546,1173 Eliminating these variations represents a major unmet need in the care of cardiomyopathies.

Cardiomyopathies are typically inherited as an autosomal dominant trait. Therefore, the prevalence would be expected to be equal among the sexes. Women are consistently less represented than men in clinical studies across different cardiomyopathies (30–40%). Despite this, the difference may be explained by bias in interaction with healthcare facilities or by diagnosis criteria based on unadjusted cardiac imaging measurements; data from large pedigrees seem to support the hypothesis that there is a real delay in the age of phenotypic expression in female carriers (at least for HCM).178,1174–1176

Females with HCM are diagnosed later than males (8–13 years later), are more severely affected, more often have LVOTO, have more severe symptoms at baseline, and more commonly develop advanced heart failure during follow-up.1177,1178 Women with LVOTO and indication for invasive procedures are often older and more symptomatic than males.1179–1181 Females and males appear to show similar survival benefit from invasive SRT.705,1181 Cardiomyopathy-related death has shown to be increased in middle-aged females with HCM compared with men and the general population; this is due to a higher rate of death from heart failure. No difference in SCD has been demonstrated in HCM regarding sex.1182,1183

Females with DCM may have a better response to therapy and seem to have a more favourable clinical course than males.186,1184 Male sex has been reported to be consistently associated with an increased SCD rate in DCM (general cohorts and particular genotypes series),186,541,872,878,1185–1187 and death from heart failure or transplant in general DCM cohorts.1188,1189

Male sex and sports have been traditionally identified as variables associated with an earlier phenotypic penetrance and a more severe disease expression in genetic carriers, and are independent predictors of malignant ventricular arrhythmic events in ARVC.522,950,1190–1195 As in HCM, females with ARVC may have an increased risk of developing heart failure.1193

Reports on sex differences in familial or genetic RCM are scarce.331,546 Compared with other types of cardiomyopathies, females seem to be as equally represented as males in RCM series.331

12. Comorbidities and cardiovascular risk factors in cardiomyopathies

12.1. Cardiovascular risk factors

The penetrance of the disease in genetic carriers of cardiomyopathy-associated variants is incomplete. Gene–environment interactions can explain part of the heterogeneity of the phenotypic expression of all cardiomyopathy phenotypes, although published data focus primarily on HCM, DCM, and ARVC.

12.2. Dilated cardiomyopathy

Individual genetic predisposition favours a dilated phenotype in the presence of trigger factors, such as inflammation, infection, toxic insults from alcohol or drugs, and tachyarrhythmias.

12.3. Hypertrophic cardiomyopathy

Hypertension and obesity have been associated with penetrance and phenotypic expression of HCM.1196 Results from the EORP Cardiomyopathy/Mycarditis Registry showed that patients with HCM had a high prevalence of cardiovascular risk factors, comparable with data from the general population.1196 Hypertension, diabetes, and obesity were associated with older age at presentation, a lower prevalence of family history of HCM and SCD, more symptoms, frequent AF, and worse LV diastolic function.1197 Hypertension and obesity were also associated with higher provocable LVOT gradients and LVH.1198

12.4. Arrhythmogenic right ventricular cardiomyopathy

The role of intense exercise in disease expression and outcomes has been studied in HCM and DCM, but the impact has shown to be particularly relevant in ARVC (Table 25). Despite significant research, the pathophysiology of ARVC is complex and not well understood. The search for genetic or environmental triggers, such as viruses and immune response, has failed to identify actionable factors. The role of inflammation on the pathophysiology is thought to be key.1199

Table 25

Modulators of the phenotypic expression of cardiomyopathies

ConditionHCMDCMARVCExpression
Hypertension+++++?Hypertrophy, dilatation, dysfunction, AF
Diabetes+++?Hypertrophy, dysfunction, AF
Obesity+++?Hypertrophy, LVOTO, AF
Toxic+++?Dilatation, dysfunction
Sports+++++Dilatation, dysfunction, ventricular arrhythmia
Virus+++Dilatation, dysfunction, ventricular arrhythmia
Pregnancy++Dilatation, dysfunction
ConditionHCMDCMARVCExpression
Hypertension+++++?Hypertrophy, dilatation, dysfunction, AF
Diabetes+++?Hypertrophy, dysfunction, AF
Obesity+++?Hypertrophy, LVOTO, AF
Toxic+++?Dilatation, dysfunction
Sports+++++Dilatation, dysfunction, ventricular arrhythmia
Virus+++Dilatation, dysfunction, ventricular arrhythmia
Pregnancy++Dilatation, dysfunction

AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVOTO, left ventricular outflow tract obstruction.

+, degree of positive association; −, absence of definitive association; ?, unknown association.

Table 25

Modulators of the phenotypic expression of cardiomyopathies

ConditionHCMDCMARVCExpression
Hypertension+++++?Hypertrophy, dilatation, dysfunction, AF
Diabetes+++?Hypertrophy, dysfunction, AF
Obesity+++?Hypertrophy, LVOTO, AF
Toxic+++?Dilatation, dysfunction
Sports+++++Dilatation, dysfunction, ventricular arrhythmia
Virus+++Dilatation, dysfunction, ventricular arrhythmia
Pregnancy++Dilatation, dysfunction
ConditionHCMDCMARVCExpression
Hypertension+++++?Hypertrophy, dilatation, dysfunction, AF
Diabetes+++?Hypertrophy, dysfunction, AF
Obesity+++?Hypertrophy, LVOTO, AF
Toxic+++?Dilatation, dysfunction
Sports+++++Dilatation, dysfunction, ventricular arrhythmia
Virus+++Dilatation, dysfunction, ventricular arrhythmia
Pregnancy++Dilatation, dysfunction

AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; LVOTO, left ventricular outflow tract obstruction.

+, degree of positive association; −, absence of definitive association; ?, unknown association.

Recommendation Table 34

Recommendation for management of cardiovascular risk factors in patients with cardiomyopathy

graphic
graphic
Recommendation Table 34

Recommendation for management of cardiovascular risk factors in patients with cardiomyopathy

graphic
graphic

13. Coronavirus disease (COVID-19) and cardiomyopathies

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, known as COVID-19, is characterized by a high variability of clinical presentation and outcomes with an adverse association between underlying cardiac disease, including heart failure, and SARS-CoV-2-related mortality.1200–1202 However, examination of SARS-CoV-2 infection in underlying causes of heart failure, particularly cardiomyopathies, has been limited.

Analyses of international registries on cardiomyopathies and SARS-CoV-2 from the pre-vaccine period have identified several markers of adverse outcomes.1203 Prior history of heart failure and particular phenotypes (amyloidosis and DCM) were significantly associated with intensive care unit admission and death compared with HCM, ARVC, and the general population. For HCM, age, baseline functional class, LVOTO, and systolic impairment were independent predictors of death.1204

SARS-CoV-2 vaccination has been demonstrated to be safe in large population studies and reports on complications related to the vaccination in patients with cardiomyopathy are anecdotal. Given this, and the potential for worse outcomes in cardiomyopathy patients who contract COVID-19, vaccination is encouraged in all cardiomyopathy patients and, in particular, in those with signs or symptoms of heart failure.

14. Key messages

  1. Cardiomyopathies are more common than previously thought and they typically require nuanced management that may differ from the conventional approach to patients with arrhythmia or heart failure.

  2. Aetiology is fundamental to the management of patients with heart muscle disease and careful and systematic description of the morphological and functional phenotype is a crucial first step in the diagnostic pathway.

  3. An approach to nomenclature and diagnosis of cardiomyopathies that is based on the predominant phenotype at presentation is recommended.

  4. Patients with cardiomyopathy may seek medical attention due to symptoms onset (HF or arrhythmia related), incidental abnormal findings, or as a result of family screening following the diagnosis in a relative.

  5. Multimodality imaging to characterize the cardiac phenotype (morphology and function)—including tissue characterization for non-ischaemic myocardial scar detection—is necessary, in combination with a detailed personal and family history, clinical examination, electrocardiography, and laboratory investigations.

  6. Imaging results should always be interpreted in the overall clinical context, including genetic testing results, rather than in isolation.

  7. Tissue characterization by CMR is of value in diagnosis, monitoring of disease progression and risk stratification in each of the main cardiomyopathy phenotypes.

  8. DPD/PYP/HMDP bone-tracer scintigraphy or SPECT represent the gold standard for the diagnosis of ATTR-related cardiac amyloidosis.

  9. The presence of non-ischaemic ventricular scar or fatty replacement on cardiac CMR and/or pathological examination, which can occur with or without ventricular dilatation and/or systolic dysfunction, can be the sole clue to the diagnosis of a cardiomyopathy and can have prognostic significance that varies with aetiology.

  10. The aim of this multiparametric and systemic approach is to generate a phenotype-based aetiological diagnosis, interpreting available data with a cardiomyopathy-oriented mindset that combines cardiological assessment with non-cardiac parameters.

  11. A multidisciplinary approach to patient care and appropriate transition of care from paediatric to adult cardiomyopathy services is needed.

  12. Genetic testing should be performed in patients with cardiomyopathy and may influence risk stratification and management.

  13. Genetic counselling, including pre- and post-test counselling, and psychological support are an essential aspect of the multidisciplinary care of patients with cardiomyopathy and their relatives.

  14. Paediatric cardiomyopathies largely represent part of the same clinical spectrum as those seen in older adolescents and adults, but infant-onset (in the first year of life) cardiomyopathies are often associated with severe phenotypes and a high rate of heart failure-related morbidity and mortality.

  15. Beyond the first year of life, genetic causes of childhood-onset cardiomyopathies are similar to those in adults.

  16. Symptom management, identification, and prevention of disease-related complications (including SCD, heart failure, and stroke) are the cornerstone of management of all cardiomyopathies.

  17. Cardiac myosin inhibitors (Mavacamten) should be considered in patients with HCM and LVOTO who remain symptomatic despite optimal medical therapy.

  18. Validated SCD risk-prediction tools (HCM Risk-SCD and HCM Risk-Kids) are the first step in sudden death prevention in patients with HCM.

  19. Additional risk markers may be of use in patients with low or intermediate risk, but there is a lack of robust data on the impact of these parameters on the personalized risk estimates generated by the risk-prediction tools.

  20. Pharmacological treatment of DCM patients does not differ from those recommended in chronic heart failure.

  21. SCD risk of DCM and NDLVC patients varies depending on the underlying cause and genetic subtype.

  22. CMR findings play an important role in guiding ICD implantation for patients with DCM and NDLVC.

  23. In DCM and NDLVC patients, ICD should be considered for certain genetic forms even if LVEF is >35%.

  24. It is of importance to define aetiology for a tailored management in patients with syndromic and metabolic cardiomyopathies (i.e. ERT/chaperone in lysosomal storage disease; tafamidis in ATTRwt, etc.).

  25. Pregnancy and the post-partum period are associated with increased cardiovascular risk in women with known cardiomyopathy.

  26. A multidisciplinary team should evaluate the patient with cardiomyopathy to assess the risk associated with pregnancy.

  27. Beta-blocker therapy on arrhythmic indication can safely be continued during pregnancy; safety data should be checked before initiation of new drugs in pregnancy.

  28. Healthy adults of all ages and individuals with known cardiac disease should exercise with moderate intensity, totalling at least 150 min per week.

  29. All patients with cardiomyopathy should have an individualized risk assessment for exercise prescription. Evaluation should be guided by three principles: (i) preventing life-threatening arrhythmias during exercise; (ii) symptom management to allow sports; and (iii) preventing sports-induced progression of the arrhythmogenic condition.

  30. Individuals who are genotype positive/phenotype negative or have a mild cardiomyopathy phenotype and absence of symptoms or any risk factors, may be able to participate in competitive sports. In some high-risk patients with HCM, ARVC, and NDLVC, high-intensity exercise and competitive sports should be discouraged.

  31. Patients with high-risk genotypes or associated factors for arrhythmic or heart failure complications or severe LVOTO should be referred for specialized investigations before undergoing elective NCS.

  32. Identification and management of risk factors and concomitant diseases is recommended as an integral part of the management of cardiomyopathy patients.

15. Gaps in evidence

Although there have been major advances in the genetics, diagnosis, and treatments of patients with cardiomyopathy over the last few years, there are a number of areas where robust evidence is still lacking and deserve to be addressed in future clinical research.

  1. Cardiomyopathy phenotypes.

  2. Epidemiology:

    • Prevalence of NDLVC phenotype (children and adults).

    • Systematic assessment of prevalence of cardiomyopathy phenotypes in childhood.

  3. Integrated patient management:

    • Embedding of telemedicine into cardiomyopathy networks.

  4. Patient pathway:

    • Laboratory tests:

      • Studies on novel ‘omic’ biomarkers (proteomics, metabolomics, and transcriptomics) are needed to assess their potential value for diagnostic and prognostic purposes in cardiomyopathies.

    • Multimodality imaging:

      • Advanced echocardiographic techniques, including speckle tracking deformation imaging, are promising but lack robust validation in the setting of cardiomyopathies.

      • A universally accepted, standardized method for the quantification of myocardial fibrosis by CMR is lacking.

      • CMR scans may be performed in patients with compatible implantable devices, but the quality is limited by artefacts.

      • Artificial intelligence enhanced electrocardiography and imaging for cardiomyopathy evaluation has been proving a novel tool to dramatically improve diagnosis and prognosis; further studies are needed for routine introduction in clinical practice.

      • Impact of CMR on screening in genotype-positive relatives of individuals with cardiomyopathy and in gene-elusive families.

    • Genetics:

      • Penetrance is poorly characterized for most pathogenic variants. This is true both for variants found through cascade screening of relatives of a patient with cardiomyopathy, and also for variants found in the wider population who may have clinical sequencing for another indication or may choose to have genome sequencing as a screening test.

      • The benefits, harm, and costs of screening of cardiomyopathy-associated genes in individuals without a personal or family history of cardiomyopathy is not known.

    • General principles in management:

      • Management of RV failure remains largely non-evidence-based.

      • Large-scale studies are required to guide ventricular arrhythmia management in patients with genetic cardiomyopathies.

      • Optimal rate control and AADs per subtype of cardiomyopathy.

      • The role of ICDs in patients with well tolerated VT.

      • All risk calculators are developed using baseline data. Therefore, the utility of their application during follow-up visits of patients remains unclear and needs to be studied.

      • Risk prediction in childhood cardiomyopathies other than HCM remains empirical—multicentre approach required to understand and develop SCD risk models in childhood.

      • Lack of controlled studies on the effect of ablation in patients with AF and cardiomyopathy.

      • Models to predict AF recurrence have not been validated in cardiomyopathy patients.

      • Lack of randomized studies assessing the efficacy of cardiac sympathetic denervation for the prevention of VT/VF recurrences.

    • Approach to paediatric cardiomyopathies:

      • Lack of randomized studies or large registries addressing the benefit and optimal dosing of drug therapy in paediatric population.

  5. Hypertrophic cardiomyopathy:

    • Epidemiology:

      • Imaging and genotype studies suggest a population prevalence of up to 1 in 200 of the population. However, HER-based studies suggest a much lower number of 3–4/10 000. Further studies into the prevalence of clinically important diseases are necessary.

    • Aetiology:

      • Aetiology of gene-elusive disease.

      • The role of polygenic risk.

      • Interaction between comorbidity and disease outcomes.

      • Genetic and environmental determinants of disease expression in variant carriers.

    • Symptom management:

      • Optimal timing of LVOTO management and its impact on disease progression.

      • Prevention of AF and heart failure.

    • Sudden death prevention:

      • Impact of genetics (Mendelian and complex) on risk of disease-related outcomes.

      • Improved prediction models that reduce residual risk and prevent unnecessary ICD implantation.

      • Refinement of risk-prediction models to include serial data.

      • Role of LVOTO in risk prediction in children (apparent discrepancy compared with adults).

    • New therapies:

      • Clinical utility of myosin inhibitors, other small molecules, and emerging genetic therapies.

  6. Dilated cardiomyopathy:

    • Genetic basis of familial DCM is still unknown in a high number of cases.

    • Detailed data about the specific clinical course in diverse genetic and non-genetic DCM forms are not available.

    • It is unknown if patients with DCM respond differently to pharmacological treatment according to underlying aetiology.

    • Optimized SCD prevention strategy remains unsolved. There are not data from prospective clinical trials in modern cohorts with contemporary medical treatment. This gap is knowledge is particularly relevant for DCM patients with LVEF > 35%.

    • Sport recommendations and utility of prophylactic pharmacological therapy to prevent DCM onset in genetic carriers.

  7. Non-dilated left ventricular cardiomyopathy:

    • Prevalence of disease.

    • Natural history and response to treatment.

    • SCD prevention.

    • Sports recommendations.

  8. Arrhythmogenic right ventricular cardiomyopathy:

    • RCTs for therapies for the management of arrhythmias and heart failure are lacking.

    • Studies on the effect of exercise remain largely retrospective.

    • Studies on the incidence and prognostication of heart failure remain limited.

    • Studies on the frequency and mode of clinical screening for asymptomatic family members are lacking.

  9. Restrictive cardiomyopathy:

    • SCD prevention.

  10. Syndromic and metabolic cardiomyopathies:

    • Lack of randomized trials or large observational cohort studies assessing the role of new target therapies addressing the RAS/MAPK pathway (i.e trametinib).

    • There are few long-term outcome studies addressing ventricular remodelling in RAS-HCM.

    • HCM Risk-Kids has not been validated in paediatric patients with RAS-HCM. Data regarding SCD risk stratification are lacking, although candidate risk factors have been identified.

    • Lack of studies addressing the optimal timing to start ERT in adolescents and adults with late-onset Pompe disease.

    • Lack of standardized protocols to treat cross-reactive immunologic material-negative patients.

    • Lack of standardization of clinical endpoints in ERT/chaperone therapy trials.

    • Lack of head-to-head comparisons between agalsidase alpha and beta.

    • Optimal time to begin treatment in asymptomatic female patients with non-classic disease.

  11. Amyloid:

    • Further studies are needed to assess the efficacy and safety of tafamidis in NYHA class III patients.

    • SCD risk stratification and indications for ICD implantation should be carefully defined, taking into account the estimated life expectancy, competitive non-cardiovascular mortality, and the high rate of pulseless electrical activity.

    • The need for drug therapy in patients with cardiac amyloidosis and subclinical cardiac involvement (i.e asymptomatic patients, positive scintigraphy with negative ECHO) has not been clearly defined.

  12. Sports:

    • ‘Return to play’ for patients with low-risk cardiomyopathies (and how to define low risk in relation to exercise).

    • SCD risk and exercise recommendations in phenotype-negative gene carriers.

    • Role of exercise in disease expression and progression.

    • Large, adequately powered randomized prospective studies are necessary to provide evidence-based recommendations for optimal exercise prescription without compromising safety.

  13. Reproductive issues:

    • Several cardiomyopathies lack specific outcome data regarding pregnancy.

    • There is a lack of randomized trials on the use of AADs, heart failure drugs, and interventions during pregnancy.

  14. Non-cardiac interventions:

    • There is a lack specific outcome data regarding risks of non-cardiac interventions.

  15. Management of cardiovascular risk factors in patients with cardiomyopathies:

    • There is a lack of data on the impact of comorbidities on penetrance, severity, and outcome of cardiomyopathies.

16. ‘What to do’ and ‘What not to do’ messages from the Guidelines

17. Supplementary data

Supplementary material is available at European Heart Journal online.

18. Data availability statement

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

19. Author information

Author/Task Force Member Affiliations: Alexandros Protonotarios, Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, United Kingdom, Inherited Cardiovascular Disease Unit, St Bartholomew’s Hospital, London, United Kingdom; Juan R. Gimeno, Inherited Heart Diseases Unit (CSUR /ERN), Hospital Universitario Virgen de la Arrixaca- IMIB- Universidad de Murcia, Murcia, Spain, European Reference Networks for rare, low prevalence and complex diseases of the heart, ERN GUARD-Heart, European Commission 6, Amsterdam, Netherlands; Eloisa Arbustini, Centre For Inherited Cardiovascular Diseases, IRCCS Foundation Policlinico San Matteo, Piazzale Golgi, 27100 Pavia, Italy; Roberto Barriales-Villa, Inherited Cardiovascular Diseases Unit, Cardiology Service, Complexo Hospitalario Universitario A Coruña (CHUAC), A Coruña, Spain, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña, A Coruña, Spain, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, A Coruña, Spain; Cristina Basso, Department of Cardiac, Thoracic, Vascular Sciences and Public Health-University of Padua, Cardiovascular Pathology Unit-Azienda Ospedaliera, Padua, Italy; Connie R. Bezzina, Amsterdam UMC location University of Amsterdam, Department of experimental cardiology, Heart centre, Meibergdreef 9, Amsterdam, Netherlands, Amsterdam cardiovascular sciences, Heart failure and arrhythmias, Amsterdam, Netherlands, European Reference Networks for rare, low prevalence and complex diseases of the heart, ERN GUARD-Heart; Elena Biagini, Cardiology, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, Cardiology, Centro di riferimento europeo delle malattie cardiovascolari, ERN GUARD-Heart, Bologna, Italy; Nico A. Blom, Paediatric Cardiology, Leiden University Medical Center, Leiden, Netherlands, Paediatric Cardiology, Amsterdam University Medical Center, Amsterdam, Netherlands; Rudolf A. de Boer, Erasmus Medical Center, Department of Cardiology, Rotterdam, Netherlands; Tim De Winter (Belgium), ESC Patient Forum, Sophia Antipolis, France; Perry M. Elliott, UCL Institute of Cardiovascular Science University College London, London, United Kingdom, St. Bartholomew’s Hospital, London, United Kingdom; Marcus Flather, Norwich Medical School, University of East Anglia, Norwich, United Kingdom, Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Pablo Garcia-Pavia, Department of cardiology, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBERCV, Madrid, Spain, Centro Nacional de Invstigaciones Cardiovasculares (CNIC), Madrid, Spain, European Reference Networks for rare, low prevalence and complex diseases of the heart, ERN GUARD-Heart, Madrid, Spain; Kristina H. Haugaa, Cardiology, Karolinska University Hospital, Stockholm, Sweden, Cardiology, Oslo University Hospital, Oslo, Norway; Jodie Ingles, Centre for Population Genomics, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Ruxandra Oana Jurcut, Expert Center for Rare Genetic Cardiovascular Diseases, Department of Cardiology, Emergency Institute of Cardiovascular Diseases “Prof.dr.C.C.Iliescu”, Bucharest, Romania, Cardiology, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania; Sabine Klaassen, Experimental and Clinical Research Center, A Cooperation Between the Max Delbrück Center and Charité - Universitätsmedizin Berlin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, Congenital Heart Disease - Pediatric Cardiology, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany, DZHK (German Centre for Cardiovascular Research) partner site Berlin, Berlin, Germany; Giuseppe Limongelli, Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy, Cardiology, Monaldi Hospital - AORN Colli, Naples, Italy, European Reference Network for Rare, Low Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart); Bart Loeys, Center for medical genetics, Antwerp university hospital/university of Antwerp, Antwerp, Belgium, Department of human genetics, Radboud university medical center, Nijmegen, Netherlands; Jens Mogensen, Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark; Iacopo Olivotto, Meyer Children’s Hospital IRCCS, University of Florence, Florence, Italy; Antonis Pantazis, Royal Brompton, and Harefield Hospitals, London, United Kingdom; Sanjay Sharma, St George's, University of London, London, United Kingdom, St George's University Hospital NHS Foundation Trust, London, United Kingdom; J. Peter van Tintelen, Department of Genetics, University Medical Center Utrecht, Utrecht, Netherlands; and James S. Ware, National Heart & Lung Institute, Imperial College London, London, United Kingdom, MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom, Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom.

20. Appendix

ESC Scientific Document Group

Includes Document Reviewers and ESC National Cardiac Societies.

Document Reviewers: Philippe Charron (CPG Review Co-ordinator) (France), Massimo Imazio (CPG Review Co-ordinator) (Italy), Magdy Abdelhamid (Egypt), Victor Aboyans (France), Michael Arad (Israel), Folkert W. Asselbergs (Netherlands), Riccardo Asteggiano (Italy), Zofia Bilinska (Poland), Damien Bonnet (France), Henning Bundgaard (Denmark), Nuno Miguel Cardim (Portugal), Jelena Čelutkienė (Lithuania), Maja Cikes (Croatia), Gaetano Maria De Ferrari (Italy), Veronica Dusi (Italy), Volkmar Falk (Germany), Laurent Fauchier (France), Estelle Gandjbakhch (France), Tiina Heliö (Finland), Konstantinos Koskinas (Switzerland), Dipak Kotecha (United Kingdom), Ulf Landmesser (Germany), George Lazaros (Greece), Basil S. Lewis (Israel), Ales Linhart (Czechia), Maja-Lisa Løchen (Norway), Benjamin Meder (Germany), Richard Mindham (United Kingdom), James Moon (United Kingdom), Jens Cosedis Nielsen (Denmark), Steffen Petersen (United Kingdom), Eva Prescott (Denmark), Mary N. Sheppard (United Kingdom), Gianfranco Sinagra (Italy), Marta Sitges (Spain), Jacob Tfelt-Hansen (Denmark), Rhian Touyz (Canada), Rogier Veltrop (Netherlands), Josef Veselka (Czechia), Karim Wahbi (France), Arthur Wilde (Netherlands), and Katja Zeppenfeld (Netherlands).

ESC National Cardiac Societies actively involved in the review process of the 2023 ESC Guidelines for the management of cardiomyopathies:

Algeria: Algerian Society of Cardiology, Brahim Kichou; Armenia: Armenian Cardiologists Association, Hamayak Sisakian; Austria: Austrian Society of Cardiology, Daniel Scherr; Belgium: Belgian Society of Cardiology, Bernhard Gerber; Bosnia and Herzegovina: Association of Cardiologists of Bosnia and Herzegovina, Alen Džubur; Bulgaria: Bulgarian Society of Cardiology, Mariana Gospodinova; Croatia: Croatian Cardiac Society, Ivo Planinc; Cyprus: Cyprus Society of Cardiology, Hera Heracleous Moustra; Czechia: Czech Society of Cardiology, David Zemánek; Denmark: Danish Society of Cardiology, Morten Steen Kvistholm Jensen; Egypt: Egyptian Society of Cardiology, Ahmad Samir; Estonia: Estonian Society of Cardiology, Kairit Palm; Finland: Finnish Cardiac Society, Tiina Heliö; France: French Society of Cardiology, Karim,Wahbi; Germany: German Cardiac Society, Eric Schulze-Bahr; Greece: Hellenic Society of Cardiology, Vlachopoulos Haralambos; Hungary: Hungarian Society of Cardiology Róbert Sepp; Iceland: Icelandic Society of Cardiology, Berglind Aðalsteinsdóttir; Ireland: Irish Cardiac Society, Deirdre Ward; Israel: Israel Heart Society, Miry Blich; Italy: Italian Federation of Cardiology, Gianfranco Sinagra; Kosovo (Republic of): Kosovo Society of Cardiology, Afrim Poniku; Kyrgyzstan: Kyrgyz Society of Cardiology, Olga Lunegova, Latvia: Latvian Society of Cardiology, Ainars Rudzitis; Lebanon: Lebanese Society of Cardiology, Roland Kassab; Lithuania: Lithuanian Society of Cardiology, Jūratė Barysienė; Luxembourg: Luxembourg Society of Cardiology, Steve Huijnen; Malta: Maltese Cardiac Society, Tiziana Felice; Moldova (Republic of): Moldavian Society of Cardiology, Eleonora Vataman; Montenegro: Montenegro Society of Cardiology, Nikola Pavlovic; Morocco: Moroccan Society of Cardiology, Nawal Doghmi; Netherlands: Netherlands Society of Cardiology, Folkert W. Asselbergs; North Macedonia: The National Society of Cardiology of North Macedonia, Elizabeta Srbinovska Kostovska; Norway: Norwegian Society of Cardiology, Vibeke Marie Almaas; Poland: Polish Cardiac Society, Elżbieta Katarzyna Biernacka; Portugal: Portuguese Society of Cardiology, Dulce Brito; Romania: Romanian Society of Cardiology, Monica Rosca; San Marino: San Marino Society of Cardiology, Marco Zavatta; Serbia: Cardiology Society of Serbia, Arsen Ristic; Slovakia: Slovak Society of Cardiology, Eva Goncalvesová; Slovenia: Slovenian Society of Cardiology, Matjaž Šinkovec; Spain: Spanish Society of Cardiology, Victoria Cañadas-Godoy; Sweden: Swedish Society of Cardiology, Pyotr G. Platonov; Switzerland: Swiss Society of Cardiology, Ardan M. Saguner; Syrian Arab Republic: Syrian Cardiovascular Association, Ahmad Rasheed Al Saadi; Tunisia: Tunisian Society of Cardiology and Cardiovascular Surgery, Ikram Kammoun; Türkiye: Turkish Society of Cardiology, Ahmet Celik; Ukraine: Ukrainian Association of Cardiology, Elena Nesukay; and Uzbekistan: Association of Cardiologists of Uzbekistan, Timur Abdullaev.

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 Pasquet (Belgium), Amina Rakisheva (Kazakhstan), Bianca Rocca (Italy), Xavier Rossello (Spain), Ilonca Vaartjes (Netherlands), Christiaan Vrints (Belgium), Adam Witkowski (Poland), and Katja Zeppenfeld (Netherlands).

21. Acknowledgements

The Task Force Chairs thank Sebastian Onciul for providing cardiac magnetic resonance images in Figure 7.

22. References

1

Authors/Task Force Members
;
Elliott
 
PM
,
Anastasakis
 
A
,
Borger
 
MA
,
Borggrefe
 
M
,
Cecchi
 
F
, et al.  
2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy: the Task Force for the Diagnosis and Management of Hypertrophic Cardiomyopathy of the European Society of Cardiology (ESC)
.
Eur Heart J
 
2014
;
35
:
2733
2779
. https://doi.org/10.1093/eurheartj/ehu284

2

Elliott
 
P
,
Andersson
 
B
,
Arbustini
 
E
,
Bilinska
 
Z
,
Cecchi
 
F
,
Charron
 
P
, et al.  
Classification of the cardiomyopathies: a position statement from the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases
.
Eur Heart J
 
2008
;
29
:
270
276
. https://doi.org/10.1093/eurheartj/ehm342

3

Wilde
 
AAM
,
Semsarian
 
C
,
Marquez
 
MF
,
Shamloo
 
AS
,
Ackerman
 
MJ
,
Ashley
 
EA
, et al.  
European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases
.
Europace
 
2022
;
24
:
1307
1367
. https://doi.org/10.1093/europace/euac030

4

Towbin
 
JA
,
McKenna
 
WJ
,
Abrams
 
DJ
,
Ackerman
 
MJ
,
Calkins
 
H
,
Darrieux
 
FCC
, et al.  
2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy
.
Heart Rhythm
 
2019
;
16
:
e301
e372
. https://doi.org/10.1016/j.hrthm.2019.05.007

5

Corrado
 
D
,
Perazzolo Marra
 
M
,
Zorzi
 
A
,
Beffagna
 
G
,
Cipriani
 
A
,
Lazzari
 
M
, et al.  
Diagnosis of arrhythmogenic cardiomyopathy: the Padua criteria
.
Int J Cardiol
 
2020
;
319
:
106
114
. https://doi.org/10.1016/j.ijcard.2020.06.005

6

Biesecker
 
LG
,
Adam
 
MP
,
Alkuraya
 
FS
,
Amemiya
 
AR
,
Bamshad
 
MJ
,
Beck
 
AE
, et al.  
A dyadic approach to the delineation of diagnostic entities in clinical genomics
.
Am J Hum Genet
 
2021
;
108
:
8
15
. https://doi.org/10.1016/j.ajhg.2020.11.013

7

Arbustini
 
E
,
Narula
 
N
,
Dec
 
GW
,
Reddy
 
KS
,
Greenberg
 
B
,
Kushwaha
 
S
, et al.  
The MOGE(S) classification for a phenotype-genotype nomenclature of cardiomyopathy: endorsed by the World Heart Federation
.
J Am Coll Cardiol
 
2013
;
62
:
2046
2072
. https://doi.org/10.1016/j.jacc.2013.08.1644

8

Disertori
 
M
,
Quintarelli
 
S
,
Grasso
 
M
,
Pilotto
 
A
,
Narula
 
N
,
Favalli
 
V
, et al.  
Autosomal recessive atrial dilated cardiomyopathy with standstill evolution associated with mutation of Natriuretic Peptide Precursor A
.
Circ Cardiovasc Genet
 
2013
;
6
:
27
36
. https://doi.org/10.1161/CIRCGENETICS.112.963520

9

Pinto
 
YM
,
Elliott
 
PM
,
Arbustini
 
E
,
Adler
 
Y
,
Anastasakis
 
A
,
Bohm
 
M
, et al.  
Proposal for a revised definition of dilated cardiomyopathy, hypokinetic non-dilated cardiomyopathy, and its implications for clinical practice: a position statement of the ESC working group on myocardial and pericardial diseases
.
Eur Heart J
 
2016
;
37
:
1850
1858
. https://doi.org/10.1093/eurheartj/ehv727

10

Marcus
 
FI
,
McKenna
 
WJ
,
Sherrill
 
D
,
Basso
 
C
,
Bauce
 
B
,
Bluemke
 
DA
, et al.  
Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the Task Force Criteria
.
Eur Heart J
 
2010
;
31
:
806
14
. https://doi.org/10.1093/eurheartj/ehq025

11

Rapezzi
 
C
,
Aimo
 
A
,
Barison
 
A
,
Emdin
 
M
,
Porcari
 
A
,
Linhart
 
A
, et al.  
Restrictive cardiomyopathy: definition and diagnosis
.
Eur Heart J
 
2022
;
43
:
4679
4693
. https://doi.org/10.1093/eurheartj/ehac543

12

van Waning
 
JI
,
Caliskan
 
K
,
Michels
 
M
,
Schinkel
 
AFL
,
Hirsch
 
A
,
Dalinghaus
 
M
, et al.  
Cardiac phenotypes, genetics, and risks in familial noncompaction cardiomyopathy
.
J Am Coll Cardiol
 
2019
;
73
:
1601
1611
. https://doi.org/10.1016/j.jacc.2018.12.085

13

Sedaghat-Hamedani
 
F
,
Haas
 
J
,
Zhu
 
F
,
Geier
 
C
,
Kayvanpour
 
E
,
Liss
 
M
, et al.  
Clinical genetics and outcome of left ventricular non-compaction cardiomyopathy
.
Eur Heart J
 
2017
;
38
:
3449
3460
. https://doi.org/10.1093/eurheartj/ehx545

14

Klaassen
 
S
,
Probst
 
S
,
Oechslin
 
E
,
Gerull
 
B
,
Krings
 
G
,
Schuler
 
P
, et al.  
Mutations in sarcomere protein genes in left ventricular noncompaction
.
Circulation
 
2008
;
117
:
2893
2901
. https://doi.org/10.1161/CIRCULATIONAHA.107.746164

15

Probst
 
S
,
Oechslin
 
E
,
Schuler
 
P
,
Greutmann
 
M
,
Boye
 
P
,
Knirsch
 
W
, et al.  
Sarcomere gene mutations in isolated left ventricular noncompaction cardiomyopathy do not predict clinical phenotype
.
Circ Cardiovasc Genet
 
2011
;
4
:
367
374
. https://doi.org/10.1161/CIRCGENETICS.110.959270

16

Hoedemaekers
 
YM
,
Caliskan
 
K
,
Michels
 
M
,
Frohn-Mulder
 
I
,
van der Smagt
 
JJ
,
Phefferkorn
 
JE
.
The importance of genetic counseling, DNA diagnostics, and cardiologic family screening in left ventricular noncompaction cardiomyopathy
.
Circ Cardiovasc Genet
 
2010
;
3
:
232
239
. https://doi.org/10.1161/CIRCGENETICS.109.903898

17

Gati
 
S
,
Papadakis
 
M
,
Papamichael
 
ND
,
Zaidi
 
A
,
Sheikh
 
N
,
Reed
 
M
, et al.  
Reversible de novo left ventricular trabeculations in pregnant women: implications for the diagnosis of left ventricular noncompaction in low-risk populations
.
Circulation
 
2014
;
130
:
475
483
. https://doi.org/10.1161/CIRCULATIONAHA.114.008554

18

Gati
 
S
,
Chandra
 
N
,
Bennett
 
RL
,
Reed
 
M
,
Kervio
 
G
,
Panoulas
 
VF
, et al.  
Increased left ventricular trabeculation in highly trained athletes: do we need more stringent criteria for the diagnosis of left ventricular non-compaction in athletes?
 
Heart
 
2013
;
99
:
401
408
. https://doi.org/10.1136/heartjnl-2012-303418

19

de la Chica
 
JA
,
Gomez-Talavera
 
S
,
Garcia-Ruiz
 
JM
,
Garcia-Lunar
 
I
,
Oliva
 
B
,
Fernandez-Alvira
 
JM
, et al.  
Association between left ventricular noncompaction and vigorous physical activity
.
J Am Coll Cardiol
 
2020
;
76
:
1723
1733
. https://doi.org/10.1016/j.jacc.2020.08.030

20

Jensen
 
B
,
van der Wal
 
AC
,
Moorman
 
AFM
,
Christoffels
 
VM
.
Excessive trabeculations in noncompaction do not have the embryonic identity
.
Int J Cardiol
 
2017
;
227
:
325
330
. https://doi.org/10.1016/j.ijcard.2016.11.089

21

Faber
 
JW
,
D’Silva
 
A
,
Christoffels
 
VM
,
Jensen
 
B
.
Lack of morphometric evidence for ventricular compaction in humans
.
J Cardiol
 
2021
;
78
:
397
405
. https://doi.org/10.1016/j.jjcc.2021.03.006

22

Anderson
 
RH
,
Jensen
 
B
,
Mohun
 
TJ
,
Petersen
 
SE
,
Aung
 
N
,
Zemrak
 
F
, et al.  
Key questions relating to left ventricular noncompaction cardiomyopathy: is the emperor still wearing any clothes?
 
Can J Cardiol
 
2017
;
33
:
747
757
. https://doi.org/10.1016/j.cjca.2017.01.017

23

Lyon
 
AR
,
Bossone
 
E
,
Schneider
 
B
,
Sechtem
 
U
,
Citro
 
R
,
Underwood
 
SR
, et al.  
Current state of knowledge on Takotsubo syndrome: a Position Statement from the Taskforce on Takotsubo Syndrome of the Heart Failure Association of the European Society of Cardiology
.
Eur J Heart Fail
 
2016
;
18
:
8
27
. https://doi.org/10.1002/ejhf.424

24

McKenna
 
WJ
,
Maron
 
BJ
,
Thiene
 
G
.
Classification, epidemiology, and global burden of cardiomyopathies
.
Circ Res
 
2017
;
121
:
722
730
. https://doi.org/10.1161/CIRCRESAHA.117.309711

25

Hershberger
 
RE
,
Hedges
 
DJ
,
Morales
 
A
.
Dilated cardiomyopathy: the complexity of a diverse genetic architecture
.
Nat Rev Cardiol
 
2013
;
10
:
531
547
. https://doi.org/10.1038/nrcardio.2013.105

26

Hada
 
Y
,
Sakamoto
 
T
,
Amano
 
K
,
Yamaguchi
 
T
,
Takenaka
 
K
,
Takahashi
 
H
, et al.  
Prevalence of hypertrophic cardiomyopathy in a population of adult Japanese workers as detected by echocardiographic screening
.
Am J Cardiol
 
1987
;
59
:
183
184
. https://doi.org/10.1016/S0002-9149(87)80107-8

27

Agnarsson
 
UT
,
Hardarson
 
T
,
Hallgrimsson
 
J
,
Sigfusson
 
N
.
The prevalence of hypertrophic cardiomyopathy in men: an echocardiographic population screening study with a review of death records
.
J Intern Med
 
1992
;
232
:
499
506
. https://doi.org/10.1111/j.1365-2796.1992.tb00623.x

28

Maron
 
BJ
,
Gardin
 
JM
,
Flack
 
JM
,
Gidding
 
SS
,
Kurosaki
 
TT
,
Bild
 
DE
.
Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary Artery Risk Development in (Young) Adults
.
Circulation
 
1995
;
92
:
785
789
. https://doi.org/10.1161/01.CIR.92.4.785

29

Maron
 
BJ
,
Mathenge
 
R
,
Casey
 
SA
,
Poliac
 
LC
,
Longe
 
TF
.
Clinical profile of hypertrophic cardiomyopathy identified de novo in rural communities
.
J Am Coll Cardiol
 
1999
;
33
:
1590
1595
. https://doi.org/10.1016/S0735-1097(99)00039-X

30

Maron
 
BJ
,
Spirito
 
P
,
Roman
 
MJ
,
Paranicas
 
M
,
Okin
 
PM
,
Best
 
LG
, et al.  
Prevalence of hypertrophic cardiomyopathy in a population-based sample of American Indians aged 51 to 77 years (the Strong Heart Study)
.
Am J Cardiol
 
2004
;
93
:
1510
1514
. https://doi.org/10.1016/j.amjcard.2004.03.007

31

Zou
 
Y
,
Song
 
L
,
Wang
 
Z
,
Ma
 
A
,
Liu
 
T
,
Gu
 
H
, et al.  
Prevalence of idiopathic hypertrophic cardiomyopathy in China: a population-based echocardiographic analysis of 8080 adults
.
Am J Med
 
2004
;
116
:
14
18
. https://doi.org/10.1016/j.amjmed.2003.05.009

32

Maro
 
EE
,
Janabi
 
M
,
Kaushik
 
R
.
Clinical and echocardiographic study of hypertrophic cardiomyopathy in Tanzania
.
Trop Doct
 
2006
;
36
:
225
227
. https://doi.org/10.1258/004947506778604904

33

Basavarajaiah
 
S
,
Wilson
 
M
,
Whyte
 
G
,
Shah
 
A
,
McKenna
 
W
,
Sharma
 
S
.
Prevalence of hypertrophic cardiomyopathy in highly trained athletes: relevance to pre-participation screening
.
J Am Coll Cardiol
 
2008
;
51
:
1033
1039
. https://doi.org/10.1016/j.jacc.2007.10.055

34

Nugent
 
AW
,
Daubeney
 
PE
,
Chondros
 
P
,
Carlin
 
JB
,
Cheung
 
M
,
Wilkinson
 
LC
, et al.  
The epidemiology of childhood cardiomyopathy in Australia
.
N Engl J Med
 
2003
;
348
:
1639
1646
. https://doi.org/10.1056/NEJMoa021737

35

Lipshultz
 
SE
,
Sleeper
 
LA
,
Towbin
 
JA
,
Lowe
 
AM
,
Orav
 
EJ
,
Cox
 
GF
, et al.  
The incidence of pediatric cardiomyopathy in two regions of the United States
.
N Engl J Med
 
2003
;
348
:
1647
1655
. https://doi.org/10.1056/NEJMoa021715

36

Arola
 
A
,
Jokinen
 
E
,
Ruuskanen
 
O
,
Saraste
 
M
,
Pesonen
 
E
,
Kuusela
 
AL
, et al.  
Epidemiology of idiopathic cardiomyopathies in children and adolescents. A nationwide study in Finland
.
Am J Epidemiol
 
1997
;
146
:
385
393
. https://doi.org/10.1093/oxfordjournals.aje.a009291

37

Codd
 
MB
,
Sugrue
 
DD
,
Gersh
 
BJ
,
Melton
 
LJ
 III
.
Epidemiology of idiopathic dilated and hypertrophic cardiomyopathy. A population-based study in Olmsted County, Minnesota, 1975–1984
.
Circulation
 
1989
;
80
:
564
572
. https://doi.org/10.1161/01.CIR.80.3.564

38

Andrews
 
RE
,
Fenton
 
MJ
,
Ridout
 
DA
,
Burch
 
M
.
New-onset heart failure due to heart muscle disease in childhood: a prospective study in the United Kingdom and Ireland
.
Circulation
 
2008
;
117
:
79
84
. https://doi.org/10.1161/CIRCULATIONAHA.106.671735

39

Peters
 
S
,
Trümmel
 
M
,
Meyners
 
W
.
Prevalence of right ventricular dysplasia-cardiomyopathy in a non-referral hospital
.
Int J Cardiol
 
2004
;
97
:
499
501
. https://doi.org/10.1016/j.ijcard.2003.10.037

40

Corrado
 
D
,
Basso
 
C
,
Pavei
 
A
,
Michieli
 
P
,
Schiavon
 
M
,
Thiene
 
G
.
Trends in sudden cardiovascular death in young competitive athletes after implementation of a preparticipation screening program
.
JAMA
 
2006
;
296
:
1593
1601
. https://doi.org/10.1001/jama.296.13.1593

41

Migliore
 
F
,
Zorzi
 
A
,
Michieli
 
P
,
Perazzolo Marra
 
M
,
Siciliano
 
M
,
Rigato
 
I
, et al.  
Prevalence of cardiomyopathy in Italian asymptomatic children with electrocardiographic T-wave inversion at preparticipation screening
.
Circulation
 
2012
;
125
:
529
538
. https://doi.org/10.1161/CIRCULATIONAHA.111.055673

42

Ware
 
JS
,
Amor-Salamanca
 
A
,
Tayal
 
U
,
Govind
 
R
,
Serrano
 
I
,
Salazar-Mendiguchia
 
J
, et al.  
Genetic etiology for alcohol-induced cardiac toxicity
.
J Am Coll Cardiol
 
2018
;
71
:
2293
2302
. https://doi.org/10.1016/j.jacc.2018.03.462

43

Garcia-Pavia
 
P
,
Kim
 
Y
,
Restrepo-Cordoba
 
MA
,
Lunde
 
IG
,
Wakimoto
 
H
,
Smith
 
AM
, et al.  
Genetic variants associated with cancer therapy-induced cardiomyopathy
.
Circulation
 
2019
;
140
:
31
41
. https://doi.org/10.1161/CIRCULATIONAHA.118.037934

44

Ware
 
JS
,
Li
 
J
,
Mazaika
 
E
,
Yasso
 
CM
,
DeSouza
 
T
,
Cappola
 
TP
, et al.  
Shared genetic predisposition in peripartum and dilated cardiomyopathies
.
N Engl J Med
 
2016
;
374
:
233
241
. https://doi.org/10.1056/NEJMoa1505517

45

Goli
 
R
,
Li
 
J
,
Brandimarto
 
J
,
Levine
 
LD
,
Riis
 
V
,
McAfee
 
Q
, et al.  
Genetic and phenotypic landscape of peripartum cardiomyopathy
.
Circulation
 
2021
;
143
:
1852
1862
. https://doi.org/10.1161/CIRCULATIONAHA.120.052395

46

Doheny
 
D
,
Srinivasan
 
R
,
Pagant
 
S
,
Chen
 
B
,
Yasuda
 
M
,
Desnick
 
RJ
.
Fabry disease: prevalence of affected males and heterozygotes with pathogenic GLA mutations identified by screening renal, cardiac and stroke clinics, 1995–2017
.
J Med Genet
 
2018
;
55
:
261
268
. https://doi.org/10.1136/jmedgenet-2017-105080

47

Tini
 
G
,
Sessarego
 
E
,
Benenati
 
S
,
Vianello
 
PF
,
Musumeci
 
B
,
Autore
 
C
, et al.  
Yield of bone scintigraphy screening for transthyretin-related cardiac amyloidosis in different conditions: methodological issues and clinical implications
.
Eur J Clin Invest
 
2021
;
51
:
e13665
. https://doi.org/10.1111/eci.13665

48

Aimo
 
A
,
Merlo
 
M
,
Porcari
 
A
,
Georgiopoulos
 
G
,
Pagura
 
L
,
Vergaro
 
G
, et al.  
Redefining the epidemiology of cardiac amyloidosis. A systematic review and meta-analysis of screening studies
.
Eur J Heart Fail
 
2022
;
24
:
2342
2351
. https://doi.org/10.1002/ejhf.2532

49

Lota
 
AS
,
Hazebroek
 
MR
,
Theotokis
 
P
,
Wassall
 
R
,
Salmi
 
S
,
Halliday
 
BP
, et al.  
Genetic architecture of acute myocarditis and the overlap with inherited cardiomyopathy
.
Circulation
 
2022
;
146
:
1123
1134
. https://doi.org/10.1161/CIRCULATIONAHA.121.058457

50

Tiron
 
C
,
Campuzano
 
O
,
Fernandez-Falgueras
 
A
,
Alcalde
 
M
,
Loma-Osorio
 
P
,
Zamora
 
E
, et al.  
Prevalence of pathogenic variants in cardiomyopathy-associated genes in myocarditis
.
Circ Genom Precis Med
 
2022
;
15
:
e003408
. https://doi.org/10.1161/CIRCGEN.121.003408

51

Seidel
 
F
,
Holtgrewe
 
M
,
Al-Wakeel-Marquard
 
N
,
Opgen-Rhein
 
B
,
Dartsch
 
J
,
Herbst
 
C
, et al.  
Pathogenic variants associated with dilated cardiomyopathy predict outcome in pediatric myocarditis
.
Circ Genom Precis Med
 
2021
;
14
:
e003250
. https://doi.org/10.1161/CIRCGEN.120.003250

52

Ammirati
 
E
,
Raimondi
 
F
,
Piriou
 
N
,
Sardo Infirri
 
L
,
Mohiddin
 
SA
,
Mazzanti
 
A
, et al.  
Acute myocarditis associated with desmosomal gene variants
.
JACC Heart Fail
 
2022
;
10
:
714
727
. https://doi.org/10.1016/j.jchf.2022.06.013

53

Cardim
 
N
,
Freitas
 
A
,
Brito
 
D
.
From hypertrophic cardiomyopathy centers to inherited cardiovascular disease centers in Europe. A small or a major step? A position paper from the Nucleus of the Working Group on Myocardial and Pericardial Diseases of the Portuguese Society of Cardiology
.
Rev Port Cardiol
 
2011
;
30
:
829
835
. https://doi.org/10.1016/j.repc.2011.09.005

54

Barriales-Villa
 
R
,
Gimeno-Blanes
 
JR
,
Zorio-Grima
 
E
,
Ripoll-Vera
 
T
,
Evangelista-Masip
 
A
,
Moya-Mitjans
 
A
, et al.  
Plan of action for inherited cardiovascular diseases: synthesis of recommendations and action algorithms
.
Rev Esp Cardiol
 
2016
;
69
:
300
309
. https://doi.org/10.1016/j.recesp.2015.11.031

55

Vriz
 
O
,
AlSergani
 
H
,
Elshaer
 
AN
,
Shaik
 
A
,
Mushtaq
 
AH
,
Lioncino
 
M
, et al.  
A complex unit for a complex disease: the HCM-Family Unit
.
Monaldi Arch Chest Dis
 
2021
;
92
. https://doi.org/10.4081/monaldi.2021.2147

56

Basso
 
C
,
Aguilera
 
B
,
Banner
 
J
,
Cohle
 
S
,
d’Amati
 
G
,
de Gouveia
 
RH
, et al.  
Guidelines for autopsy investigation of sudden cardiac death: 2017 update from the Association for European Cardiovascular Pathology
.
Virchows Arch
 
2017
;
471
:
691
705
. https://doi.org/10.1007/s00428-017-2221-0

57

Fellmann
 
F
,
van El
 
CG
,
Charron
 
P
,
Michaud
 
K
,
Howard
 
HC
,
Boers
 
SN
, et al.  
European recommendations integrating genetic testing into multidisciplinary management of sudden cardiac death
.
Eur J Hum Genet
 
2019
;
27
:
1763
1773
. https://doi.org/10.1038/s41431-019-0445-y

58

de Hosson
 
M
,
Goossens
 
PJJ
,
De Backer
 
J
,
De Wolf
 
D
,
Van Hecke
 
A
.
Needs and experiences of adolescents with congenital heart disease and parents in the transitional process: a qualitative study
.
J Pediatr Nurs
 
2021
;
61
:
90
95
. https://doi.org/10.1016/j.pedn.2021.03.016

59

de Hosson
 
M
,
De Backer
 
J
,
De Wolf
 
D
,
De Groote
 
K
,
Demulier
 
L
,
Mels
 
S
, et al.  
Development of a transition program for adolescents with congenital heart disease
.
Eur J Pediatr
 
2020
;
179
:
339
348
. https://doi.org/10.1007/s00431-019-03515-4

60

Tini
 
G
,
Vianello
 
PF
,
Rizzola
 
G
,
La Malfa
 
G
,
Porto
 
I
,
Canepa
 
M
.
Telehealth monitoring for hypertrophic cardiomyopathy and amyloid cardiomyopathy patients: lessons from the coronavirus disease 2019 lockdown in Italy
.
J Cardiovasc Med
 
2020
;
21
:
622
623
. https://doi.org/10.2459/JCM.0000000000001024

61

Directive 2011/24/EU of the European Parliament and of the Council of 9 March 2011. In: The European Parliament and the Council of the European Union, (ed); 2011
. https://eur-lex.europa.eu/legal-content/EN/TXT/HTML/?uri=CELEX:52022DC0210  (5 April 2023 date last accessed).

62

Rapezzi
 
C
,
Arbustini
 
E
,
Caforio
 
AL
,
Charron
 
P
,
Gimeno-Blanes
 
J
,
Helio
 
T
, et al.  
Diagnostic work-up in cardiomyopathies: bridging the gap between clinical phenotypes and final diagnosis. A position statement from the ESC Working Group on Myocardial and Pericardial Diseases
.
Eur Heart J
 
2013
;
34
:
1448
1458
. https://doi.org/10.1093/eurheartj/ehs397

63

Elliott
 
P
,
Charron
 
P
,
Blanes
 
JR
,
Tavazzi
 
L
,
Tendera
 
M
,
Konte
 
M
, et al.  
European cardiomyopathy pilot registry: EURObservational research programme of the European Society of Cardiology
.
Eur Heart J
 
2016
;
37
:
164
173
. https://doi.org/10.1093/eurheartj/ehv497

64

van Velzen
 
HG
,
Schinkel
 
AFL
,
Baart
 
SJ
,
Oldenburg
 
RA
,
Frohn-Mulder
 
IME
,
van Slegtenhorst
 
MA
, et al.  
Outcomes of contemporary family screening in hypertrophic cardiomyopathy
.
Circ Genom Precis Med
 
2018
;
11
:
e001896
. https://doi.org/10.1161/CIRCGEN.117.001896

65

Ranthe
 
MF
,
Carstensen
 
L
,
Oyen
 
N
,
Jensen
 
MK
,
Axelsson
 
A
,
Wohlfahrt
 
J
, et al.  
Risk of cardiomyopathy in younger persons with a family history of death from cardiomyopathy: a nationwide family study in a cohort of 3.9 million persons
.
Circulation
 
2015
;
132
:
1013
1019
. https://doi.org/10.1161/CIRCULATIONAHA.114.013478

66

Gimeno
 
JR
,
Lacunza
 
J
,
Garcia-Alberola
 
A
,
Cerdan
 
MC
,
Oliva
 
MJ
,
Garcia-Molina
 
E
, et al.  
Penetrance and risk profile in inherited cardiac diseases studied in a dedicated screening clinic
.
Am J Cardiol
 
2009
;
104
:
406
410
. https://doi.org/10.1016/j.amjcard.2009.03.055

67

Ploski
 
R
,
Rydzanicz
 
M
,
Ksiazczyk
 
TM
,
Franaszczyk
 
M
,
Pollak
 
A
,
Kosinska
 
J
, et al.  
Evidence for troponin C (TNNC1) as a gene for autosomal recessive restrictive cardiomyopathy with fatal outcome in infancy
.
Am J Med Genet A
 
2016
;
170
:
3241
3248
. https://doi.org/10.1002/ajmg.a.37860

68

Surmacz
 
R
,
Franaszczyk
 
M
,
Pyda
 
M
,
Ploski
 
R
,
Bilinska
 
ZT
,
Bobkowski
 
W
.
Autosomal recessive transmission of familial nonsyndromic dilated cardiomyopathy due to compound desmoplakin gene mutations
.
Pol Arch Intern Med
 
2018
;
128
:
785
787
. https://doi.org/10.20452/pamw.4365

69

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
3726
. https://doi.org/10.1093/eurheartj/ehab368

69a

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
3639
. https://doi.org/10.1093/eurheartj/ehad195.
In press.

70

Bonny
 
A
,
Lellouche
 
N
,
Ditah
 
I
,
Hidden-Lucet
 
F
,
Yitemben
 
MT
,
Granger
 
B
, et al.  
C-reactive protein in arrhythmogenic right ventricular dysplasia/cardiomyopathy and relationship with ventricular tachycardia
.
Cardiol Res Pract
 
2010
;
2010
:
919783
. https://doi.org/10.4061/2010/919783

71

Donal
 
E
,
Delgado
 
V
,
Bucciarelli-Ducci
 
C
,
Galli
 
E
,
Haugaa
 
KH
,
Charron
 
P
, et al.  
Multimodality imaging in the diagnosis, risk stratification, and management of patients with dilated cardiomyopathies: an expert consensus document from the European Association of Cardiovascular Imaging
.
Eur Heart J Cardiovasc Imaging
 
2019
;
20
:
1075
1093
. https://doi.org/10.1093/ehjci/jez178

72

Haugaa
 
KH
,
Basso
 
C
,
Badano
 
LP
,
Bucciarelli-Ducci
 
C
,
Cardim
 
N
,
Gaemperli
 
O
, et al.  
Comprehensive multi-modality imaging approach in arrhythmogenic cardiomyopathy–an expert consensus document of the European Association of Cardiovascular Imaging
.
Eur Heart J Cardiovasc Imaging
 
2017
;
18
:
237
253
. https://doi.org/10.1093/ehjci/jew229

73

Lang
 
RM
,
Badano
 
LP
,
Mor-Avi
 
V
,
Afilalo
 
J
,
Armstrong
 
A
,
Ernande
 
L
, et al.  
Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging
.
Eur Heart J Cardiovasc Imaging
 
2015
;
16
:
233
270
. https://doi.org/10.1093/ehjci/jev014

74

Lancellotti
 
P
,
Nkomo
 
VT
,
Badano
 
LP
,
Bergler-Klein
 
J
,
Bogaert
 
J
,
Davin
 
L
, et al.  
Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography
.
Eur Heart J Cardiovasc Imaging
 
2013
;
14
:
721
740
. https://doi.org/10.1093/ehjci/jet123

75

Charron
 
P
,
Arad
 
M
,
Arbustini
 
E
,
Basso
 
C
,
Bilinska
 
Z
,
Elliott
 
P
, et al.  
Genetic counselling and testing in cardiomyopathies: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases
.
Eur Heart J
 
2010
;
31
:
2715
2726
. https://doi.org/10.1093/eurheartj/ehq271

76

Liu
 
D
,
Hu
 
K
,
Nordbeck
 
P
,
Ertl
 
G
,
Störk
 
S
,
Weidemann
 
F
.
Longitudinal strain bull’s eye plot patterns in patients with cardiomyopathy and concentric left ventricular hypertrophy
.
Eur J Med Res
 
2016
;
21
:
21
. https://doi.org/10.1186/s40001-016-0216-y

77

Haugaa
 
KH
,
Hasselberg
 
NE
,
Edvardsen
 
T
.
Mechanical dispersion by strain echocardiography: a predictor of ventricular arrhythmias in subjects with lamin A/C mutations
.
JACC Cardiovasc Imaging
 
2015
;
8
:
104
106
. https://doi.org/10.1016/j.jcmg.2014.04.029

78

Haugaa
 
KH
,
Goebel
 
B
,
Dahlslett
 
T
,
Meyer
 
K
,
Jung
 
C
,
Lauten
 
A
, et al.  
Risk assessment of ventricular arrhythmias in patients with nonischemic dilated cardiomyopathy by strain echocardiography
.
J Am Soc Echocardiogr
 
2012
;
25
:
667
673
. https://doi.org/10.1016/j.echo.2012.02.004

79

Leren
 
IS
,
Saberniak
 
J
,
Haland
 
TF
,
Edvardsen
 
T
,
Haugaa
 
KH
.
Combination of ECG and echocardiography for identification of arrhythmic events in early ARVC
.
JACC Cardiovasc Imaging
 
2017
;
10
:
503
513
. https://doi.org/10.1016/j.jcmg.2016.06.011

80

Haland
 
TF
,
Almaas
 
VM
,
Hasselberg
 
NE
,
Saberniak
 
J
,
Leren
 
IS
,
Hopp
 
E
, et al.  
Strain echocardiography is related to fibrosis and ventricular arrhythmias in hypertrophic cardiomyopathy
.
Eur Heart J Cardiovasc Imaging
 
2016
;
17
:
613
621
. https://doi.org/10.1093/ehjci/jew005

81

Norrish
 
G
,
Ding
 
T
,
Field
 
E
,
Ziolkowska
 
L
,
Olivotto
 
I
,
Limongelli
 
G
, et al.  
Development of a novel risk prediction model for sudden cardiac death in childhood hypertrophic cardiomyopathy (HCM Risk-Kids)
.
JAMA Cardiol
 
2019
;
4
:
918
927
. https://doi.org/10.1001/jamacardio.2019.2861

82

Lopez
 
L
,
Frommelt
 
PC
,
Colan
 
SD
,
Trachtenberg
 
FL
,
Gongwer
 
R
,
Stylianou
 
M
, et al.  
Pediatric heart network echocardiographic Z scores: comparison with other published models
.
J Am Soc Echocardiogr
 
2021
;
34
:
185
192
. https://doi.org/10.1016/j.echo.2020.09.019

83

Adabag
 
AS
,
Kuskowski
 
MA
,
Maron
 
BJ
.
Determinants for clinical diagnosis of hypertrophic cardiomyopathy
.
Am J Cardiol
 
2006
;
98
:
1507
1511
. https://doi.org/10.1016/j.amjcard.2006.07.029

84

Klues
 
HG
,
Schiffers
 
A
,
Maron
 
BJ
.
Phenotypic spectrum and patterns of left ventricular hypertrophy in hypertrophic cardiomyopathy: morphologic observations and significance as assessed by two-dimensional echocardiography in 600 patients
.
J Am Coll Cardiol
 
1995
;
26
:
1699
1708
. https://doi.org/10.1016/0735-1097(95)00390-8

85

Shapiro
 
LM
,
McKenna
 
WJ
.
Distribution of left ventricular hypertrophy in hypertrophic cardiomyopathy: a two-dimensional echocardiographic study
.
J Am Coll Cardiol
 
1983
;
2
:
437
444
. https://doi.org/10.1016/S0735-1097(83)80269-1

86

Maron
 
MS
,
Olivotto
 
I
,
Betocchi
 
S
,
Casey
 
SA
,
Lesser
 
JR
,
Losi
 
MA
, et al.  
Effect of left ventricular outflow tract obstruction on clinical outcome in hypertrophic cardiomyopathy
.
N Engl J Med
 
2003
;
348
:
295
303
. https://doi.org/10.1056/NEJMoa021332

87

Nistri
 
S
,
Olivotto
 
I
,
Betocchi
 
S
,
Losi
 
MA
,
Valsecchi
 
G
,
Pinamonti
 
B
, et al.  
Prognostic significance of left atrial size in patients with hypertrophic cardiomyopathy (from the Italian Registry for Hypertrophic Cardiomyopathy)
.
Am J Cardiol
 
2006
;
98
:
960
965
. https://doi.org/10.1016/j.amjcard.2006.05.013

88

Debonnaire
 
P
,
Joyce
 
E
,
Hiemstra
 
Y
,
Mertens
 
BJ
,
Atsma
 
DE
,
Schalij
 
MJ
, et al.  
Left atrial size and function in hypertrophic cardiomyopathy patients and risk of new-onset atrial fibrillation
.
Circ Arrhythm Electrophysiol
 
2017
;
10
:
e004052
. https://doi.org/10.1161/CIRCEP.116.004052

89

Harris
 
KM
,
Spirito
 
P
,
Maron
 
MS
,
Zenovich
 
AG
,
Formisano
 
F
,
Lesser
 
JR
, et al.  
Prevalence, clinical profile, and significance of left ventricular remodeling in the end-stage phase of hypertrophic cardiomyopathy
.
Circulation
 
2006
;
114
:
216
225
. https://doi.org/10.1161/CIRCULATIONAHA.105.583500

90

Alba
 
AC
,
Gaztanaga
 
J
,
Foroutan
 
F
,
Thavendiranathan
 
P
,
Merlo
 
M
,
Alonso-Rodriguez
 
D
, et al.  
Prognostic value of late gadolinium enhancement for the prediction of cardiovascular outcomes in dilated cardiomyopathy: an international, multi-institutional study of the MINICOR group
.
Circ Cardiovasc Imaging
 
2020
;
13
:
e010105
. https://doi.org/10.1161/CIRCIMAGING.119.010105

91

Leong
 
DP
,
Chakrabarty
 
A
,
Shipp
 
N
,
Molaee
 
P
,
Madsen
 
PL
,
Joerg
 
L
, et al.  
Effects of myocardial fibrosis and ventricular dyssynchrony on response to therapy in new-presentation idiopathic dilated cardiomyopathy: insights from cardiovascular magnetic resonance and echocardiography
.
Eur Heart J
 
2012
;
33
:
640
648
. https://doi.org/10.1093/eurheartj/ehr391

92

Yoerger
 
DM
,
Marcus
 
F
,
Sherrill
 
D
,
Calkins
 
H
,
Towbin
 
JA
,
Zareba
 
W
, et al.  
Echocardiographic findings in patients meeting task force criteria for arrhythmogenic right ventricular dysplasia: new insights from the multidisciplinary study of right ventricular dysplasia
.
J Am Coll Cardiol
 
2005
;
45
:
860
865
. https://doi.org/10.1016/j.jacc.2004.10.070

93

Pieles
 
GE
,
Grosse-Wortmann
 
L
,
Hader
 
M
,
Fatah
 
M
,
Chungsomprasong
 
P
,
Slorach
 
C
, et al.  
Association of echocardiographic parameters of right ventricular remodeling and myocardial performance with modified task force criteria in adolescents with arrhythmogenic right ventricular cardiomyopathy
.
Circ Cardiovasc Imaging
 
2019
;
12
:
e007693
. https://doi.org/10.1161/CIRCIMAGING.118.007693

94

Mehta
 
D
,
Lubitz
 
SA
,
Frankel
 
Z
,
Wisnivesky
 
JP
,
Einstein
 
AJ
,
Goldman
 
M
, et al.  
Cardiac involvement in patients with sarcoidosis: diagnostic and prognostic value of outpatient testing
.
Chest
 
2008
;
133
:
1426
1435
. https://doi.org/10.1378/chest.07-2784

95

Skold
 
CM
,
Larsen
 
FF
,
Rasmussen
 
E
,
Pehrsson
 
SK
,
Eklund
 
AG
.
Determination of cardiac involvement in sarcoidosis by magnetic resonance imaging and Doppler echocardiography
.
J Intern Med
 
2002
;
252
:
465
471
. https://doi.org/10.1046/j.1365-2796.2002.01058.x

96

Joyce
 
E
,
Ninaber
 
MK
,
Katsanos
 
S
,
Debonnaire
 
P
,
Kamperidis
 
V
,
Bax
 
JJ
, et al.  
Subclinical left ventricular dysfunction by echocardiographic speckle–tracking strain analysis relates to outcome in sarcoidosis
.
Eur J Heart Fail
 
2015
;
17
:
51
62
. https://doi.org/10.1002/ejhf.205

97

Pagourelias
 
ED
,
Mirea
 
O
,
Duchenne
 
J
,
Van Cleemput
 
J
,
Delforge
 
M
,
Bogaert
 
J
, et al.  
Echo parameters for differential diagnosis in cardiac amyloidosis: a head-to-head comparison of deformation and nondeformation parameters
.
Circ Cardiovasc Imaging
 
2017
;
10
:
e005588
. https://doi.org/10.1161/CIRCIMAGING.116.005588

98

Phelan
 
D
,
Collier
 
P
,
Thavendiranathan
 
P
,
Popovic
 
ZB
,
Hanna
 
M
,
Plana
 
JC
, et al.  
Relative apical sparing of longitudinal strain using two-dimensional speckle-tracking echocardiography is both sensitive and specific for the diagnosis of cardiac amyloidosis
.
Heart
 
2012
;
98
:
1442
1448
. https://doi.org/10.1136/heartjnl-2012-302353

99

Linhart
 
A
,
Kampmann
 
C
,
Zamorano
 
JL
,
Sunder-Plassmann
 
G
,
Beck
 
M
,
Mehta
 
A
, et al.  
Cardiac manifestations of Anderson–Fabry disease: results from the international Fabry outcome survey
.
Eur Heart J
 
2007
;
28
:
1228
1235
. https://doi.org/10.1093/eurheartj/ehm153

100

Boldrini
 
M
,
Cappelli
 
F
,
Chacko
 
L
,
Restrepo-Cordoba
 
MA
,
Lopez-Sainz
 
A
,
Giannoni
 
A
, et al.  
Multiparametric echocardiography scores for the diagnosis of cardiac amyloidosis
.
JACC Cardiovasc Imaging
 
2020
;
13
:
909
920
. https://doi.org/10.1016/j.jcmg.2019.10.011

101

Pieroni
 
M
,
Chimenti
 
C
,
De Cobelli
 
F
,
Morgante
 
E
,
Del Maschio
 
A
,
Gaudio
 
C
, et al.  
Fabry’s disease cardiomyopathy: echocardiographic detection of endomyocardial glycosphingolipid compartmentalization
.
J Am Coll Cardiol
 
2006
;
47
:
1663
1671
. https://doi.org/10.1016/j.jacc.2005.11.070

102

Zemrak
 
F
,
Ahlman
 
MA
,
Captur
 
G
,
Mohiddin
 
SA
,
Kawel-Boehm
 
N
,
Prince
 
MR
, et al.  
The relationship of left ventricular trabeculation to ventricular function and structure over a 9.5-year follow-up: the MESA study
.
J Am Coll Cardiol
 
2014
;
64
:
1971
1980
. https://doi.org/10.1016/j.jacc.2014.08.035

103

Steeden
 
JA
,
Quail
 
M
,
Gotschy
 
A
,
Mortensen
 
KH
,
Hauptmann
 
A
,
Arridge
 
S
, et al.  
Rapid whole-heart CMR with single volume super-resolution
.
J Cardiovasc Magn Reson
 
2020
;
22
:
56
. https://doi.org/10.1186/s12968-020-00651-x

104

Kiblboeck
 
D
,
Reiter
 
C
,
Kammler
 
J
,
Schmit
 
P
,
Blessberger
 
H
,
Kellermair
 
J
, et al.  
Artefacts in 1.5 Tesla and 3 Tesla cardiovascular magnetic resonance imaging in patients with leadless cardiac pacemakers
.
J Cardiovasc Magn Reson
 
2018
;
20
:
47
. https://doi.org/10.1186/s12968-018-0469-4

105

Rajiah
 
P
,
Kay
 
F
,
Bolen
 
M
,
Patel
 
AR
,
Landeras
 
L
.
Cardiac magnetic resonance in patients with cardiac implantable electronic devices: challenges and solutions
.
J Thorac Imaging
 
2020
;
35
:
W1
W17
. https://doi.org/10.1097/RTI.0000000000000462

106

Gandjbakhch
 
E
,
Dacher
 
JN
,
Taieb
 
J
,
Chauvin
 
M
,
Anselme
 
F
,
Bartoli
 
A
, et al.  
Joint Position Paper of the Working Group of Pacing and Electrophysiology of the French Society of Cardiology and the French Society of Diagnostic and Interventional Cardiac and Vascular Imaging on magnetic resonance imaging in patients with cardiac electronic implantable devices
.
Arch Cardiovasc Dis
 
2020
;
113
:
473
484
.

107

Nazarian
 
S
,
Hansford
 
R
,
Rahsepar
 
AA
,
Weltin
 
V
,
McVeigh
 
D
,
Gucuk Ipek
 
E
, et al.  
Safety of magnetic resonance imaging in patients with cardiac devices
.
N Engl J Med
 
2017
;
377
:
2555
2564
. https://doi.org/10.1056/NEJMoa1604267

108

Russo
 
RJ
,
Costa
 
HS
,
Silva
 
PD
,
Anderson
 
JL
,
Arshad
 
A
,
Biederman
 
RW
, et al.  
Assessing the risks associated with MRI in patients with a pacemaker or defibrillator
.
N Engl J Med
 
2017
;
376
:
755
764
. https://doi.org/10.1056/NEJMoa1603265

109

Gakenheimer-Smith
 
L
,
Etheridge
 
SP
,
Niu
 
MC
,
Ou
 
Z
,
Presson
 
AP
,
Whitaker
 
P
, et al.  
MRI in pediatric and congenital heart disease patients with CIEDs and epicardial or abandoned leads
.
Pacing Clin Electrophysiol
 
2020
;
43
:
797
804
. https://doi.org/10.1111/pace.13984

110

Vigen
 
KK
,
Reeder
 
SB
,
Hood
 
MN
,
Steckner
 
M
,
Leiner
 
T
,
Dombroski
 
DA
, et al.  
Recommendations for imaging patients with cardiac implantable electronic devices (CIEDs)
.
J Magn Reson Imaging
 
2021
;
53
:
1311
1317
. https://doi.org/10.1002/jmri.27320

111

Bhuva
 
AN
,
Feuchter
 
P
,
Hawkins
 
A
,
Cash
 
L
,
Boubertakh
 
R
,
Evanson
 
J
, et al.  
MRI for patients with cardiac implantable electronic devices: simplifying complexity with a ‘one-stop’ service model
.
BMJ Qual Saf
 
2019
;
28
:
853
858
. https://doi.org/10.1136/bmjqs-2018-009079

112

Seewoster
 
T
,
Lobe
 
S
,
Hilbert
 
S
,
Bollmann
 
A
,
Sommer
 
P
,
Lindemann
 
F
, et al.  
Cardiovascular magnetic resonance imaging in patients with cardiac implantable electronic devices: best practice and real-world experience
.
Europace
 
2019
;
21
:
1220
1228
. https://doi.org/10.1093/europace/euz112

113

Stühlinger
 
M
,
Burri
 
H
,
Vernooy
 
K
,
Garcia
 
R
,
Lenarczyk
 
R
,
Sultan
 
A
, et al.  
EHRA consensus on prevention and management of interference due to medical procedures in patients with cardiac implantable electronic devices
.
Europace
 
2022
;
24
:
1512
1537
. https://doi.org/10.1093/europace/euac040

114

Primary
 
P
,
Ian Paterson
 
D
,
White
 
JA
,
Butler
 
CR
,
Connelly
 
KA
,
Guerra
 
PG
, et al.  
2021 Update on safety of magnetic resonance imaging: joint statement from Canadian Cardiovascular Society/Canadian Society for Cardiovascular Magnetic Resonance/Canadian Heart Rhythm Society
.
Can J Cardiol
 
2021
;
37
:
835
847
. https://doi.org/10.1016/j.cjca.2021.02.012

115

Messroghli
 
DR
,
Moon
 
JC
,
Ferreira
 
VM
,
Grosse-Wortmann
 
L
,
He
 
T
,
Kellman
 
P
, et al.  
Clinical recommendations for cardiovascular magnetic resonance mapping of T1, T2, T2* and extracellular volume: a consensus statement by the Society for Cardiovascular Magnetic Resonance (SCMR) endorsed by the European Association for Cardiovascular Imaging (EACVI)
.
J Cardiovasc Magn Reson
 
2017
;
19
:
75
. https://doi.org/10.1186/s12968-017-0389-8

116

Baggiano
 
A
,
Boldrini
 
M
,
Martinez-Naharro
 
A
,
Kotecha
 
T
,
Petrie
 
A
,
Rezk
 
T
, et al.  
Noncontrast magnetic resonance for the diagnosis of cardiac amyloidosis
.
JACC Cardiovasc Imaging
 
2020
;
13
:
69
80
. https://doi.org/10.1016/j.jcmg.2019.03.026

117

Nordin
 
S
,
Kozor
 
R
,
Vijapurapu
 
R
,
Augusto
 
JB
,
Knott
 
KD
,
Captur
 
G
, et al.  
Myocardial storage, inflammation, and cardiac phenotype in Fabry disease after one year of enzyme replacement therapy
.
Circ Cardiovasc Imaging
 
2019
;
12
:
e009430
. https://doi.org/10.1161/CIRCIMAGING.119.009430

118

Ray
 
JG
,
Vermeulen
 
MJ
,
Bharatha
 
A
,
Montanera
 
WJ
,
Park
 
AL
.
Association between MRI exposure during pregnancy and fetal and childhood outcomes
.
JAMA
 
2016
;
316
:
952
961
. https://doi.org/10.1001/jama.2016.12126

119

Andreini
 
D
,
Dello Russo
 
A
,
Pontone
 
G
,
Mushtaq
 
S
,
Conte
 
E
,
Perchinunno
 
M
, et al.  
CMR for identifying the substrate of ventricular arrhythmia in patients with normal echocardiography
.
JACC Cardiovasc Imaging
 
2020
;
13
:
410
421
. https://doi.org/10.1016/j.jcmg.2019.04.023

120

Halliday
 
BP
,
Baksi
 
AJ
,
Gulati
 
A
,
Ali
 
A
,
Newsome
 
S
,
Izgi
 
C
, et al.  
Outcome in dilated cardiomyopathy related to the extent, location, and pattern of late gadolinium enhancement
.
JACC Cardiovasc Imaging
 
2019
;
12
:
1645
1655
. https://doi.org/10.1016/j.jcmg.2018.07.015

121

Barison
 
A
,
Aimo
 
A
,
Ortalda
 
A
,
Todiere
 
G
,
Grigoratos
 
C
,
Passino
 
C
, et al.  
Late gadolinium enhancement as a predictor of functional recovery, need for defibrillator implantation and prognosis in non-ischemic dilated cardiomyopathy
.
Int J Cardiol
 
2018
;
250
:
195
200
. https://doi.org/10.1016/j.ijcard.2017.10.043

122

Holmstrom
 
M
,
Kivisto
 
S
,
Helio
 
T
,
Jurkko
 
R
,
Kaartinen
 
M
,
Antila
 
M
, et al.  
Late gadolinium enhanced cardiovascular magnetic resonance of lamin A/C gene mutation related dilated cardiomyopathy
.
J Cardiovasc Magn Reson
 
2011
;
13
:
30
. https://doi.org/10.1186/1532-429X-13-30

123

Olivotto
 
I
,
Maron
 
MS
,
Autore
 
C
,
Lesser
 
JR
,
Rega
 
L
,
Casolo
 
G
, et al.  
Assessment and significance of left ventricular mass by cardiovascular magnetic resonance in hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2008
;
52
:
559
566
. https://doi.org/10.1016/j.jacc.2008.04.047

124

Neubauer
 
S
,
Kolm
 
P
,
Ho
 
CY
,
Kwong
 
RY
,
Desai
 
MY
,
Dolman
 
SF
, et al.  
Distinct subgroups in hypertrophic cardiomyopathy in the NHLBI HCM registry
.
J Am Coll Cardiol
 
2019
;
74
:
2333
2345
. https://doi.org/10.1016/j.jacc.2019.08.1057

125

Miller
 
RJH
,
Heidary
 
S
,
Pavlovic
 
A
,
Schlachter
 
A
,
Dash
 
R
,
Fleischmann
 
D
, et al.  
Defining genotype-phenotype relationships in patients with hypertrophic cardiomyopathy using cardiovascular magnetic resonance imaging
.
PLoS One
 
2019
;
14
:
e0217612
. https://doi.org/10.1371/journal.pone.0217612

126

Quarta
 
G
,
Husain
 
SI
,
Flett
 
AS
,
Sado
 
DM
,
Chao
 
CY
,
Tome Esteban
 
MT
, et al.  
Arrhythmogenic right ventricular cardiomyopathy mimics: role of cardiovascular magnetic resonance
.
J Cardiovasc Magn Reson
 
2013
;
15
:
16
. https://doi.org/10.1186/1532-429X-15-16

127

Menghetti
 
L
,
Basso
 
C
,
Nava
 
A
,
Angelini
 
A
,
Thiene
 
G
.
Spin-echo nuclear magnetic resonance for tissue characterisation in arrhythmogenic right ventricular cardiomyopathy
.
Heart
 
1996
;
76
:
467
470
. https://doi.org/10.1136/hrt.76.6.467

128

Sen-Chowdhry
 
S
,
Syrris
 
P
,
Ward
 
D
,
Asimaki
 
A
,
Sevdalis
 
E
,
McKenna
 
WJ
.
Clinical and genetic characterization of families with arrhythmogenic right ventricular dysplasia/cardiomyopathy provides novel insights into patterns of disease expression
.
Circulation
 
2007
;
115
:
1710
1720
. https://doi.org/10.1161/CIRCULATIONAHA.106.660241

129

Aquaro
 
GD
,
Barison
 
A
,
Todiere
 
G
,
Grigoratos
 
C
,
Ait Ali
 
L
,
Di Bella
 
G
, et al.  
Usefulness of combined functional assessment by cardiac magnetic resonance and tissue characterization versus task force criteria for diagnosis of arrhythmogenic right ventricular cardiomyopathy
.
Am J Cardiol
 
2016
;
118
:
1730
1736
. https://doi.org/10.1016/j.amjcard.2016.08.056

130

Petersen
 
SE
,
Selvanayagam
 
JB
,
Wiesmann
 
F
,
Robson
 
MD
,
Francis
 
JM
,
Anderson
 
RH
, et al.  
Left ventricular non-compaction: insights from cardiovascular magnetic resonance imaging
.
J Am Coll Cardiol
 
2005
;
46
:
101
105
. https://doi.org/10.1016/j.jacc.2005.03.045

131

Masso
 
AH
,
Uribe
 
C
,
Willerson
 
JT
,
Cheong
 
BY
,
Davis
 
BR
.
Left ventricular noncompaction detected by cardiac magnetic resonance screening: a reexamination of diagnostic criteria
.
Tex Heart Inst J
 
2020
;
47
:
183
193
. https://doi.org/10.14503/THIJ-19-7157

132

Grothoff
 
M
,
Pachowsky
 
M
,
Hoffmann
 
J
,
Posch
 
M
,
Klaassen
 
S
,
Lehmkuhl
 
L
, et al.  
Value of cardiovascular MR in diagnosing left ventricular non-compaction cardiomyopathy and in discriminating between other cardiomyopathies
.
Eur Radiol
 
2012
;
22
:
2699
2709
. https://doi.org/10.1007/s00330-012-2554-7

133

Jacquier
 
A
,
Thuny
 
F
,
Jop
 
B
,
Giorgi
 
R
,
Cohen
 
F
,
Gaubert
 
JY
, et al.  
Measurement of trabeculated left ventricular mass using cardiac magnetic resonance imaging in the diagnosis of left ventricular non-compaction
.
Eur Heart J
 
2010
;
31
:
1098
1104
. https://doi.org/10.1093/eurheartj/ehp595

134

Sado
 
DM
,
White
 
SK
,
Piechnik
 
SK
,
Banypersad
 
SM
,
Treibel
 
T
,
Captur
 
G
, et al.  
Identification and assessment of Anderson–Fabry disease by cardiovascular magnetic resonance noncontrast myocardial T1 mapping
.
Circ Cardiovasc Imaging
 
2013
;
6
:
392
398
. https://doi.org/10.1161/CIRCIMAGING.112.000070

135

Deva
 
DP
,
Hanneman
 
K
,
Li
 
Q
,
Ng
 
MY
,
Wasim
 
S
,
Morel
 
C
, et al.  
Cardiovascular magnetic resonance demonstration of the spectrum of morphological phenotypes and patterns of myocardial scarring in Anderson–Fabry disease
.
J Cardiovasc Magn Reson
 
2016
;
18
:
14
. https://doi.org/10.1186/s12968-016-0233-6

136

Francone
 
M
.
Role of cardiac magnetic resonance in the evaluation of dilated cardiomyopathy: diagnostic contribution and prognostic significance
.
ISRN Radiol
 
2014
;
2014
:
365404
. https://doi.org/10.1155/2014/365404

137

Di Marco
 
A
,
Anguera
 
I
,
Schmitt
 
M
,
Klem
 
I
,
Neilan
 
TG
,
White
 
JA
, et al.  
Late gadolinium enhancement and the risk for ventricular arrhythmias or sudden death in dilated cardiomyopathy: systematic review and meta-analysis
.
JACC Heart Fail
 
2017
;
5
:
28
38
. https://doi.org/10.1016/j.jchf.2016.09.017

138

Klem
 
I
,
Klein
 
M
,
Khan
 
M
,
Yang
 
EY
,
Nabi
 
F
,
Ivanov
 
A
, et al.  
Relationship of LVEF and myocardial scar to long-term mortality risk and mode of death in patients with nonischemic cardiomyopathy
.
Circulation
 
2021
;
143
:
1343
1358
. https://doi.org/10.1161/CIRCULATIONAHA.120.048477

139

Rastegar
 
N
,
Te Riele
 
ASJM
,
James
 
CA
,
Bhonsale
 
A
,
Murray
 
B
,
Tichnell
 
C
, et al.  
Fibrofatty changes: incidence at cardiac MR imaging in patients with arrhythmogenic right ventricular dysplasia/cardiomyopathy
.
Radiology
 
2016
;
280
:
405
412
. https://doi.org/10.1148/radiol.2016150988

140

te Riele
 
ASJM
,
Bhonsale
 
A
,
James
 
CA
,
Rastegar
 
N
,
Murray
 
B
,
Burt
 
JR
, et al.  
Incremental value of cardiac magnetic resonance imaging in arrhythmic risk stratification of arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers
.
J Am Coll Cardiol
 
2013
;
62
:
1761
1769
. https://doi.org/10.1016/j.jacc.2012.11.087

141

Chan
 
RH
,
Maron
 
BJ
,
Olivotto
 
I
,
Pencina
 
MJ
,
Assenza
 
GE
,
Haas
 
T
, et al.  
Prognostic value of quantitative contrast-enhanced cardiovascular magnetic resonance for the evaluation of sudden death risk in patients with hypertrophic cardiomyopathy
.
Circulation
 
2014
;
130
:
484
495
. https://doi.org/10.1161/CIRCULATIONAHA.113.007094

142

He
 
D
,
Ye
 
M
,
Zhang
 
L
,
Jiang
 
B
.
Prognostic significance of late gadolinium enhancement on cardiac magnetic resonance in patients with hypertrophic cardiomyopathy
.
Heart Lung
 
2018
;
47
:
122
126
. https://doi.org/10.1016/j.hrtlng.2017.10.008

143

Weissler-Snir
 
A
,
Dorian
 
P
,
Rakowski
 
H
,
Care
 
M
,
Spears
 
D
.
Primary prevention implantable cardioverter-defibrillators in hypertrophic cardiomyopathy–are there predictors of appropriate therapy?
 
Heart Rhythm
 
2021
;
18
:
63
70
. https://doi.org/10.1016/j.hrthm.2020.08.009

144

Raman
 
B
,
Ariga
 
R
,
Spartera
 
M
,
Sivalokanathan
 
S
,
Chan
 
K
,
Dass
 
S
, et al.  
Progression of myocardial fibrosis in hypertrophic cardiomyopathy: mechanisms and clinical implications
.
Eur Heart J Cardiovasc Imaging
 
2019
;
20
:
157
167
. https://doi.org/10.1093/ehjci/jey135

145

Ho
 
CY
,
Abbasi
 
SA
,
Neilan
 
TG
,
Shah
 
RV
,
Chen
 
Y
,
Heydari
 
B
, et al.  
T1 measurements identify extracellular volume expansion in hypertrophic cardiomyopathy sarcomere mutation carriers with and without left ventricular hypertrophy
.
Circ Cardiovasc Imaging
 
2013
;
6
:
415
422
. https://doi.org/10.1161/CIRCIMAGING.112.000333

146

Moon
 
JC
,
Fisher
 
NG
,
McKenna
 
WJ
,
Pennell
 
DJ
.
Detection of apical hypertrophic cardiomyopathy by cardiovascular magnetic resonance in patients with non-diagnostic echocardiography
.
Heart
 
2004
;
90
:
645
649
. https://doi.org/10.1136/hrt.2003.014969

147

Aquaro
 
GD
,
De Luca
 
A
,
Cappelletto
 
C
,
Raimondi
 
F
,
Bianco
 
F
,
Botto
 
N
, et al.  
Prognostic value of magnetic resonance phenotype in patients with arrhythmogenic right ventricular cardiomyopathy
.
J Am Coll Cardiol
 
2020
;
75
:
2753
2765
. https://doi.org/10.1016/j.jacc.2020.04.023

148

Martinez-Naharro
 
A
,
Abdel-Gadir
 
A
,
Treibel
 
TA
,
Zumbo
 
G
,
Knight
 
DS
,
Rosmini
 
S
, et al.  
CMR-verified regression of cardiac AL amyloid after chemotherapy
.
JACC Cardiovasc Imaging
 
2018
;
11
:
152
154
. https://doi.org/10.1016/j.jcmg.2017.02.012

149

Fontana
 
M
,
Martinez-Naharro
 
A
,
Chacko
 
L
,
Rowczenio
 
D
,
Gilbertson
 
JA
,
Whelan
 
CJ
, et al.  
Reduction in CMR derived extracellular volume with patisiran indicates cardiac amyloid regression
.
JACC Cardiovasc Imaging
 
2021
;
14
:
189
199
. https://doi.org/10.1016/j.jcmg.2020.07.043

150

Martinez-Naharro
 
A
,
Kotecha
 
T
,
Norrington
 
K
,
Boldrini
 
M
,
Rezk
 
T
,
Quarta
 
C
, et al.  
Native T1 and extracellular volume in transthyretin amyloidosis
.
JACC Cardiovasc Imaging
 
2019
;
12
:
810
819
. https://doi.org/10.1016/j.jcmg.2018.02.006

151

Puntmann
 
VO
,
Isted
 
A
,
Hinojar
 
R
,
Foote
 
L
,
Carr-White
 
G
,
Nagel
 
E
.
T1 and T2 mapping in recognition of early cardiac involvement in systemic sarcoidosis
.
Radiology
 
2017
;
285
:
63
72
. https://doi.org/10.1148/radiol.2017162732

152

Pennell
 
DJ
,
Porter
 
JB
,
Cappellini
 
MD
,
Chan
 
LL
,
El-Beshlawy
 
A
,
Aydinok
 
Y
, et al.  
Deferasirox for up to 3 years leads to continued improvement of myocardial T2* in patients with beta-thalassemia major
.
Haematologica
 
2012
;
97
:
842
848
. https://doi.org/10.3324/haematol.2011.049957

153

Dalal
 
D
,
Tandri
 
H
,
Judge
 
DP
,
Amat
 
N
,
Macedo
 
R
,
Jain
 
R
, et al.  
Morphologic variants of familial arrhythmogenic right ventricular dysplasia/cardiomyopathy a genetics–magnetic resonance imaging correlation study
.
J Am Coll Cardiol
 
2009
;
53
:
1289
1299
. https://doi.org/10.1016/j.jacc.2008.12.045

154

Stokke
 
MK
,
Castrini
 
AI
,
Aneq
 
MA
,
Jensen
 
HK
,
Madsen
 
T
,
Hansen
 
J
, et al.  
Absence of ECG Task Force Criteria does not rule out structural changes in genotype positive ARVC patients
.
Int J Cardiol
 
2020
;
317
:
152
158
. https://doi.org/10.1016/j.ijcard.2020.05.095

155

Moon
 
JC
,
Sachdev
 
B
,
Elkington
 
AG
,
McKenna
 
WJ
,
Mehta
 
A
,
Pennell
 
DJ
, et al.  
Gadolinium enhanced cardiovascular magnetic resonance in Anderson–Fabry disease. Evidence for a disease specific abnormality of the myocardial interstitium
.
Eur Heart J
 
2003
;
24
:
2151
2155
. https://doi.org/10.1016/j.ehj.2003.09.017

156

Huurman
 
R
,
van der Velde
 
N
,
Schinkel
 
AFL
,
Hassing
 
HC
,
Budde
 
RPJ
,
van Slegtenhorst
 
MA
, et al.  
Contemporary family screening in hypertrophic cardiomyopathy: the role of cardiovascular magnetic resonance
.
Eur Heart J Cardiovasc Imaging
 
2022
;
23
:
1144
1154
. https://doi.org/10.1093/ehjci/jeac099

157

Valente
 
AM
,
Lakdawala
 
NK
,
Powell
 
AJ
,
Evans
 
SP
,
Cirino
 
AL
,
Orav
 
EJ
, et al.  
Comparison of echocardiographic and cardiac magnetic resonance imaging in hypertrophic cardiomyopathy sarcomere mutation carriers without left ventricular hypertrophy
.
Circ Cardiovasc Genet
 
2013
;
6
:
230
237
. https://doi.org/10.1161/CIRCGENETICS.113.000037

158

Germans
 
T
,
Russel
 
IK
,
Gotte
 
MJ
,
Spreeuwenberg
 
MD
,
Doevendans
 
PA
,
Pinto
 
YM
, et al.  
How do hypertrophic cardiomyopathy mutations affect myocardial function in carriers with normal wall thickness? Assessment with cardiovascular magnetic resonance
.
J Cardiovasc Magn Reson
 
2010
;
12
:
13
. https://doi.org/10.1186/1532-429X-12-13

159

Germans
 
T
,
Wilde
 
AA
,
Dijkmans
 
PA
,
Chai
 
W
,
Kamp
 
O
,
Pinto
 
YM
, et al.  
Structural abnormalities of the inferoseptal left ventricular wall detected by cardiac magnetic resonance imaging in carriers of hypertrophic cardiomyopathy mutations
.
J Am Coll Cardiol
 
2006
;
48
:
2518
2523
. https://doi.org/10.1016/j.jacc.2006.08.036

160

Aziz
 
W
,
Claridge
 
S
,
Ntalas
 
I
,
Gould
 
J
,
de Vecchi
 
A
,
Razeghi
 
O
, et al.  
Emerging role of cardiac computed tomography in heart failure
.
ESC Heart Fail
 
2019
;
6
:
909
920
. https://doi.org/10.1002/ehf2.12479

161

Galand
 
V
,
Ghoshhajra
 
B
,
Szymonifka
 
J
,
Das
 
S
,
Leclercq
 
C
,
Martins
 
RP
, et al.  
Utility of computed tomography to predict ventricular arrhythmias in patients with nonischemic cardiomyopathy receiving cardiac resynchronization therapy
.
Am J Cardiol
 
2020
;
125
:
607
612
. https://doi.org/10.1016/j.amjcard.2019.11.003

162

Palmisano
 
A
,
Vignale
 
D
,
Peretto
 
G
,
Busnardo
 
E
,
Calcagno
 
C
,
Campochiaro
 
C
, et al.  
Hybrid FDG-PET/MR or FDG-PET/CT to detect disease activity in patients with persisting arrhythmias after myocarditis
.
JACC Cardiovasc Imaging
 
2021
;
14
:
288
292
. https://doi.org/10.1016/j.jcmg.2020.03.009

163

Wicks
 
EC
,
Menezes
 
LJ
,
Barnes
 
A
,
Mohiddin
 
SA
,
Sekhri
 
N
,
Porter
 
JC
, et al.  
Diagnostic accuracy and prognostic value of simultaneous hybrid 18F-fluorodeoxyglucose positron emission tomography/magnetic resonance imaging in cardiac sarcoidosis
.
Eur Heart J Cardiovasc Imaging
 
2018
;
19
:
757
767
. https://doi.org/10.1093/ehjci/jex340

164

Youssef
 
G
,
Leung
 
E
,
Mylonas
 
I
,
Nery
 
P
,
Williams
 
K
,
Wisenberg
 
G
, et al.  
The use of 18F-FDG PET in the diagnosis of cardiac sarcoidosis: a systematic review and metaanalysis including the Ontario experience
.
J Nucl Med
 
2012
;
53
:
241
248
. https://doi.org/10.2967/jnumed.111.090662

165

Bravo
 
PE
,
Di Carli
 
MF
,
Dorbala
 
S
.
Role of PET to evaluate coronary microvascular dysfunction in non-ischemic cardiomyopathies
.
Heart Fail Rev
 
2017
;
22
:
455
464
. https://doi.org/10.1007/s10741-017-9628-1

166

Perugini
 
E
,
Guidalotti
 
PL
,
Salvi
 
F
,
Cooke
 
RM
,
Pettinato
 
C
,
Riva
 
L
, et al.  
Noninvasive etiologic diagnosis of cardiac amyloidosis using 99mTc-3,3-diphosphono-1,2-propanodicarboxylic acid scintigraphy
.
J Am Coll Cardiol
 
2005
;
46
:
1076
1084
. https://doi.org/10.1016/j.jacc.2005.05.073

167

Hutt
 
DF
,
Fontana
 
M
,
Burniston
 
M
,
Quigley
 
AM
,
Petrie
 
A
,
Ross
 
JC
, et al.  
Prognostic utility of the Perugini grading of 99mTc-DPD scintigraphy in transthyretin (ATTR) amyloidosis and its relationship with skeletal muscle and soft tissue amyloid
.
Eur Heart J Cardiovasc Imaging
 
2017
;
18
:
1344
1350
. https://doi.org/10.1093/ehjci/jew325

168

Gillmore
 
JD
,
Maurer
 
MS
,
Falk
 
RH
,
Merlini
 
G
,
Damy
 
T
,
Dispenzieri
 
A
, et al.  
Nonbiopsy diagnosis of cardiac transthyretin amyloidosis
.
Circulation
 
2016
;
133
:
2404
2412
. https://doi.org/10.1161/CIRCULATIONAHA.116.021612

169

Langer
 
C
,
Lutz
 
M
,
Eden
 
M
,
Ludde
 
M
,
Hohnhorst
 
M
,
Gierloff
 
C
, et al.  
Hypertrophic cardiomyopathy in cardiac CT: a validation study on the detection of intramyocardial fibrosis in consecutive patients
.
Int J Cardiovasc Imaging
 
2014
;
30
:
659
667
. https://doi.org/10.1007/s10554-013-0358-8

170

Asferg
 
C
,
Usinger
 
L
,
Kristensen
 
TS
,
Abdulla
 
J
.
Accuracy of multi-slice computed tomography for measurement of left ventricular ejection fraction compared with cardiac magnetic resonance imaging and two-dimensional transthoracic echocardiography: a systematic review and meta-analysis
.
Eur J Radiol
 
2012
;
81
:
e757
e762
. https://doi.org/10.1016/j.ejrad.2012.02.002

171

Premaratne
 
M
,
Shamsaei
 
M
,
Chow
 
JD
,
Haddad
 
T
,
Erthal
 
F
,
Curran
 
H
, et al.  
Using coronary calcification to exclude an ischemic etiology for cardiomyopathy: a validation study and systematic review
.
Int J Cardiol
 
2017
;
230
:
518
522
. https://doi.org/10.1016/j.ijcard.2016.12.068

172

Dweck
 
MR
,
Abgral
 
R
,
Trivieri
 
MG
,
Robson
 
PM
,
Karakatsanis
 
N
,
Mani
 
V
, et al.  
Hybrid magnetic resonance imaging and positron emission tomography with fluorodeoxyglucose to diagnose active cardiac sarcoidosis
.
JACC Cardiovasc Imaging
 
2018
;
11
:
94
107
. https://doi.org/10.1016/j.jcmg.2017.02.021

173

Okumura
 
W
,
Iwasaki
 
T
,
Toyama
 
T
,
Iso
 
T
,
Arai
 
M
,
Oriuchi
 
N
, et al.  
Usefulness of fasting 18F-FDG PET in identification of cardiac sarcoidosis
.
J Nucl Med
 
2004
;
45
:
1989
1998
.

174

Besler
 
C
,
Urban
 
D
,
Watzka
 
S
,
Lang
 
D
,
Rommel
 
KP
,
Kandolf
 
R
, et al.  
Endomyocardial miR-133a levels correlate with myocardial inflammation, improved left ventricular function, and clinical outcome in patients with inflammatory cardiomyopathy
.
Eur J Heart Fail
 
2016
;
18
:
1442
1451
. https://doi.org/10.1002/ejhf.579

175

Ardehali
 
H
,
Qasim
 
A
,
Cappola
 
T
,
Howard
 
D
,
Hruban
 
R
,
Hare
 
JM
, et al.  
Endomyocardial biopsy plays a role in diagnosing patients with unexplained cardiomyopathy
.
Am Heart J
 
2004
;
147
:
919
923
. https://doi.org/10.1016/j.ahj.2003.09.020

176

Ardehali
 
H
,
Howard
 
DL
,
Hariri
 
A
,
Qasim
 
A
,
Hare
 
JM
,
Baughman
 
KL
, et al.  
A positive endomyocardial biopsy result for sarcoid is associated with poor prognosis in patients with initially unexplained cardiomyopathy
.
Am Heart J
 
2005
;
150
:
459
463
. https://doi.org/10.1016/j.ahj.2004.10.006

177

Hahn
 
VS
,
Yanek
 
LR
,
Vaishnav
 
J
,
Ying
 
W
,
Vaidya
 
D
,
Lee
 
YZJ
, et al.  
Endomyocardial biopsy characterization of heart failure with preserved ejection fraction and prevalence of cardiac amyloidosis
.
JACC Heart Fail
 
2020
;
8
:
712
724
. https://doi.org/10.1016/j.jchf.2020.04.007

178

Lorenzini
 
M
,
Norrish
 
G
,
Field
 
E
,
Ochoa
 
JP
,
Cicerchia
 
M
,
Akhtar
 
MM
, et al.  
Penetrance of hypertrophic cardiomyopathy in sarcomere protein mutation carriers
.
J Am Coll Cardiol
 
2020
;
76
:
550
559
. https://doi.org/10.1016/j.jacc.2020.06.011

179

Dalal
 
D
,
James
 
C
,
Devanagondi
 
R
,
Tichnell
 
C
,
Tucker
 
A
,
Prakasa
 
K
, et al.  
Penetrance of mutations in plakophilin-2 among families with arrhythmogenic right ventricular dysplasia/cardiomyopathy
.
J Am Coll Cardiol
 
2006
;
48
:
1416
1424
. https://doi.org/10.1016/j.jacc.2006.06.045

180

Claes
 
GR
,
van Tienen
 
FH
,
Lindsey
 
P
,
Krapels
 
IP
,
Helderman-van den Enden
 
AT
,
Hoos
 
MB
, et al.  
Hypertrophic remodelling in cardiac regulatory myosin light chain (MYL2) founder mutation carriers
.
Eur Heart J
 
2016
;
37
:
1815
1822
. https://doi.org/10.1093/eurheartj/ehv522

181

James
 
CA
,
Bhonsale
 
A
,
Tichnell
 
C
,
Murray
 
B
,
Russell
 
SD
,
Tandri
 
H
, et al.  
Exercise increases age-related penetrance and arrhythmic risk in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers
.
J Am Coll Cardiol
 
2013
;
62
:
1290
1297
. https://doi.org/10.1016/j.jacc.2013.06.033

182

Tadros
 
R
,
Francis
 
C
,
Xu
 
X
,
Vermeer
 
AMC
,
Harper
 
AR
,
Huurman
 
R
, et al.  
Shared genetic pathways contribute to risk of hypertrophic and dilated cardiomyopathies with opposite directions of effect
.
Nat Genet
 
2021
;
53
:
128
134
. https://doi.org/10.1038/s41588-020-00762-2

183

Harper
 
AR
,
Goel
 
A
,
Grace
 
C
,
Thomson
 
KL
,
Petersen
 
SE
,
Xu
 
X
, et al.  
Common genetic variants and modifiable risk factors underpin hypertrophic cardiomyopathy susceptibility and expressivity
.
Nat Genet
 
2021
;
53
:
135
142
. https://doi.org/10.1038/s41588-020-00764-0

184

Pugh
 
TJ
,
Kelly
 
MA
,
Gowrisankar
 
S
,
Hynes
 
E
,
Seidman
 
MA
,
Baxter
 
SM
, et al.  
The landscape of genetic variation in dilated cardiomyopathy as surveyed by clinical DNA sequencing
.
Genet Med
 
2014
;
16
:
601
608
. https://doi.org/10.1038/gim.2013.204

185

Escobar-Lopez
 
L
,
Ochoa
 
JP
,
Mirelis
 
JG
,
Espinosa
 
MA
,
Navarro
 
M
,
Gallego-Delgado
 
M
, et al.  
Association of genetic variants with outcomes in patients with nonischemic dilated cardiomyopathy
.
J Am Coll Cardiol
 
2021
;
78
:
1682
1699
. https://doi.org/10.1016/j.jacc.2021.08.039

186

Gigli
 
M
,
Merlo
 
M
,
Graw
 
SL
,
Barbati
 
G
,
Rowland
 
TJ
,
Slavov
 
DB
, et al.  
Genetic risk of arrhythmic phenotypes in patients with dilated cardiomyopathy
.
J Am Coll Cardiol
 
2019
;
74
:
1480
1490
. https://doi.org/10.1016/j.jacc.2019.06.072

187

Garnier
 
S
,
Harakalova
 
M
,
Weiss
 
S
,
Mokry
 
M
,
Regitz-Zagrosek
 
V
,
Hengstenberg
 
C
, et al.  
Genome-wide association analysis in dilated cardiomyopathy reveals two new players in systolic heart failure on chromosomes 3p25.1 and 22q11.23
.
Eur Heart J
 
2021
;
42
:
2000
2011
. https://doi.org/10.1093/eurheartj/ehab030

188

Pua
 
CJ
,
Tham
 
N
,
Chin
 
CWL
,
Walsh
 
R
,
Khor
 
CC
,
Toepfer
 
CN
, et al.  
Genetic studies of hypertrophic cardiomyopathy in Singaporeans identify variants in TNNI3 and TNNT2 that are common in Chinese patients
.
Circ Genom Precis Med
 
2020
;
13
:
424
434
. https://doi.org/10.1161/CIRCGEN.119.002823

189

Jordan
 
E
,
Peterson
 
L
,
Ai
 
T
,
Asatryan
 
B
,
Bronicki
 
L
,
Brown
 
E
, et al.  
Evidence-based assessment of genes in dilated cardiomyopathy
.
Circulation
 
2021
;
144
:
7
19
. https://doi.org/10.1161/CIRCULATIONAHA.120.053033

190

James
 
CA
,
Jongbloed
 
JDH
,
Hershberger
 
RE
,
Morales
 
A
,
Judge
 
DP
,
Syrris
 
P
, et al.  
International evidence based reappraisal of genes associated with arrhythmogenic right ventricular cardiomyopathy using the clinical genome resource framework
.
Circ Genom Precis Med
 
2021
;
14
:
e003273
. https://doi.org/10.1161/CIRCGEN.120.003273

191

Ingles
 
J
,
Goldstein
 
J
,
Thaxton
 
C
,
Caleshu
 
C
,
Corty
 
EW
,
Crowley
 
SB
, et al.  
Evaluating the clinical validity of hypertrophic cardiomyopathy genes
.
Circ Genom Precis Med
 
2019
;
12
:
e002460
. https://doi.org/10.1161/CIRCGEN.119.002460

191a

ClinGen Clinical Genome Resource
. https://search.clinicalgenome.org/kb/gene-validity  (12 July 2023 date last accessed).

192

Miller
 
DT
,
Lee
 
K
,
Chung
 
WK
,
Gordon
 
AS
,
Herman
 
GE
,
Klein
 
TE
, et al.  
ACMG SF v3.0 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics (ACMG)
.
Genet Med
 
2021
;
23
:
1381
1390
. https://doi.org/10.1038/s41436-021-01172-3

193

Miller
 
DT
,
Lee
 
K
,
Gordon
 
AS
,
Amendola
 
LM
,
Adelman
 
K
,
Bale
 
SJ
, et al.  
Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2021 update: a policy statement of the American College of Medical Genetics and Genomics (ACMG)
.
Genet Med
 
2021
;
23
:
1391
1398
. https://doi.org/10.1038/s41436-021-01171-4

194

Watkins
 
H
.
Time to think differently about sarcomere-negative hypertrophic cardiomyopathy
.
Circulation
 
2021
;
143
:
2415
2417
. https://doi.org/10.1161/CIRCULATIONAHA.121.053527

195

Kumuthini
 
J
,
Zick
 
B
,
Balasopoulou
 
A
,
Chalikiopoulou
 
C
,
Dandara
 
C
,
El-Kamah
 
G
, et al.  
The clinical utility of polygenic risk scores in genomic medicine practices: a systematic review
.
Hum Genet
 
2022
;
141
:
1697
1704
. https://doi.org/10.1007/s00439-022-02452-x

196

Pirruccello
 
JP
,
Bick
 
A
,
Wang
 
M
,
Chaffin
 
M
,
Friedman
 
S
,
Yao
 
J
, et al.  
Analysis of cardiac magnetic resonance imaging in 36,000 individuals yields genetic insights into dilated cardiomyopathy
.
Nat Commun
 
2020
;
11
:
2254
. https://doi.org/10.1038/s41467-020-15823-7

197

Biddinger
 
KJ
,
Jurgens
 
SJ
,
Maamari
 
D
,
Gaziano
 
L
,
Choi
 
SH
,
Morrill
 
VN
, et al.  
Rare and common genetic variation underlying the risk of hypertrophic cardiomyopathy in a National Biobank
.
JAMA Cardiol
 
2022
;
7
:
715
722
. https://doi.org/10.1001/jamacardio.2022.1061

198

Richards
 
S
,
Aziz
 
N
,
Bale
 
S
,
Bick
 
D
,
Das
 
S
,
Gastier-Foster
 
J
, et al.  
Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology
.
Genet Med
 
2015
;
17
:
405
424
. https://doi.org/10.1038/gim.2015.30

199

Kelly
 
MA
,
Caleshu
 
C
,
Morales
 
A
,
Buchan
 
J
,
Wolf
 
Z
,
Harrison
 
SM
, et al.  
Adaptation and validation of the ACMG/AMP variant classification framework for MYH7-associated inherited cardiomyopathies: recommendations by ClinGen’s Inherited Cardiomyopathy Expert Panel
.
Genet Med
 
2018
;
20
:
351
359
. https://doi.org/10.1038/gim.2017.218

200

Arbustini
 
E
,
Behr
 
ER
,
Carrier
 
L
,
van Duijn
 
C
,
Evans
 
P
,
Favalli
 
V
, et al.  
Interpretation and actionability of genetic variants in cardiomyopathies: a position statement from the European Society of Cardiology Council on cardiovascular genomics
.
Eur Heart J
 
2022
;
43
:
1901
1916
. https://doi.org/10.1093/eurheartj/ehab895

201

National Society of Genetic Counselors Definition Task Force
;
Resta
 
R
,
Biesecker
 
BB
,
Bennett
 
RL
,
Blum
 
S
,
Hahn
 
SE
.
A new definition of genetic counseling: National Society of Genetic Counselors’ Task Force report
.
J Genet Couns
 
2006
;
15
:
77
83
. https://doi.org/10.1007/s10897-005-9014-3

202

Biesecker
 
BB
.
Goals of genetic counseling
.
Clin Genet
 
2001
;
60
:
323
330
. https://doi.org/10.1034/j.1399-0004.2001.600501.x

203

Ingles
 
J
,
Yeates
 
L
,
Semsarian
 
C
.
The emerging role of the cardiac genetic counselor
.
Heart Rhythm
 
2011
;
8
:
1958
1962
. https://doi.org/10.1016/j.hrthm.2011.07.017

204

Bordet
 
C
,
Brice
 
S
,
Maupain
 
C
,
Gandjbakhch
 
E
,
Isidor
 
B
,
Palmyre
 
A
, et al.  
Psychosocial impact of predictive genetic testing in hereditary heart diseases: the PREDICT study
.
J Clin Med
 
2020
;
9
:
1365
. https://doi.org/10.3390/jcm9051365

205

Ingles
 
J
.
Psychological issues in managing families with inherited cardiovascular diseases
.
Cold Spring Harb Perspect Med
 
2020
;
10
:
a036558
. https://doi.org/10.1101/cshperspect.a036558

206

Edwards
 
A
,
Gray
 
J
,
Clarke
 
A
,
Dundon
 
J
,
Elwyn
 
G
,
Gaff
 
C
, et al.  
Interventions to improve risk communication in clinical genetics: systematic review
.
Patient Educ Couns
 
2008
;
71
:
4
25
. https://doi.org/10.1016/j.pec.2007.11.026

207

Austin
 
J
,
Semaka
 
A
,
Hadjipavlou
 
G
.
Conceptualizing genetic counseling as psychotherapy in the era of genomic medicine
.
J Genet Couns
 
2014
;
23
:
903
909
. https://doi.org/10.1007/s10897-014-9728-1

208

Michie
 
S
,
Marteau
 
TM
,
Bobrow
 
M
.
Genetic counselling: the psychological impact of meeting patients’ expectations
.
J Med Genet
 
1997
;
34
:
237
241
. https://doi.org/10.1136/jmg.34.3.237

209

Ison
 
HE
,
Ware
 
SM
,
Schwantes-An
 
TH
,
Freeze
 
S
,
Elmore
 
L
,
Spoonamore
 
KG
.
The impact of cardiovascular genetic counseling on patient empowerment
.
J Genet Couns
 
2019
;
28
:
570
577
. https://doi.org/10.1002/jgc4.1050

210

Borry
 
P
,
Evers-Kiebooms
 
G
,
Cornel
 
MC
,
Clarke
 
A
,
Dierickx
 
K
.
Public Professional Policy Committee (PPPC) of the European Society of Human Genetics (ESHG). Genetic testing in asymptomatic minors: background considerations towards ESHG recommendations
.
Eur J Hum Genet
 
2009
;
17
:
711
719
. https://doi.org/10.1038/ejhg.2009.25

211

Ormondroyd
 
E
,
Oates
 
S
,
Parker
 
M
,
Blair
 
E
,
Watkins
 
H
.
Pre-symptomatic genetic testing for inherited cardiac conditions: a qualitative exploration of psychosocial and ethical implications
.
Eur J Hum Genet
 
2014
;
22
:
88
93
. https://doi.org/10.1038/ejhg.2013.81

212

Spanaki
 
A
,
O’Curry
 
S
,
Winter-Beatty
 
J
,
Mead-Regan
 
S
,
Hawkins
 
K
,
English
 
J
, et al.  
Psychosocial adjustment and quality of life in children undergoing screening in a specialist paediatric hypertrophic cardiomyopathy clinic
.
Cardiol Young
 
2016
;
26
:
961
967
. https://doi.org/10.1017/S1047951115001717

213

Ingles
 
J
,
Semsarian
 
C
.
Conveying a probabilistic genetic test result to families with an inherited heart disease
.
Heart Rhythm
 
2014
;
11
:
1073
1078
. https://doi.org/10.1016/j.hrthm.2014.03.017

214

Whyte
 
S
,
Green
 
A
,
McAllister
 
M
,
Shipman
 
H
.
Family communication in inherited cardiovascular conditions in Ireland
.
J Genet Couns
 
2016
;
25
:
1317
1326
. https://doi.org/10.1007/s10897-016-9974-5

215

Daly
 
MB
,
Montgomery
 
S
,
Bingler
 
R
,
Ruth
 
K
.
Communicating genetic test results within the family: is it lost in translation? A survey of relatives in the randomized six-step study
.
Fam Cancer
 
2016
;
15
:
697
706
. https://doi.org/10.1007/s10689-016-9889-1

216

Burns
 
C
,
McGaughran
 
J
,
Davis
 
A
,
Semsarian
 
C
,
Ingles
 
J
.
Factors influencing uptake of familial long QT syndrome genetic testing
.
Am J Med Genet A
 
2016
;
170A
:
418
425
. https://doi.org/10.1002/ajmg.a.37455

217

Kaphingst
 
KA
,
Blanchard
 
M
,
Milam
 
L
,
Pokharel
 
M
,
Elrick
 
A
,
Goodman
 
MS
.
Relationships between health literacy and genomics-related knowledge, self-efficacy, perceived importance, and communication in a medically underserved population
.
J Health Commun
 
2016
;
21
:
58
68
. https://doi.org/10.1080/10810730.2016.1144661

218

Yeates
 
L
,
McDonald
 
K
,
Burns
 
C
,
Semsarian
 
C
,
Carter
 
S
,
Ingles
 
J
.
Decision-making and experiences of preimplantation genetic diagnosis in inherited heart diseases: a qualitative study
.
Eur J Hum Genet
 
2022
;
30
:
187
193
. https://doi.org/10.1038/s41431-021-00963-1

219

Landstrom
 
AP
,
Kim
 
JJ
,
Gelb
 
BD
,
Helm
 
BM
,
Kannankeril
 
PJ
,
Semsarian
 
C
, et al.  
Genetic testing for heritable cardiovascular diseases in pediatric patients: a scientific statement from the American Heart Association
.
Circ Genom Precis Med
 
2021
;
14
:
e000086
. https://doi.org/10.1161/HCG.0000000000000086

220

Akolekar
 
R
,
Beta
 
J
,
Picciarelli
 
G
,
Ogilvie
 
C
,
D’Antonio
 
F
.
Procedure-related risk of miscarriage following amniocentesis and chorionic villus sampling: a systematic review and meta-analysis
.
Ultrasound Obstet Gynecol
 
2015
;
45
:
16
26
. https://doi.org/10.1002/uog.14636

221

Meiser
 
B
,
Irle
 
J
,
Lobb
 
E
,
Barlow-Stewart
 
K
.
Assessment of the content and process of genetic counseling: a critical review of empirical studies
.
J Genet Couns
 
2008
;
17
:
434
451
. https://doi.org/10.1007/s10897-008-9173-0

222

Waddell-Smith
 
KE
,
Donoghue
 
T
,
Oates
 
S
,
Graham
 
A
,
Crawford
 
J
,
Stiles
 
MK
, et al.  
Inpatient detection of cardiac-inherited disease: the impact of improving family history taking
.
Open Heart
 
2016
;
3
:
e000329
. https://doi.org/10.1136/openhrt-2015-000329

223

Murray
 
B
,
Tichnell
 
C
,
Burch
 
AE
,
Calkins
 
H
,
James
 
CA
.
Strength of the genetic counselor: patient relationship is associated with extent of increased empowerment in patients with arrhythmogenic cardiomyopathy
.
J Genet Couns
 
2022
;
31
:
388
397
. https://doi.org/10.1002/jgc4.1499

224

Ingles
 
J
,
Lind
 
JM
,
Phongsavan
 
P
,
Semsarian
 
C
.
Psychosocial impact of specialized cardiac genetic clinics for hypertrophic cardiomyopathy
.
Genet Med
 
2008
;
10
:
117
120
. https://doi.org/10.1097/GIM.0b013e3181612cc7

225

Furqan
 
A
,
Arscott
 
P
,
Girolami
 
F
,
Cirino
 
AL
,
Michels
 
M
,
Day
 
SM
, et al.  
Care in specialized centers and data sharing increase agreement in hypertrophic cardiomyopathy genetic test interpretation
.
Circ Cardiovasc Genet
 
2017
;
10
:
e001700
. https://doi.org/10.1161/CIRCGENETICS.116.001700

226

Reuter
 
C
,
Grove
 
ME
,
Orland
 
K
,
Spoonamore
 
K
,
Caleshu
 
C
.
Clinical cardiovascular genetic counselors take a leading role in team-based variant classification
.
J Genet Couns
 
2018
;
27
:
751
760
. https://doi.org/10.1007/s10897-017-0175-7

227

Ingles
 
J
,
McGaughran
 
J
,
Scuffham
 
PA
,
Atherton
 
J
,
Semsarian
 
C
.
A cost-effectiveness model of genetic testing for the evaluation of families with hypertrophic cardiomyopathy
.
Heart
 
2012
;
98
:
625
630
. https://doi.org/10.1136/heartjnl-2011-300368

228

Wordsworth
 
S
,
Leal
 
J
,
Blair
 
E
,
Legood
 
R
,
Thomson
 
K
,
Seller
 
A
, et al.  
DNA testing for hypertrophic cardiomyopathy: a cost-effectiveness model
.
Eur Heart J
 
2010
;
31
:
926
935
. https://doi.org/10.1093/eurheartj/ehq067

229

Catchpool
 
M
,
Ramchand
 
J
,
Martyn
 
M
,
Hare
 
DL
,
James
 
PA
,
Trainer
 
AH
, et al.  
A cost-effectiveness model of genetic testing and periodical clinical screening for the evaluation of families with dilated cardiomyopathy
.
Genet Med
 
2019
;
21
:
2815
2822
. https://doi.org/10.1038/s41436-019-0582-2

230

Groeneweg
 
JA
,
Bhonsale
 
A
,
James
 
CA
,
te Riele
 
AS
,
Dooijes
 
D
,
Tichnell
 
C
, et al.  
Clinical presentation, long-term follow-up, and outcomes of 1001 arrhythmogenic right ventricular dysplasia/cardiomyopathy patients and family members
.
Circ Cardiovasc Genet
 
2015
;
8
:
437
446
. https://doi.org/10.1161/CIRCGENETICS.114.001003

231

Alfares
 
AA
,
Kelly
 
MA
,
McDermott
 
G
,
Funke
 
BH
,
Lebo
 
MS
,
Baxter
 
SB
, et al.  
Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: expanded panels offer limited additional sensitivity
.
Genet Med
 
2015
;
17
:
880
888
. https://doi.org/10.1038/gim.2014.205

232

Ingles
 
J
,
Yeates
 
L
,
O’Brien
 
L
,
McGaughran
 
J
,
Scuffham
 
PA
,
Atherton
 
J
, et al.  
Genetic testing for inherited heart diseases: longitudinal impact on health-related quality of life
.
Genet Med
 
2012
;
14
:
749
752
. https://doi.org/10.1038/gim.2012.47

233

Friess
 
MR
,
Marino
 
BS
,
Cassedy
 
A
,
Wilmot
 
I
,
Jefferies
 
JL
,
Lorts
 
A
.
Health-related quality of life assessment in children followed in a cardiomyopathy clinic
.
Pediatr Cardiol
 
2015
;
36
:
516
523
. https://doi.org/10.1007/s00246-014-1042-z

234

Wakefield
 
CE
,
Hanlon
 
LV
,
Tucker
 
KM
,
Patenaude
 
AF
,
Signorelli
 
C
,
McLoone
 
JK
, et al.  
The psychological impact of genetic information on children: a systematic review
.
Genet Med
 
2016
;
18
:
755
762
. https://doi.org/10.1038/gim.2015.181

235

Christian
 
S
,
Somerville
 
M
,
Taylor
 
S
,
Atallah
 
J
.
When to offer predictive genetic testing to children at risk of an inherited arrhythmia or cardiomyopathy
.
Circ Genom Precis Med
 
2018
;
11
:
e002300
. https://doi.org/10.1161/CIRCGEN.118.002300

236

MacLeod
 
R
,
Beach
 
A
,
Henriques
 
S
,
Knopp
 
J
,
Nelson
 
K
,
Kerzin-Storrar
 
L
.
Experiences of predictive testing in young people at risk of Huntington’s disease, familial cardiomyopathy or hereditary breast and ovarian cancer
.
Eur J Hum Genet
 
2014
;
22
:
396
401
. https://doi.org/10.1038/ejhg.2013.143

237

Knight
 
LM
,
Miller
 
E
,
Kovach
 
J
,
Arscott
 
P
,
von Alvensleben
 
JC
,
Bradley
 
D
, et al.  
Genetic testing and cascade screening in pediatric long QT syndrome and hypertrophic cardiomyopathy
.
Heart Rhythm
 
2020
;
17
:
106
112
. https://doi.org/10.1016/j.hrthm.2019.06.015

238

Ho
 
CY
,
Day
 
SM
,
Ashley
 
EA
,
Michels
 
M
,
Pereira
 
AC
,
Jacoby
 
D
, et al.  
Genotype and lifetime burden of disease in hypertrophic cardiomyopathy: insights from the Sarcomeric Human Cardiomyopathy Registry (SHaRe)
.
Circulation
 
2018
;
138
:
1387
1398
. https://doi.org/10.1161/CIRCULATIONAHA.117.033200

239

Marey
 
I
,
Fressart
 
V
,
Rambaud
 
C
,
Fornes
 
P
,
Martin
 
L
,
Grotto
 
S
, et al.  
Clinical impact of post-mortem genetic testing in cardiac death and cardiomyopathy
.
Open Med (Wars)
 
2020
;
15
:
435
446
. https://doi.org/10.1515/med-2020-0150

240

Isbister
 
JC
,
Nowak
 
N
,
Butters
 
A
,
Yeates
 
L
,
Gray
 
B
,
Sy
 
RW
, et al.  
“Concealed cardiomyopathy” as a cause of previously unexplained sudden cardiac arrest
.
Int J Cardiol
 
2021
;
324
:
96
101
. https://doi.org/10.1016/j.ijcard.2020.09.031

241

Dellefave-Castillo
 
LM
,
Cirino
 
AL
,
Callis
 
TE
,
Esplin
 
ED
,
Garcia
 
J
,
Hatchell
 
KE
, et al.  
Assessment of the diagnostic yield of combined cardiomyopathy and arrhythmia genetic testing
.
JAMA Cardiol
 
2022
;
7
:
966
974
. https://doi.org/10.1001/jamacardio.2022.2455

242

Williams
 
N
,
Manderski
 
E
,
Stewart
 
S
,
Bao
 
R
,
Tang
 
Y
.
Lessons learned from testing cardiac channelopathy and cardiomyopathy genes in individuals who died suddenly: a two-year prospective study in a large medical examiner’s office with an in-house molecular genetics laboratory and genetic counseling services
.
J Genet Couns
 
2020
;
29
:
293
302
. https://doi.org/10.1002/jgc4.1157

243

Isbister
 
JC
,
Nowak
 
N
,
Yeates
 
L
,
Singer
 
ES
,
Sy
 
RW
,
Ingles
 
J
, et al.  
Concealed cardiomyopathy in autopsy-inconclusive cases of sudden cardiac death and implications for families
.
J Am Coll Cardiol
 
2022
;
80
:
2057
2068
. https://doi.org/10.1016/j.jacc.2022.09.029

244

Michie
 
S
,
Bobrow
 
M
,
Marteau
 
TM
.
Predictive genetic testing in children and adults: a study of emotional impact
.
J Med Genet
 
2001
;
38
:
519
526
. https://doi.org/10.1136/jmg.38.8.519

245

Rath
 
A
,
Weintraub
 
R
.
Overview of cardiomyopathies in childhood
.
Front Pediatr
 
2021
;
9
:
708732
. https://doi.org/10.3389/fped.2021.708732

246

Lipshultz
 
SE
,
Law
 
YM
,
Asante-Korang
 
A
,
Austin
 
ED
,
Dipchand
 
AI
,
Everitt
 
MD
, et al.  
Cardiomyopathy in children: classification and diagnosis: a scientific statement from the American Heart Association
.
Circulation
 
2019
;
140
:
e9
e68
. https://doi.org/10.1161/CIR.0000000000000682

247

Kindel
 
SJ
,
Miller
 
EM
,
Gupta
 
R
,
Cripe
 
LH
,
Hinton
 
RB
,
Spicer
 
RL
, et al.  
Pediatric cardiomyopathy: importance of genetic and metabolic evaluation
.
J Card Fail
 
2012
;
18
:
396
403
. https://doi.org/10.1016/j.cardfail.2012.01.017

248

Norrish
 
G
,
Field
 
E
,
McLeod
 
K
,
Ilina
 
M
,
Stuart
 
G
,
Bhole
 
V
, et al.  
Clinical presentation and survival of childhood hypertrophic cardiomyopathy: a retrospective study in United Kingdom
.
Eur Heart J
 
2019
;
40
:
986
993
. https://doi.org/10.1093/eurheartj/ehy798

249

Towbin
 
JA
,
Lowe
 
AM
,
Colan
 
SD
,
Sleeper
 
LA
,
Orav
 
EJ
,
Clunie
 
S
, et al.  
Incidence, causes, and outcomes of dilated cardiomyopathy in children
.
JAMA
 
2006
;
296
:
1867
1876
. https://doi.org/10.1001/jama.296.15.1867

250

Shamszad
 
P
,
Hall
 
M
,
Rossano
 
JW
,
Denfield
 
SW
,
Knudson
 
JD
,
Penny
 
DJ
, et al.  
Characteristics and outcomes of heart failure-related intensive care unit admissions in children with cardiomyopathy
.
J Card Fail
 
2013
;
19
:
672
677
. https://doi.org/10.1016/j.cardfail.2013.08.006

251

Pelliccia
 
F
,
Alfieri
 
O
,
Calabro
 
P
,
Cecchi
 
F
,
Ferrazzi
 
P
,
Gragnano
 
F
, et al.  
Multidisciplinary evaluation and management of obstructive hypertrophic cardiomyopathy in 2020: towards the HCM Heart Team
.
Int J Cardiol
 
2020
;
304
:
86
92
. https://doi.org/10.1016/j.ijcard.2020.01.021

252

Law
 
SP
,
Oron
 
AP
,
Kemna
 
MS
,
Albers
 
EL
,
McMullan
 
DM
,
Chen
 
JM
, et al.  
Comparison of transplant waitlist outcomes for pediatric candidates supported by ventricular assist devices versus medical therapy
.
Pediatr Crit Care Med
 
2018
;
19
:
442
450
. https://doi.org/10.1097/PCC.0000000000001503

253

Ullmo
 
S
,
Vial
 
Y
,
Di Bernardo
 
S
,
Roth-Kleiner
 
M
,
Mivelaz
 
Y
,
Sekarski
 
N
, et al.  
Pathologic ventricular hypertrophy in the offspring of diabetic mothers: a retrospective study
.
Eur Heart J
 
2007
;
28
:
1319
1325
. https://doi.org/10.1093/eurheartj/ehl416

254

Yunis
 
KA
,
Bitar
 
FF
,
Hayek
 
P
,
Mroueh
 
SM
,
Mikati
 
M
.
Transient hypertrophic cardiomyopathy in the newborn following multiple doses of antenatal corticosteroids
.
Am J Perinatol
 
1999
;
16
:
17
21
. https://doi.org/10.1055/s-2007-993830

255

Brickman
 
WJ
,
Silverman
 
BL
.
Cardiovascular effects of growth hormone
.
Endocrine
 
2000
;
12
:
153
161
. https://doi.org/10.1385/ENDO:12:2:153

256

Monda
 
E
,
Rubino
 
M
,
Lioncino
 
M
,
Di Fraia
 
F
,
Pacileo
 
R
,
Verrillo
 
F
, et al.  
Hypertrophic cardiomyopathy in children: pathophysiology, diagnosis, and treatment of non-sarcomeric causes
.
Front Pediatr
 
2021
;
9
:
632293
. https://doi.org/10.3389/fped.2021.632293

257

Fourey
 
D
,
Care
 
M
,
Siminovitch
 
KA
,
Weissler-Snir
 
A
,
Hindieh
 
W
,
Chan
 
RH
, et al.  
Prevalence and clinical implication of double mutations in hypertrophic cardiomyopathy: revisiting the gene-dose effect
.
Circ Cardiovasc Genet
 
2017
;
10
:
e001685
. https://doi.org/10.1161/CIRCGENETICS.116.001685

258

Kaltenecker
 
E
,
Schleihauf
 
J
,
Meierhofer
 
C
,
Shehu
 
N
,
Mkrtchyan
 
N
,
Hager
 
A
, et al.  
Long-term outcomes of childhood onset Noonan compared to sarcomere hypertrophic cardiomyopathy
.
Cardiovasc Diagn Ther
 
2019
;
9
:
S299
S309
. https://doi.org/10.21037/cdt.2019.05.01

259

Kishnani
 
PS
,
Hwu
 
WL
,
Mandel
 
H
,
Nicolino
 
M
,
Yong
 
F
,
Corzo
 
D
, et al.  
A retrospective, multinational, multicenter study on the natural history of infantile-onset Pompe disease
.
J Pediatr
 
2006
;
148
:
671
676.e2
. https://doi.org/10.1016/j.jpeds.2005.11.033

260

Linglart
 
L
,
Gelb
 
BD
.
Congenital heart defects in Noonan syndrome: diagnosis, management, and treatment
.
Am J Med Genet C Semin Med Genet
 
2020
;
184
:
73
80
. https://doi.org/10.1002/ajmg.c.31765

261

Gelb
 
BD
,
Roberts
 
AE
,
Tartaglia
 
M
.
Cardiomyopathies in Noonan syndrome and the other RASopathies
.
Prog Pediatr Cardiol
 
2015
;
39
:
13
19
. https://doi.org/10.1016/j.ppedcard.2015.01.002

262

Calcagni
 
G
,
Limongelli
 
G
,
D’Ambrosio
 
A
,
Gesualdo
 
F
,
Digilio
 
MC
,
Baban
 
A
, et al.  
Cardiac defects, morbidity and mortality in patients affected by RASopathies. CARNET study results
.
Int J Cardiol
 
2017
;
245
:
92
98
. https://doi.org/10.1016/j.ijcard.2017.07.068

263

Lioncino
 
M
,
Monda
 
E
,
Verrillo
 
F
,
Moscarella
 
E
,
Calcagni
 
G
,
Drago
 
F
, et al.  
Hypertrophic cardiomyopathy in RASopathies: diagnosis, clinical characteristics, prognostic implications, and management
.
Heart Fail Clin
 
2022
;
18
:
19
29
. https://doi.org/10.1016/j.hfc.2021.07.004

264

Calcagni
 
G
,
Adorisio
 
R
,
Martinelli
 
S
,
Grutter
 
G
,
Baban
 
A
,
Versacci
 
P
, et al.  
Clinical presentation and natural history of hypertrophic cardiomyopathy in RASopathies
.
Heart Fail Clin
 
2018
;
14
:
225
235
. https://doi.org/10.1016/j.hfc.2017.12.005

265

Poterucha
 
JT
,
Johnson
 
JN
,
O’Leary
 
PW
,
Connolly
 
HM
,
Niaz
 
T
,
Maleszewski
 
JJ
, et al.  
Surgical ventricular septal myectomy for patients with Noonan syndrome and symptomatic left ventricular outflow tract obstruction
.
Am J Cardiol
 
2015
;
116
:
1116
1121
. https://doi.org/10.1016/j.amjcard.2015.06.037

266

Hemmati
 
P
,
Dearani
 
JA
,
Daly
 
RC
,
King
 
KS
,
Ammash
 
NM
,
Cetta
 
F
, et al.  
Early outcomes of cardiac surgery in patients with Noonan syndrome
.
Semin Thorac Cardiovasc Surg
 
2019
;
31
:
507
513
. https://doi.org/10.1053/j.semtcvs.2018.12.004

267

Moran
 
AM
,
Colan
 
SD
.
Verapamil therapy in infants with hypertrophic cardiomyopathy
.
Cardiol Young
 
1998
;
8
:
310
319
. https://doi.org/10.1017/S1047951100006818

268

van der Ploeg
 
AT
,
Reuser
 
AJ
.
Pompe’s disease
.
Lancet
 
2008
;
372
:
1342
1353
. https://doi.org/10.1016/S0140-6736(08)61555-X

269

Kishnani
 
PS
,
Corzo
 
D
,
Nicolino
 
M
,
Byrne
 
B
,
Mandel
 
H
,
Hwu
 
WL
, et al.  
Recombinant human acid [alpha]-glucosidase: major clinical benefits in infantile-onset Pompe disease
.
Neurology
 
2007
;
68
:
99
109
. https://doi.org/10.1212/01.wnl.0000251268.41188.04

270

Tanaka
 
M
,
Ino
 
H
,
Ohno
 
K
,
Hattori
 
K
,
Sato
 
W
,
Ozawa
 
T
, et al.  
Mitochondrial mutation in fatal infantile cardiomyopathy
.
Lancet
 
1990
;
336
:
1452
. https://doi.org/10.1016/0140-6736(90)93162-I

271

Holmgren
 
D
,
Wahlander
 
H
,
Eriksson
 
BO
,
Oldfors
 
A
,
Holme
 
E
,
Tulinius
 
M
.
Cardiomyopathy in children with mitochondrial disease; clinical course and cardiological findings
.
Eur Heart J
 
2003
;
24
:
280
288
. https://doi.org/10.1016/S0195-668X(02)00387-1

272

Arad
 
M
,
Maron
 
BJ
,
Gorham
 
JM
,
Johnson
 
WH
 Jr
,
Saul
 
JP
,
Perez-Atayde
 
AR
, et al.  
Glycogen storage diseases presenting as hypertrophic cardiomyopathy
.
N Engl J Med
 
2005
;
352
:
362
372
. https://doi.org/10.1056/NEJMoa033349

273

Ansong
 
AK
,
Li
 
JS
,
Nozik-Grayck
 
E
,
Ing
 
R
,
Kravitz
 
RM
,
Idriss
 
SF
, et al.  
Electrocardiographic response to enzyme replacement therapy for Pompe disease
.
Genet Med
 
2006
;
8
:
297
301
. https://doi.org/10.1097/01.gim.0000195896.04069.5f

274

Schoser
 
B
,
Attarian
 
S
,
Borges
 
J
,
Bouhour
 
F
,
Chien
 
Y
,
Choi
 
Y
, et al.  
Efficacy and safety results of the avalglucosidase alfa phase 3 COMET trial in late-onset Pompe disease patients
.
Eur J Neurol
 
2021
;
28
:
68
68
.

275

van der Ploeg
 
AT
,
Clemens
 
PR
,
Corzo
 
D
,
Escolar
 
DM
,
Florence
 
J
,
Groeneveld
 
GJ
, et al.  
A randomized study of alglucosidase alfa in late-onset Pompe’s disease
.
N Engl J Med
 
2010
;
362
:
1396
1406
. https://doi.org/10.1056/NEJMoa0909859

276

Lee
 
TM
,
Hsu
 
DT
,
Kantor
 
P
,
Towbin
 
JA
,
Ware
 
SM
,
Colan
 
SD
, et al.  
Pediatric cardiomyopathies
.
Circ Res
 
2017
;
121
:
855
873
. https://doi.org/10.1161/CIRCRESAHA.116.309386

277

Chang
 
RR
,
Allada
 
V
.
Electrocardiographic and echocardiographic features that distinguish anomalous origin of the left coronary artery from pulmonary artery from idiopathic dilated cardiomyopathy
.
Pediatr Cardiol
 
2001
;
22
:
3
10
. https://doi.org/10.1007/s002460010142

278

Cooper
 
LT
,
Baughman
 
KL
,
Feldman
 
AM
,
Frustaci
 
A
,
Jessup
 
M
,
Kuhl
 
U
, et al.  
The role of endomyocardial biopsy in the management of cardiovascular disease: a scientific statement from the American Heart Association, the American College of Cardiology, and the European Society of Cardiology. Endorsed by the Heart Failure Society of America and the Heart Failure Association of the European Society of Cardiology
.
J Am Coll Cardiol
 
2007
;
50
:
1914
1931
. https://doi.org/10.1093/eurheartj/ehm456

279

Law
 
YM
,
Lal
 
AK
,
Chen
 
S
,
Cihakova
 
D
,
Cooper
 
LT
 Jr
,
Deshpande
 
S
, et al.  
Diagnosis and management of myocarditis in children: a scientific statement from the American Heart Association
.
Circulation
 
2021
;
144
:
e123
e135
. https://doi.org/10.1161/CIR.0000000000001001

280

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
1724
. https://doi.org/10.1056/NEJM199912023412302

281

Towbin
 
JA
,
Hejtmancik
 
JF
,
Brink
 
P
,
Gelb
 
B
,
Zhu
 
XM
,
Chamberlain
 
JS
, et al.  
X-linked dilated cardiomyopathy. Molecular genetic evidence of linkage to the Duchenne muscular dystrophy (dystrophin) gene at the Xp21 locus
.
Circulation
 
1993
;
87
:
1854
1865
. https://doi.org/10.1161/01.CIR.87.6.1854

282

D’Amario
 
D
,
Amodeo
 
A
,
Adorisio
 
R
,
Tiziano
 
FD
,
Leone
 
AM
,
Perri
 
G
, et al.  
A current approach to heart failure in Duchenne muscular dystrophy
.
Heart
 
2017
;
103
:
1770
1779
. https://doi.org/10.1136/heartjnl-2017-311269

283

Towbin
 
JA
.
Left ventricular noncompaction: a new form of heart failure
.
Heart Fail Clin
 
2010
;
6
:
453
469
. https://doi.org/10.1016/j.hfc.2010.06.005

284

Towbin
 
JA
,
Lorts
 
A
,
Jefferies
 
JL
.
Left ventricular non-compaction cardiomyopathy
.
Lancet
 
2015
;
386
:
813
825
. https://doi.org/10.1016/S0140-6736(14)61282-4

285

Pignatelli
 
RH
,
McMahon
 
CJ
,
Dreyer
 
WJ
,
Denfield
 
SW
,
Price
 
J
,
Belmont
 
JW
, et al.  
Clinical characterization of left ventricular noncompaction in children: a relatively common form of cardiomyopathy
.
Circulation
 
2003
;
108
:
2672
2678
. https://doi.org/10.1161/01.CIR.0000100664.10777.B8

286

Webber
 
SA
,
Lipshultz
 
SE
,
Sleeper
 
LA
,
Lu
 
M
,
Wilkinson
 
JD
,
Addonizio
 
LJ
, et al.  
Outcomes of restrictive cardiomyopathy in childhood and the influence of phenotype: a report from the Pediatric Cardiomyopathy Registry
.
Circulation
 
2012
;
126
:
1237
1244
. https://doi.org/10.1161/CIRCULATIONAHA.112.104638

287

Pinto
 
JR
,
Parvatiyar
 
MS
,
Jones
 
MA
,
Liang
 
J
,
Potter
 
JD
.
A troponin T mutation that causes infantile restrictive cardiomyopathy increases Ca2+ sensitivity of force development and impairs the inhibitory properties of troponin
.
J Biol Chem
 
2008
;
283
:
2156
2166
. https://doi.org/10.1074/jbc.M707066200

288

Peled
 
Y
,
Gramlich
 
M
,
Yoskovitz
 
G
,
Feinberg
 
MS
,
Afek
 
A
,
Polak-Charcon
 
S
, et al.  
Titin mutation in familial restrictive cardiomyopathy
.
Int J Cardiol
 
2014
;
171
:
24
30
. https://doi.org/10.1016/j.ijcard.2013.11.037

289

Mogensen
 
J
,
Kubo
 
T
,
Duque
 
M
,
Uribe
 
W
,
Shaw
 
A
,
Murphy
 
R
, et al.  
Idiopathic restrictive cardiomyopathy is part of the clinical expression of cardiac troponin I mutations
.
J Clin Invest
 
2003
;
111
:
209
216
. https://doi.org/10.1172/JCI200316336

290

Protonotarios
 
N
,
Tsatsopoulou
 
A
.
Naxos disease and Carvajal syndrome: cardiocutaneous disorders that highlight the pathogenesis and broaden the spectrum of arrhythmogenic right ventricular cardiomyopathy
.
Cardiovasc Pathol
 
2004
;
13
:
185
194
. https://doi.org/10.1016/j.carpath.2004.03.609

291

McKoy
 
G
,
Protonotarios
 
N
,
Crosby
 
A
,
Tsatsopoulou
 
A
,
Anastasakis
 
A
,
Coonar
 
A
, et al.  
Identification of a deletion in plakoglobin in arrhythmogenic right ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease)
.
Lancet
 
2000
;
355
:
2119
2124
. https://doi.org/10.1016/S0140-6736(00)02379-5

292

Carvajal-Huerta
 
L
.
Epidermolytic palmoplantar keratoderma with woolly hair and dilated cardiomyopathy
.
J Am Acad Dermatol
 
1998
;
39
:
418
421
. https://doi.org/10.1016/S0190-9622(98)70317-2

293

Smedsrud
 
MK
,
Chivulescu
 
M
,
Forsa
 
MI
,
Castrini
 
I
,
Aabel
 
EW
,
Rootwelt-Norberg
 
C
, et al.  
Highly malignant disease in childhood-onset arrhythmogenic right ventricular cardiomyopathy
.
Eur Heart J
 
2022
;
43
:
4694
4703
. https://doi.org/10.1093/eurheartj/ehac485

294

Kontorovich
 
AR
,
Patel
 
N
,
Moscati
 
A
,
Richter
 
F
,
Peter
 
I
,
Purevjav
 
E
, et al.  
Myopathic cardiac genotypes increase risk for myocarditis
.
JACC Basic Transl Sci
 
2021
;
6
:
584
592
. https://doi.org/10.1016/j.jacbts.2021.06.001

295

Poller
 
W
,
Haas
 
J
,
Klingel
 
K
,
Kuhnisch
 
J
,
Gast
 
M
,
Kaya
 
Z
, et al.  
Familial recurrent myocarditis triggered by exercise in patients with a truncating variant of the desmoplakin gene
.
J Am Heart Assoc
 
2020
;
9
:
e015289
. https://doi.org/10.1161/JAHA.119.015289

296

Martins
 
D
,
Ovaert
 
C
,
Khraiche
 
D
,
Boddaert
 
N
,
Bonnet
 
D
,
Raimondi
 
F
.
Myocardial inflammation detected by cardiac MRI in arrhythmogenic right ventricular cardiomyopathy: a paediatric case series
.
Int J Cardiol
 
2018
;
271
:
81
86
. https://doi.org/10.1016/j.ijcard.2018.05.116

297

Bariani
 
R
,
Cipriani
 
A
,
Rizzo
 
S
,
Celeghin
 
R
,
Bueno Marinas
 
M
,
Giorgi
 
B
, et al.  
‘Hot phase’ clinical presentation in arrhythmogenic cardiomyopathy
.
Europace
 
2021
;
23
:
907
917
. https://doi.org/10.1093/europace/euaa343

298

Ross
 
RD
.
The Ross classification for heart failure in children after 25 years: a review and an age-stratified revision
.
Pediatr Cardiol
 
2012
;
33
:
1295
1300
. https://doi.org/10.1007/s00246-012-0306-8

299

Zeppenfeld
 
K
,
Tfelt-Hansen
 
J
,
de Riva
 
M
,
Winkel
 
BG
,
Behr
 
ER
,
Blom
 
NA
, et al.  
2022 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death
.
Eur Heart J
 
2022
;
43
:
3997
4126
. https://doi.org/10.1093/eurheartj/ehac262

300

Knuuti
 
J
,
Wijns
 
W
,
Saraste
 
A
,
Capodanno
 
D
,
Barbato
 
E
,
Funck-Brentano
 
C
, et al.  
2019 ESC Guidelines for the diagnosis and management of chronic coronary syndromes
.
Eur Heart J
 
2020
;
41
:
407
477
. https://doi.org/10.1093/eurheartj/ehz425

301

Brignole
 
M
,
Moya
 
A
,
de Lange
 
FJ
,
Deharo
 
JC
,
Elliott
 
PM
,
Fanciulli
 
A
, et al.  
2018 ESC Guidelines for the diagnosis and management of syncope
.
Eur Heart J
 
2018
;
39
:
1883
1948
. https://doi.org/10.1093/eurheartj/ehy037

302

Ahmad
 
F
,
Seidman
 
JG
,
Seidman
 
CE
.
The genetic basis for cardiac remodeling
.
Annu Rev Genomics Hum Genet
 
2005
;
6
:
185
216
. https://doi.org/10.1146/annurev.genom.6.080604.162132

303

Ferro
 
MD
,
Stolfo
 
D
,
Altinier
 
A
,
Gigli
 
M
,
Perrieri
 
M
,
Ramani
 
F
, et al.  
Association between mutation status and left ventricular reverse remodelling in dilated cardiomyopathy
.
Heart
 
2017
;
103
:
1704
1710
. https://doi.org/10.1136/heartjnl-2016-311017

304

de Boer
 
RA
,
Heymans
 
S
,
Backs
 
J
,
Carrier
 
L
,
Coats
 
AJS
,
Dimmeler
 
S
, et al.  
Targeted therapies in genetic dilated and hypertrophic cardiomyopathies: from molecular mechanisms to therapeutic targets. A position paper from the Heart Failure Association (HFA) and the Working Group on Myocardial Function of the European Society of Cardiology (ESC)
.
Eur J Heart Fail
 
2022
;
24
:
406
420
.

305

Oghina
 
S
,
Bougouin
 
W
,
Bezard
 
M
,
Kharoubi
 
M
,
Komajda
 
M
,
Cohen-Solal
 
A
, et al.  
The impact of patients with cardiac amyloidosis in HFpEF trials
.
JACC Heart Fail
 
2021
;
9
:
169
178
. https://doi.org/10.1016/j.jchf.2020.12.005

306

Bozkurt
 
B
,
Coats
 
AJS
,
Tsutsui
 
H
,
Abdelhamid
 
M
,
Adamopoulos
 
S
,
Albert
 
N
, et al.  
Universal definition and classification of heart failure: a report of the Heart Failure Society of America, Heart Failure Association of the European Society of Cardiology, Japanese Heart Failure Society and Writing Committee of the Universal Definition of Heart Failure
.
J Card Fail
 
2021
;
3
:
S1071-9164(21)00050-6
. https://doi.org/10.1016/j.cardfail.2021.01.022

307

Halliday
 
BP
,
Wassall
 
R
,
Lota
 
AS
,
Khalique
 
Z
,
Gregson
 
J
,
Newsome
 
S
, et al.  
Withdrawal of pharmacological treatment for heart failure in patients with recovered dilated cardiomyopathy (TRED-HF): an open-label, pilot, randomised trial
.
Lancet
 
2019
;
393
:
61
73
. https://doi.org/10.1016/S0140-6736(18)32484-X

308

Mureddu
 
GF
,
Tarantini
 
L
,
Agabiti
 
N
,
Faggiano
 
P
,
Masson
 
S
,
Latini
 
R
, et al.  
Evaluation of different strategies for identifying asymptomatic left ventricular dysfunction and pre-clinical (stage B) heart failure in the elderly. Results from ‘PREDICTOR’, a population based-study in central Italy
.
Eur J Heart Fail
 
2013
;
15
:
1102
1112
. https://doi.org/10.1093/eurjhf/hft098

309

Green
 
EM
,
Wakimoto
 
H
,
Anderson
 
RL
,
Evanchik
 
MJ
,
Gorham
 
JM
,
Harrison
 
BC
, et al.  
A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice
.
Science
 
2016
;
351
:
617
621
. https://doi.org/10.1126/science.aad3456

310

Masarone
 
D
,
Valente
 
F
,
Rubino
 
M
,
Vastarella
 
R
,
Gravino
 
R
,
Rea
 
A
, et al.  
Pediatric heart failure: a practical guide to diagnosis and management
.
Pediatr Neonatol
 
2017
;
58
:
303
312
. https://doi.org/10.1016/j.pedneo.2017.01.001

311

Loss
 
KL
,
Shaddy
 
RE
,
Kantor
 
PF
.
Recent and upcoming drug therapies for pediatric heart failure
.
Front Pediatr
 
2021
;
9
:
681224
. https://doi.org/10.3389/fped.2021.681224

312

Arya
 
A
,
Azad
 
S
,
Sitaraman
 
R
.
Angiotensin receptor and neprylisin inhibitor: a new drug in pediatric cardiologist’s armamentarium
.
Ann Pediatr Cardiol
 
2020
;
13
:
334
336
. https://doi.org/10.4103/apc.APC_9_20

313

Biagini
 
E
,
Spirito
 
P
,
Leone
 
O
,
Picchio
 
FM
,
Coccolo
 
F
,
Ragni
 
L
, et al.  
Heart transplantation in hypertrophic cardiomyopathy
.
Am J Cardiol
 
2008
;
101
:
387
392
. https://doi.org/10.1016/j.amjcard.2007.09.085

314

Bograd
 
AJ
,
Mital
 
S
,
Schwarzenberger
 
JC
,
Mosca
 
RS
,
Quaegebeur
 
JM
,
Addonizio
 
LJ
, et al.  
Twenty-year experience with heart transplantation for infants and children with restrictive cardiomyopathy: 1986–2006
.
Am J Transplant
 
2008
;
8
:
201
207
. https://doi.org/10.1111/j.1600-6143.2007.02027.x

315

Marstrand
 
P
,
Han
 
L
,
Day
 
SM
,
Olivotto
 
I
,
Ashley
 
EA
,
Michels
 
M
, et al.  
Hypertrophic cardiomyopathy with left ventricular systolic dysfunction: insights from the SHaRe registry
.
Circulation
 
2020
;
141
:
1371
1383
. https://doi.org/10.1161/CIRCULATIONAHA.119.044366

316

DePasquale
 
EC
,
Nasir
 
K
,
Jacoby
 
DL
.
Outcomes of adults with restrictive cardiomyopathy after heart transplantation
.
J Heart Lung Transplant
 
2012
;
31
:
1269
1275
. https://doi.org/10.1016/j.healun.2012.09.018

317

Khush
 
KK
,
Cherikh
 
WS
,
Chambers
 
DC
,
Harhay
 
MO
,
Hayes
 
D
 Jr
,
Hsich
 
E
, et al.  
The international thoracic organ transplant registry of the International Society for Heart and Lung Transplantation: thirty-sixth adult heart transplantation report – 2019; focus theme: donor and recipient size match
.
J Heart Lung Transplant
 
2019
;
38
:
1056
1066
. https://doi.org/10.1016/j.healun.2019.08.004

318

Lund
 
LH
,
Edwards
 
LB
,
Kucheryavaya
 
AY
,
Dipchand
 
AI
,
Benden
 
C
,
Christie
 
JD
, et al.  
The Registry of the International Society for Heart and Lung Transplantation: Thirtieth Official Adult Heart Transplant Report–2013; focus theme: age
.
J Heart Lung Transplant
 
2013
;
32
:
951
964
. https://doi.org/10.1016/j.healun.2013.08.006

319

Mehra
 
MR
,
Canter
 
CE
,
Hannan
 
MM
,
Semigran
 
MJ
,
Uber
 
PA
,
Baran
 
DA
, et al.  
The 2016 International Society for Heart Lung Transplantation listing criteria for heart transplantation: a 10-year update
.
J Heart Lung Transplant
 
2016
;
35
:
1
23
. https://doi.org/10.1016/j.healun.2015.10.023

320

Bansal
 
A
,
Akhtar
 
F
,
Desai
 
S
,
Velasco-Gonzalez
 
C
,
Bansal
 
A
,
Teagle
 
A
, et al.  
Six-month outcomes in postapproval HeartMate3 patients: a single-center US experience
.
J Card Surg
 
2022
;
37
:
1907
1914
. https://doi.org/10.1111/jocs.16452

321

Mehra
 
MR
,
Uriel
 
N
,
Naka
 
Y
,
Cleveland
 
JC
 Jr
,
Yuzefpolskaya
 
M
,
Salerno
 
CT
, et al.  
A fully magnetically levitated left ventricular assist device – final report
.
N Engl J Med
 
2019
;
380
:
1618
1627
. https://doi.org/10.1056/NEJMoa1900486

322

Mehra
 
MR
,
Goldstein
 
DJ
,
Uriel
 
N
,
Cleveland
 
JC
 Jr
,
Yuzefpolskaya
 
M
,
Salerno
 
C
, et al.  
Two-year outcomes with a magnetically levitated cardiac pump in heart failure
.
N Engl J Med
 
2018
;
378
:
1386
1395
. https://doi.org/10.1056/NEJMoa1800866

323

Zimpfer
 
D
,
Gustafsson
 
F
,
Potapov
 
E
,
Pya
 
Y
,
Schmitto
 
J
,
Berchtold-Herz
 
M
, et al.  
Two-year outcome after implantation of a full magnetically levitated left ventricular assist device: results from the ELEVATE Registry
.
Eur Heart J
 
2020
;
41
:
3801
3809
. https://doi.org/10.1093/eurheartj/ehaa639

324

Kirklin
 
JK
,
Naftel
 
DC
,
Pagani
 
FD
,
Kormos
 
RL
,
Stevenson
 
LW
,
Blume
 
ED
, et al.  
Seventh INTERMACS annual report: 15,000 patients and counting
.
J Heart Lung Transplant
 
2015
;
34
:
1495
1504
. https://doi.org/10.1016/j.healun.2015.10.003

325

Rose
 
EA
,
Gelijns
 
AC
,
Moskowitz
 
AJ
,
Heitjan
 
DF
,
Stevenson
 
LW
,
Dembitsky
 
W
, et al.  
Randomized evaluation of mechanical assistance for the treatment of Congestive Heart Failure Study Group. Long-term use of a left ventricular assist device for end-stage heart failure
.
N Engl J Med
 
2001
;
345
:
1435
1443
. https://doi.org/10.1056/NEJMoa012175

326

Rogers
 
JG
,
Butler
 
J
,
Lansman
 
SL
,
Gass
 
A
,
Portner
 
PM
,
Pasque
 
MK
, et al.  
Chronic mechanical circulatory support for inotrope-dependent heart failure patients who are not transplant candidates: results of the INTrEPID Trial
.
J Am Coll Cardiol
 
2007
;
50
:
741
747
. https://doi.org/10.1016/j.jacc.2007.03.063

327

Slaughter
 
MS
,
Rogers
 
JG
,
Milano
 
CA
,
Russell
 
SD
,
Conte
 
JV
,
Feldman
 
D
, et al.  
Advanced heart failure treated with continuous-flow left ventricular assist device
.
N Engl J Med
 
2009
;
361
:
2241
2251
. https://doi.org/10.1056/NEJMoa0909938

328

Goldstein
 
DJ
,
Naka
 
Y
,
Horstmanshof
 
D
,
Ravichandran
 
AK
,
Schroder
 
J
,
Ransom
 
J
, et al.  
Association of clinical outcomes with left ventricular assist device use by bridge to transplant or destination therapy intent: the multicenter study of MagLev technology in patients undergoing mechanical circulatory support therapy with HeartMate 3 (MOMENTUM 3) randomized clinical trial
.
JAMA Cardiol
 
2020
;
5
:
411
419
. https://doi.org/10.1001/jamacardio.2019.5323

329

Theochari
 
CA
,
Michalopoulos
 
G
,
Oikonomou
 
EK
,
Giannopoulos
 
S
,
Doulamis
 
IP
,
Villela
 
MA
, et al.  
Heart transplantation versus left ventricular assist devices as destination therapy or bridge to transplantation for 1-year mortality: a systematic review and meta-analysis
.
Ann Cardiothorac Surg
 
2018
;
7
:
3
11
. https://doi.org/10.21037/acs.2017.09.18

330

Jorde
 
UP
,
Kushwaha
 
SS
,
Tatooles
 
AJ
,
Naka
 
Y
,
Bhat
 
G
,
Long
 
JW
, et al.  
Results of the destination therapy post-food and drug administration approval study with a continuous flow left ventricular assist device: a prospective study using the INTERMACS registry (Interagency Registry for Mechanically Assisted Circulatory Support)
.
J Am Coll Cardiol
 
2014
;
63
:
1751
1757
. https://doi.org/10.1016/j.jacc.2014.01.053

331

Charron
 
P
,
Elliott
 
PM
,
Gimeno
 
JR
,
Caforio
 
ALP
,
Kaski
 
JP
,
Tavazzi
 
L
, et al.  
The Cardiomyopathy registry of the EURObservational Research Programme of the European Society of Cardiology: baseline data and contemporary management of adult patients with cardiomyopathies
.
Eur Heart J
 
2018
;
39
:
1784
1793
. https://doi.org/10.1093/eurheartj/ehx819

332

Mizia-Stec
 
K
,
Caforio
 
ALP
,
Charron
 
P
,
Gimeno
 
JR
,
Elliott
 
P
,
Kaski
 
JP
, et al.  
Atrial fibrillation, anticoagulation management and risk of stroke in the Cardiomyopathy/Myocarditis registry of the EURObservational Research Programme of the European Society of Cardiology
.
ESC Heart Fail
 
2020
;
7
:
3601
3609
. https://doi.org/10.1002/ehf2.12854

333

Gimeno
 
JR
,
Elliott
 
PM
,
Tavazzi
 
L
,
Tendera
 
M
,
Kaski
 
JP
,
Laroche
 
C
, et al.  
Prospective follow-up in various subtypes of cardiomyopathies: insights from the ESC EORP Cardiomyopathy Registry
.
Eur Heart J Qual Care Clin Outcomes
 
2021
;
7
:
134
142
. https://doi.org/10.1093/ehjqcco/qcaa075

334

Fauchier
 
L
,
Bisson
 
A
,
Bodin
 
A
,
Herbert
 
J
,
Spiesser
 
P
,
Pierre
 
B
, et al.  
Ischemic stroke in patients with hypertrophic cardiomyopathy according to presence or absence of atrial fibrillation
.
Stroke
 
2022
;
53
:
497
504
. https://doi.org/10.1161/STROKEAHA.121.034213

335

Buckley
 
BJR
,
Harrison
 
SL
,
Gupta
 
D
,
Fazio-Eynullayeva
 
E
,
Underhill
 
P
,
Lip
 
GYH
.
Atrial fibrillation in patients with cardiomyopathy: prevalence and clinical outcomes from real-world data
.
J Am Heart Assoc
 
2021
;
10
:
e021970
. https://doi.org/10.1161/JAHA.121.021970

336

Hindricks
 
G
,
Potpara
 
T
,
Dagres
 
N
,
Arbelo
 
E
,
Bax
 
JJ
,
Blomstrom-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
. https://doi.org/10.1093/eurheartj/ehaa612

337

Lip
 
GYH
.
The ABC pathway: an integrated approach to improve AF management
.
Nat Rev Cardiol
 
2017
;
14
:
627
628
. https://doi.org/10.1038/nrcardio.2017.153

338

Proietti
 
M
,
Romiti
 
GF
,
Olshansky
 
B
,
Lane
 
DA
,
Lip
 
GYH
.
Improved outcomes by integrated care of anticoagulated patients with atrial fibrillation using the simple ABC (Atrial Fibrillation Better Care) pathway
.
Am J Med
 
2018
;
131
:
1359
1366.e6
. https://doi.org/10.1016/j.amjmed.2018.06.012

339

Yoon
 
M
,
Yang
 
P-S
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
T-H
,
Uhm
 
J-S
, et al.  
Improved population-based clinical outcomes of patients with atrial fibrillation by compliance with the simple ABC (Atrial Fibrillation Better Care) pathway for integrated care management: a nationwide cohort study
.
Thromb Haemost
 
2019
;
119
:
1695
1703
. https://doi.org/10.1055/s-0039-1693516

340

Pastori
 
D
,
Farcomeni
 
A
,
Pignatelli
 
P
,
Violi
 
F
,
Lip
 
GYH
.
ABC (Atrial Fibrillation Better Care) pathway and healthcare costs in atrial fibrillation: the ATHERO-AF study
.
Am J Med
 
2019
;
132
:
856
861
. https://doi.org/10.1016/j.amjmed.2019.01.003

341

Pastori
 
D
,
Pignatelli
 
P
,
Menichelli
 
D
,
Violi
 
F
,
Lip
 
GYH
.
Integrated care management of patients with atrial fibrillation and risk of cardiovascular events: the ABC (Atrial Fibrillation Better Care) pathway in the ATHERO-AF study cohort
.
Mayo Clin Proc
 
2019
;
94
:
1261
1267
. https://doi.org/10.1016/j.mayocp.2018.10.022

342

Proietti
 
M
,
Lip
 
GYH
,
Laroche
 
C
,
Fauchier
 
L
,
Marin
 
F
,
Nabauer
 
M
, et al.  
Relation of outcomes to ABC (Atrial Fibrillation Better Care) pathway adherent care in European patients with atrial fibrillation: an analysis from the ESC-EHRA EORP Atrial Fibrillation General Long-Term (AFGen LT) Registry
.
Europace
 
2021
;
23
:
174
183
. https://doi.org/10.1093/europace/euaa274

343

Pastori
 
D
,
Menichelli
 
D
,
Violi
 
F
,
Pignatelli
 
P
,
Lip
 
GYH
;
ATHERO-AF study group
.
The Atrial Fibrillation Better Care (ABC) pathway and cardiac complications in atrial fibrillation: a potential sex-based difference. The ATHERO-AF study
.
Eur J Intern Med
 
2021
;
85
:
80
85
. https://doi.org/10.1016/j.ejim.2020.12.011

344

Stevens
 
D
,
Harrison
 
SL
,
Kolamunnage-Dona
 
R
,
Lip
 
GYH
,
Lane
 
DA
.
The Atrial Fibrillation Better Care pathway for managing atrial fibrillation: a review
.
Europace
 
2021
;
23
:
1511
1527
. https://doi.org/10.1093/europace/euab092

345

Yao
 
Y
,
Guo
 
Y
,
Lip
 
GYH
;
mAF-App II Trial investigators
.
The effects of implementing a mobile health-technology supported pathway on atrial fibrillation-related adverse events among patients with multimorbidity: the mAFA-II randomized clinical trial
.
JAMA Netw Open
 
2021
;
4
:
e2140071
. https://doi.org/10.1001/jamanetworkopen.2021.40071

346

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
414
. https://doi.org/10.1055/a-1515-9630

347

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
2996
. https://doi.org/10.1093/eurheartj/ehx739

348

Wang
 
YF
,
Jiang
 
C
,
He
 
L
,
Du
 
X
,
Sang
 
CH
,
Long
 
DY
, et al.  
Integrated care of atrial fibrillation using the ABC (Atrial fibrillation Better Care) pathway improves clinical outcomes in Chinese population: an analysis from the Chinese atrial fibrillation registry
.
Front Cardiovasc Med
 
2021
;
8
:
762245
. https://doi.org/10.3389/fcvm.2021.762245

349

Proietti
 
M
,
Romiti
 
GF
,
Olshansky
 
B
,
Lane
 
DA
,
Lip
 
GYH
.
Comprehensive management with the ABC (Atrial fibrillation Better Care) pathway in clinically complex patients with atrial fibrillation: a post hoc ancillary analysis from the AFFIRM trial
.
J Am Heart Assoc
 
2020
;
9
:
e014932
. https://doi.org/10.1161/JAHA.119.014932

350

Gumprecht
 
J
,
Domek
 
M
,
Proietti
 
M
,
Li
 
YG
,
Asaad
 
N
,
Rashed
 
W
, et al.  
Compliance of atrial fibrillation treatment with the Atrial fibrillation Better Care (ABC) pathway improves the clinical outcomes in the Middle East population: a report from the Gulf Survey of Atrial Fibrillation Events (SAFE) registry
.
J Clin Med
 
2020
;
9
:
1286
. https://doi.org/10.3390/jcm9051286

351

Proietti
 
M
,
Vitolo
 
M
,
Lip
 
GYH
.
Integrated care and outcomes in patients with atrial fibrillation and comorbidities
.
Eur J Clin Invest
 
2021
;
51
:
e13498
. https://doi.org/10.1111/eci.13498

352

Rivera-Caravaca
 
JM
,
Roldan
 
V
,
Martinez-Montesinos
 
L
,
Vicente
 
V
,
Lip
 
GYH
,
Marin
 
F
.
The Atrial Fibrillation Better Care (ABC) pathway and clinical outcomes in patients with atrial fibrillation: the prospective Murcia AF Project Phase II Cohort
.
J Gen Intern Med
 
2023
;
38
:
315
323
. https://doi.org/10.1007/s11606-022-07567-5

353

Vitolo
 
M
,
Proietti
 
M
,
Malavasi
 
VL
,
Bonini
 
N
,
Romiti
 
GF
,
Imberti
 
JF
, et al.  
Adherence to the “Atrial fibrillation Better Care” (ABC) pathway in patients with atrial fibrillation and cancer: a report from the ESC-EHRA EURObservational Research Programme in atrial fibrillation (EORP-AF) General Long-Term Registry
.
Eur J Intern Med
 
2022
;
105
:
54
62
. https://doi.org/10.1016/j.ejim.2022.08.004

354

Patel
 
SM
,
Palazzolo
 
MG
,
Murphy
 
SA
,
Antman
 
EM
,
Braunwald
 
E
,
Lanz
 
HJ
, et al.  
Evaluation of the atrial fibrillation better care pathway in the ENGAGE AF-TIMI 48 trial
.
Europace
 
2022
;
24
:
1730
1738
. https://doi.org/10.1093/europace/euac082

355

Yang
 
PS
,
Sung
 
JH
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
,
Lip
 
GYH
, et al.  
Application of the simple atrial fibrillation better care pathway for integrated care management in frail patients with atrial fibrillation: a nationwide cohort study
.
J Arrhythm
 
2020
;
36
:
668
677
. https://doi.org/10.1002/joa3.12364

356

Romiti
 
GF
,
Proietti
 
M
,
Vitolo
 
M
,
Bonini
 
N
,
Fawzy
 
AM
,
Ding
 
WY
, et al.  
Clinical complexity and impact of the ABC (Atrial fibrillation Better Care) pathway in patients with atrial fibrillation: a report from the ESC-EHRA EURObservational Research Programme in AF General Long-Term Registry
.
BMC Med
 
2022
;
20
:
326
. https://doi.org/10.1186/s12916-022-02526-7

357

Yang
 
PS
,
Sung
 
JH
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
,
Uhm
 
JS
, et al.  
The effect of integrated care management on dementia in atrial fibrillation
.
J Clin Med
 
2020
;
9
:
1696
. https://doi.org/10.3390/jcm9061696

358

Kotalczyk
 
A
,
Guo
 
Y
,
Stefil
 
M
,
Wang
 
Y
,
Lip
 
GYH
,
Chi
 
ORI
.
Effects of the atrial fibrillation better care pathway on outcomes among clinically complex Chinese patients with atrial fibrillation with multimorbidity and polypharmacy: a report from the ChiOTEAF registry
.
J Am Heart Assoc
 
2022
;
11
:
e024319
. https://doi.org/10.1161/JAHA.121.024319

359

Esteve-Pastor
 
MA
,
Ruiz-Ortiz
 
M
,
Muniz
 
J
,
Roldan-Rabadan
 
I
,
Otero
 
D
,
Cequier
 
A
, et al.  
Impact of integrated care management on clinical outcomes in atrial fibrillation patients: a report from the FANTASIIA registry
.
Front Cardiovasc Med
 
2022
;
9
:
856222
. https://doi.org/10.3389/fcvm.2022.856222

360

Guo
 
Y
,
Imberti
 
JF
,
Kotalczyk
 
A
,
Wang
 
Y
,
Lip
 
GYH
,
Chi
 
ORI
.
4S-AF scheme and ABC pathway guided management improves outcomes in atrial fibrillation patients
.
Eur J Clin Invest
 
2022
;
52
:
e13751
. https://doi.org/10.1111/eci.13751

361

Guo
 
Y
,
Lane
 
DA
,
Wang
 
L
,
Zhang
 
H
,
Wang
 
H
,
Zhang
 
W
, et al.  
Mobile health technology to improve care for patients with atrial fibrillation
.
J Am Coll Cardiol
 
2020
;
75
:
1523
1534
. https://doi.org/10.1016/j.jacc.2020.01.052

362

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
111
. https://doi.org/10.1016/j.ejim.2020.09.024

363

Koniaris
 
LS
,
Goldhaber
 
SZ
.
Anticoagulation in dilated cardiomyopathy
.
J Am Coll Cardiol
 
1998
;
31
:
745
748
. https://doi.org/10.1016/S0735-1097(98)00003-5

364

Eapen
 
ZJ
,
Mi
 
X
,
Fonarow
 
GC
,
Setoguchi
 
S
,
Piccini
 
JP
,
Mills
 
RM
, et al.  
Anticoagulation and clinical outcomes in heart failure patients with atrial fibrillation: findings from the ADHERE registry
.
J Atr Fibrillation
 
2013
;
6
:
953
.

365

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
472
. https://doi.org/10.1136/heartjnl-2013-304276

366

Camm
 
CF
,
Camm
 
AJ
.
Atrial fibrillation and anticoagulation in hypertrophic cardiomyopathy
.
Arrhythm Electrophysiol Rev
 
2017
;
6
:
63
68
. https://doi.org/10.15420/aer.2017:4:2

367

Nasser
 
MF
,
Gandhi
 
S
,
Siegel
 
RJ
,
Rader
 
F
.
Anticoagulation for stroke prevention in patients with hypertrophic cardiomyopathy and atrial fibrillation: a review
.
Heart Rhythm
 
2021
;
18
:
297
302
. https://doi.org/10.1016/j.hrthm.2020.09.018

368

van Rijsingen
 
IAW
,
Bakker
 
A
,
Azim
 
D
,
Hermans-van Ast
 
JF
,
van der Kooi
 
AJ
,
van Tintelen
 
JP
, et al.  
Lamin A/C mutation is independently associated with an increased risk of arterial and venous thromboembolic complications
.
Int J Cardiol
 
2013
;
168
:
472
477
. https://doi.org/10.1016/j.ijcard.2012.09.118

369

Guttmann
 
OP
,
Pavlou
 
M
,
O’Mahony
 
C
,
Monserrat
 
L
,
Anastasakis
 
A
,
Rapezzi
 
C
, et al.  
Prediction of thrombo-embolic risk in patients with hypertrophic cardiomyopathy (HCM Risk-CVA)
.
Eur J Heart Fail
 
2015
;
17
:
837
845
. https://doi.org/10.1002/ejhf.316

370

Garcia-Pavia
 
P
,
Bengel
 
F
,
Brito
 
D
,
Damy
 
T
,
Duca
 
F
,
Dorbala
 
S
, et al.  
Expert consensus on the monitoring of transthyretin amyloid cardiomyopathy
.
Eur J Heart Fail
 
2021
;
23
:
895
905
. https://doi.org/10.1002/ejhf.2198

371

Jung
 
H
,
Yang
 
P-S
,
Sung
 
J-H
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
T-H
, et al.  
Hypertrophic cardiomyopathy in patients with atrial fibrillation: prevalence and associated stroke risks in a nationwide cohort study
.
Thromb Haemost
 
2019
;
119
:
285
293
. https://doi.org/10.1055/s-0038-1676818

372

Jung
 
H
,
Sung
 
J-H
,
Yang
 
P-S
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
T-H
, et al.  
Stroke risk stratification for atrial fibrillation patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2018
;
72
:
2409
2411
. https://doi.org/10.1016/j.jacc.2018.07.098

373

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
1396
. https://doi.org/10.1002/ejhf.2566

374

Jung
 
H
,
Yang
 
PS
,
Jang
 
E
,
Yu
 
HT
,
Kim
 
TH
,
Uhm
 
JS
, et al.  
Effectiveness and safety of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation with hypertrophic cardiomyopathy: a nationwide cohort study
.
Chest
 
2019
;
155
:
354
363
. https://doi.org/10.1016/j.chest.2018.11.009

375

Garcia-Pavia
 
P
,
Rapezzi
 
C
,
Adler
 
Y
,
Arad
 
M
,
Basso
 
C
,
Brucato
 
A
, et al.  
Diagnosis and treatment of cardiac amyloidosis: a position statement of the ESC Working Group on Myocardial and Pericardial Diseases
.
Eur Heart J
 
2021
;
42
:
1554
1568
. https://doi.org/10.1093/eurheartj/ehab072

376

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
962
. https://doi.org/10.1016/S0140-6736(13)62343-0

377

Xiong
 
Q
,
Lau
 
YC
,
Senoo
 
K
,
Lane
 
DA
,
Hong
 
K
,
Lip
 
GYH
.
Non-vitamin K antagonist oral anticoagulants (NOACs) in patients with concomitant atrial fibrillation and heart failure: a systemic review and meta-analysis of randomized trials
.
Eur J Heart Fail
 
2015
;
17
:
1192
1200
. https://doi.org/10.1002/ejhf.343

378

Noseworthy
 
PA
,
Yao
 
X
,
Shah
 
ND
,
Gersh
 
BJ
.
Stroke and bleeding risks in NOAC- and warfarin-treated patients with hypertrophic cardiomyopathy and atrial fibrillation
.
J Am Coll Cardiol
 
2016
;
67
:
3020
3021
. https://doi.org/10.1016/j.jacc.2016.04.026

379

Lee
 
HJ
,
Kim
 
HK
,
Jung
 
JH
,
Han
 
KD
,
Lee
 
H
,
Park
 
JB
, et al.  
Novel oral anticoagulants for primary stroke prevention in hypertrophic cardiomyopathy patients with atrial fibrillation
.
Stroke
 
2019
;
50
:
2582
2586
. https://doi.org/10.1161/STROKEAHA.119.026048

380

Lin
 
Y
,
Xiong
 
H
,
Su
 
J
,
Lin
 
J
,
Zhou
 
Q
,
Lin
 
M
, et al.  
Effectiveness and safety of non-vitamin K antagonist oral anticoagulants in patients with hypertrophic cardiomyopathy with non-valvular atrial fibrillation
.
Heart Vessels
 
2022
;
37
:
1224
1231
. https://doi.org/10.1007/s00380-022-02021-2

381

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
1373
. https://doi.org/10.1056/NEJMoa1001337

382

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
942
. https://doi.org/10.1093/europace/eul106

383

Sartipy
 
U
,
Savarese
 
G
,
Dahlstrom
 
U
,
Fu
 
M
,
Lund
 
LH
.
Association of heart rate with mortality in sinus rhythm and atrial fibrillation in heart failure with preserved ejection fraction
.
Eur J Heart Fail
 
2019
;
21
:
471
479
. https://doi.org/10.1002/ejhf.1389

384

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
. https://doi.org/10.1016/j.amjcard.2019.12.025

385

Van Gelder
 
IC
,
Rienstra
 
M
,
Crijns
 
HJ
,
Olshansky
 
B
.
Rate control in atrial fibrillation
.
Lancet
 
2016
;
388
:
818
828
. https://doi.org/10.1016/S0140-6736(16)31258-2

386

Kotecha
 
D
,
Flather
 
MD
,
Altman
 
DG
,
Holmes
 
J
,
Rosano
 
G
,
Wikstrand
 
J
, et al.  
Heart rate and rhythm and the benefit of beta-blockers in patients with heart failure
.
J Am Coll Cardiol
 
2017
;
69
:
2885
2896
. https://doi.org/10.1016/j.jacc.2017.04.001

387

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
2508
. https://doi.org/10.1001/jama.2020.23138

388

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
. https://doi.org/10.1093/eurheartj/ehy555

389

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
4739
. https://doi.org/10.1093/eurheartj/ehab569

390

Huang
 
W
,
Wang
 
S
,
Su
 
L
,
Fu
 
G
,
Su
 
Y
,
Chen
 
K
, et al.  
His-bundle pacing vs biventricular pacing following atrioventricular nodal ablation in patients with atrial fibrillation and reduced ejection fraction: a multicenter, randomized, crossover study–The ALTERNATIVE-AF trial
.
Heart Rhythm
 
2022
;
19
:
1948
1955
. https://doi.org/10.1016/j.hrthm.2022.07.009

391

Guttmann
 
OP
,
Pavlou
 
M
,
O’Mahony
 
C
,
Monserrat
 
L
,
Anastasakis
 
A
,
Rapezzi
 
C
, et al.  
Predictors of atrial fibrillation in hypertrophic cardiomyopathy
.
Heart
 
2017
;
103
:
672
678
. https://doi.org/10.1136/heartjnl-2016-309672

392

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
315
. https://doi.org/10.1056/NEJMoa2029980

393

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
324
. https://doi.org/10.1056/NEJMoa2029554

394

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
345
. https://doi.org/10.1093/europace/euq450

395

Kober
 
L
,
Torp-Pedersen
 
C
,
McMurray
 
JJ
,
Gotzsche
 
O
,
Levy
 
S
,
Crijns
 
H
, et al.  
Increased mortality after dronedarone therapy for severe heart failure
.
N Engl J Med
 
2008
;
358
:
2678
2687
. https://doi.org/10.1056/NEJMoa0800456

396

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
2276
. https://doi.org/10.1056/NEJMoa1109867

397

Providencia
 
R
,
Elliott
 
P
,
Patel
 
K
,
McCready
 
J
,
Babu
 
G
,
Srinivasan
 
N
, et al.  
Catheter ablation for atrial fibrillation in hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
Heart
 
2016
;
102
:
1533
1543
. https://doi.org/10.1136/heartjnl-2016-309406

398

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
1274
. https://doi.org/10.1001/jama.2019.0693

399

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
1644
. https://doi.org/10.1161/CIRCULATIONAHA.115.019406

400

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
1390
. https://doi.org/10.1161/CIRCULATIONAHA.120.050991

401

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
427
. https://doi.org/10.1056/NEJMoa1707855

402

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
1316
. https://doi.org/10.1056/NEJMoa2019422

403

Rillig
 
A
,
Magnussen
 
C
,
Ozga
 
AK
,
Suling
 
A
,
Brandes
 
A
,
Breithardt
 
G
, et al.  
Early rhythm control therapy in patients with atrial fibrillation and heart failure
.
Circulation
 
2021
;
144
:
845
858
. https://doi.org/10.1161/CIRCULATIONAHA.121.056323

404

Khan
 
MN
,
Jais
 
P
,
Cummings
 
J
,
Di Biase
 
L
,
Sanders
 
P
,
Martin
 
DO
, et al.  
Pulmonary-vein isolation for atrial fibrillation in patients with heart failure
.
N Engl J Med
 
2008
;
359
:
1778
1785
. https://doi.org/10.1056/NEJMoa0708234

405

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
747
. https://doi.org/10.1136/hrt.2010.207340

406

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
1903
. https://doi.org/10.1016/j.jacc.2013.01.069

407

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
38
. https://doi.org/10.1161/CIRCEP.113.000806

408

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
1961
. https://doi.org/10.1016/j.jacc.2017.08.041

409

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
2366
. https://doi.org/10.1111/jce.15622

410

Androulakis
 
E
,
Sohrabi
 
C
,
Briasoulis
 
A
,
Bakogiannis
 
C
,
Saberwal
 
B
,
Siasos
 
G
, et al.  
Catheter ablation for atrial fibrillation in patients with heart failure with preserved ejection fraction: a systematic review and meta-analysis
.
J Clin Med
 
2022
;
11
:
288
. https://doi.org/10.3390/jcm11020288

411

Bunch
 
TJ
,
Munger
 
TM
,
Friedman
 
PA
,
Asirvatham
 
SJ
,
Brady
 
PA
,
Cha
 
YM
, et al.  
Substrate and procedural predictors of outcomes after catheter ablation for atrial fibrillation in patients with hypertrophic cardiomyopathy
.
J Cardiovasc Electrophysiol
 
2008
;
19
:
1009
1014
. https://doi.org/10.1111/j.1540-8167.2008.01192.x

412

Bassiouny
 
M
,
Lindsay
 
BD
,
Lever
 
H
,
Saliba
 
W
,
Klein
 
A
,
Banna
 
M
, et al.  
Outcomes of nonpharmacologic treatment of atrial fibrillation in patients with hypertrophic cardiomyopathy
.
Heart Rhythm
 
2015
;
12
:
1438
1447
. https://doi.org/10.1016/j.hrthm.2015.03.042

413

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
2436
. https://doi.org/10.1161/CIRCULATIONAHA.117.029267

414

Chen
 
X
,
Dong
 
JZ
,
Du
 
X
,
Wu
 
JH
,
Yu
 
RH
,
Long
 
DY
, et al.  
Long-term outcome of catheter ablation for atrial fibrillation in patients with apical hypertrophic cardiomyopathy
.
J Cardiovasc Electrophysiol
 
2018
;
29
:
951
957
. https://doi.org/10.1111/jce.13645

415

Di Donna
 
P
,
Olivotto
 
I
,
Delcre
 
SDL
,
Caponi
 
D
,
Scaglione
 
M
,
Nault
 
I
, et al.  
Efficacy of catheter ablation for atrial fibrillation in hypertrophic cardiomyopathy: impact of age, atrial remodelling, and disease progression
.
Europace
 
2010
;
12
:
347
355
. https://doi.org/10.1093/europace/euq013

416

Santangeli
 
P
,
Di Biase
 
L
,
Themistoclakis
 
S
,
Raviele
 
A
,
Schweikert
 
RA
,
Lakkireddy
 
D
, et al.  
Catheter ablation of atrial fibrillation in hypertrophic cardiomyopathy: long-term outcomes and mechanisms of arrhythmia recurrence
.
Circ Arrhythm Electrophysiol
 
2013
;
6
:
1089
1094
. https://doi.org/10.1161/CIRCEP.113.000339

417

Ha
 
HS
,
Wang
 
N
,
Wong
 
S
,
Phan
 
S
,
Liao
 
J
,
Kumar
 
N
, et al.  
Catheter ablation for atrial fibrillation in hypertrophic cardiomyopathy patients: a systematic review
.
J Interv Card Electrophysiol
 
2015
;
44
:
161
170
. https://doi.org/10.1007/s10840-015-0047-8

418

Zhao
 
DS
,
Shen
 
Y
,
Zhang
 
Q
,
Lin
 
G
,
Lu
 
YH
,
Chen
 
BT
, et al.  
Outcomes of catheter ablation of atrial fibrillation in patients with hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
Europace
 
2016
;
18
:
508
520
. https://doi.org/10.1093/europace/euv339

419

Lapenna
 
E
,
Pozzoli
 
A
,
De Bonis
 
M
,
La Canna
 
G
,
Nisi
 
T
,
Nascimbene
 
S
, et al.  
Mid-term outcomes of concomitant surgical ablation of atrial fibrillation in patients undergoing cardiac surgery for hypertrophic cardiomyopathy
.
Eur J Cardiothorac Surg
 
2017
;
51
:
1112
1118
. https://doi.org/10.1093/ejcts/ezx017

420

Gasperetti
 
A
,
James
 
CA
,
Chen
 
L
,
Schenker
 
N
,
Casella
 
M
,
Kany
 
S
, et al.  
Efficacy of catheter ablation for atrial arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy–a multicenter study
.
J Clin Med
 
2021
;
10
:
4962
. https://doi.org/10.3390/jcm10214962

421

Maron
 
BJ
,
Olivotto
 
I
,
Bellone
 
P
,
Conte
 
MR
,
Cecchi
 
F
,
Flygenring
 
BP
, et al.  
Clinical profile of stroke in 900 patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2002
;
39
:
301
307
. https://doi.org/10.1016/S0735-1097(01)01727-2

422

Losi
 
M-A
,
Betocchi
 
S
,
Aversa
 
M
,
Lombardi
 
R
,
Miranda
 
M
,
D’Alessandro
 
G
, et al.  
Determinants of atrial fibrillation development in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2004
;
94
:
895
900
. https://doi.org/10.1016/j.amjcard.2004.06.024

423

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
. https://doi.org/10.1161/JAHA.114.001002

424

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
1340
. https://doi.org/10.1002/clc.23050

425

Choi
 
Y-J
,
Choi
 
E-K
,
Han
 
K-D
,
Jung
 
J-H
,
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
135
. https://doi.org/10.1016/j.ijcard.2018.08.038

426

Yeung
 
C
,
Enriquez
 
A
,
Suarez-Fuster
 
L
,
Baranchuk
 
A
.
Atrial fibrillation in patients with inherited cardiomyopathies
.
Europace
 
2019
;
21
:
22
32
. https://doi.org/10.1093/europace/euy064

427

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
1865
. https://doi.org/10.1016/j.amjcard.2017.02.040

428

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
105
. https://doi.org/10.1016/j.amjcard.2016.08.092

429

Robinson
 
K
,
Frenneaux
 
MP
,
Stockins
 
B
,
Karatasakis
 
G
,
Poloniecki
 
JD
,
McKenna
 
WJ
.
Atrial fibrillation in hypertrophie cardiomyopathy: a longitudinal study
.
J Am Coll Cardiol
 
1990
;
15
:
1279
1285
. https://doi.org/10.1016/S0735-1097(10)80014-2

430

Gaita
 
F
,
Di Donna
 
P
,
Olivotto
 
I
,
Scaglione
 
M
,
Ferrero
 
I
,
Montefusco
 
A
, et al.  
Usefulness and safety of transcatheter ablation of atrial fibrillation in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2007
;
99
:
1575
1581
. https://doi.org/10.1016/j.amjcard.2006.12.087

431

Zheng
 
S
,
Jiang
 
W
,
Dai
 
J
,
Li
 
K
,
Shi
 
H
,
Wu
 
W
, et al.  
Five-year outcomes after catheter ablation for atrial fibrillation in patients with hypertrophic cardiomyopathy
.
J Cardiovasc Electrophysiol
 
2020
;
31
:
621
628
. https://doi.org/10.1111/jce.14349

432

Cao
 
ZJ
,
Guo
 
XG
,
Sun
 
Q
,
Yang
 
JD
,
Wei
 
HQ
,
Zhang
 
S
, et al.  
Pulmonary vein isolation implemented by second-generation cryoballoon for treating hypertrophic cardiomyopathy patients with symptomatic atrial fibrillation: a case-control study
.
J Geriatr Cardiol
 
2020
;
17
:
476
485
.

433

Castagno
 
D
,
Di Donna
 
P
,
Olivotto
 
I
,
Frontera
 
A
,
Calo
 
L
,
Scaglione
 
M
, et al.  
Transcatheter ablation for atrial fibrillation in patients with hypertrophic cardiomyopathy: long-term results and clinical outcomes
.
J Cardiovasc Electrophysiol
 
2021
;
32
:
657
666
. https://doi.org/10.1111/jce.14880

434

Dinshaw
 
L
,
Munkler
 
P
,
Schaffer
 
B
,
Klatt
 
N
,
Jungen
 
C
,
Dickow
 
J
, et al.  
Ablation of atrial fibrillation in patients with hypertrophic cardiomyopathy: treatment strategy, characteristics of consecutive atrial tachycardia and long-term outcome
.
J Am Heart Assoc
 
2021
;
10
:
e017451
. https://doi.org/10.1161/JAHA.120.017451

435

Creta
 
A
,
Elliott
 
P
,
Earley
 
MJ
,
Dhinoja
 
M
,
Finlay
 
M
,
Sporton
 
S
, et al.  
Catheter ablation of atrial fibrillation in patients with hypertrophic cardiomyopathy: a European observational multicentre study
.
Europace
 
2021
;
23
:
1409
1417
. https://doi.org/10.1093/europace/euab022

436

Grünig
 
E
,
Tasman
 
JA
,
Kücherer
 
H
,
Franz
 
W
,
Kübler
 
W
,
Katus
 
HA
.
Frequency and phenotypes of familial dilated cardiomyopathy
.
J Am Coll Cardiol
 
1998
;
31
:
186
194
. https://doi.org/10.1016/S0735-1097(97)00434-8

437

Bourfiss
 
M
,
Riele
 
ASJMT
,
Mast
 
TP
,
Cramer
 
MJ
,
Heijden
 
JF
,
Veen
 
TABV
, 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
1428
. https://doi.org/10.1111/jce.13094

438

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
1260
. https://doi.org/10.1016/j.jacc.2008.06.044

439

Van Rijsingen
 
IAW
,
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
384
. https://doi.org/10.1093/eurjhf/hfs191

440

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
2307
. https://doi.org/10.1016/j.jacc.2016.08.058

441

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
 
2017
;
39
:
853
860
. https://doi.org/10.1093/eurheartj/ehx596

442

Cikes
 
M
,
Claggett
 
B
,
Shah
 
AM
,
Desai
 
AS
,
Lewis
 
EF
,
Shah
 
SJ
, et al.  
Atrial fibrillation in heart failure with preserved ejection fraction: the TOPCAT trial
.
JACC Heart Fail
 
2018
;
6
:
689
697
. https://doi.org/10.1016/j.jchf.2018.05.005

443

Zafrir
 
B
,
Lund
 
LH
,
Laroche
 
C
,
Ruschitzka
 
F
,
Crespo-Leiro
 
MG
,
Coats
 
AJS
, et al.  
Prognostic implications of atrial fibrillation in heart failure with reduced, mid-range, and preserved ejection fraction: a report from 14 964 patients in the European Society of Cardiology Heart Failure Long-Term Registry
.
Eur Heart J
 
2018
;
39
:
4277
4284
. https://doi.org/10.1093/eurheartj/ehy626

444

Cikes
 
M
,
Planinc
 
I
,
Claggett
 
B
,
Cunningham
 
J
,
Milicic
 
D
,
Sweitzer
 
N
, et al.  
Atrial fibrillation in heart failure with preserved ejection fraction: the PARAGON-HF trial
.
JACC Heart Fail
 
2022
;
10
:
336
346
. https://doi.org/10.1016/j.jchf.2022.01.018

445

Ducharme
 
A
,
Swedberg
 
K
,
Pfeffer
 
MA
,
Cohen-Solal
 
A
,
Granger
 
CB
,
Maggioni
 
AP
, et al.  
Prevention of atrial fibrillation in patients with symptomatic chronic heart failure by candesartan in the Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity (CHARM) program
.
Am Heart J
 
2006
;
152
:
86
92
. https://doi.org/10.1016/j.ahj.2005.06.036

446

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
. https://doi.org/10.1016/j.amjcard.2020.11.039

447

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
722
. https://doi.org/10.1016/j.amjcard.2010.04.031

448

Camm
 
CF
,
James
 
CA
,
Tichnell
 
C
,
Murray
 
B
,
Bhonsale
 
A
,
Te Riele
 
ASJM
, et al.  
Prevalence of atrial arrhythmias in arrhythmogenic right ventricular dysplasia/cardiomyopathy
.
Heart Rhythm
 
2013
;
10
:
1661
1668
. https://doi.org/10.1016/j.hrthm.2013.08.032

449

Saguner
 
AM
,
Brunckhorst
 
C
,
Duru
 
F
.
Atrial arrhythmias in arrhythmogenic cardiomyopathy: at the beginning or at the end of the disease story? Reply
.
Circ J
 
2015
;
79
:
447
. https://doi.org/10.1253/circj.CJ-14-1234

450

Müssigbrodt
 
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
1187
. https://doi.org/10.1093/europace/eux179

451

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
2861
. https://doi.org/10.1253/circj.CJ-14-0474

452

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
;
9
:
CD005049
. https://doi.org/10.1002/14651858.CD005049.pub4

453

Muchtar
 
E
,
Gertz
 
MA
,
Kumar
 
SK
,
Lin
 
G
,
Boilson
 
B
,
Clavell
 
A
, et al.  
Digoxin use in systemic light-chain (AL) amyloidosis: contra-indicated or cautious use?
 
Amyloid
 
2018
;
25
:
86
92
. https://doi.org/10.1080/13506129.2018.1449744

454

Donnelly
 
JP
,
Sperry
 
BW
,
Gabrovsek
 
A
,
Ikram
 
A
,
Tang
 
WHW
,
Estep
 
J
, et al.  
Digoxin use in cardiac amyloidosis
.
Am J Cardiol
 
2020
;
133
:
134
138
. https://doi.org/10.1016/j.amjcard.2020.07.034

455

Pollak
 
A
,
Falk
 
RH
.
Left ventricular systolic dysfunction precipitated by verapamil in cardiac amyloidosis
.
Chest
 
1993
;
104
:
618
620
. https://doi.org/10.1378/chest.104.2.618

456

Yang
 
YJ
,
Yuan
 
JQ
,
Fan
 
CM
,
Pu
 
JL
,
Fang
 
PH
,
Ma
 
J
, et al.  
Incidence of ischemic stroke and systemic embolism in patients with hypertrophic cardiomyopathy, nonvalvular atrial fibrillation, CHA2DS2-VASc score of 1 and without anticoagulant therapy
.
Heart Vessels
 
2016
;
31
:
1148
1153
. https://doi.org/10.1007/s00380-015-0718-5

457

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
1501
. https://doi.org/10.1136/heartjnl-2016-310720

458

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
1754
. https://doi.org/10.1253/circj.CJ-19-0186

459

Tsuda
 
T
,
Hayashi
 
K
,
Fujino
 
N
,
Konno
 
T
,
Tada
 
H
,
Nomura
 
A
, et al.  
Effect of hypertrophic cardiomyopathy on the prediction of thromboembolism in patients with nonvalvular atrial fibrillation
.
Heart Rhythm
 
2019
;
16
:
829
837
. https://doi.org/10.1016/j.hrthm.2018.11.029

460

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
. https://doi.org/10.4022/jafib.2207

461

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
24227
. https://doi.org/10.18632/aging.104133

462

Komatsu
 
J
,
Imai
 
RI
,
Nakaoka
 
Y
,
Nishida
 
K
,
Seki
 
SI
,
Kubo
 
T
, et al.  
Importance of paroxysmal atrial fibrillation and sex differences in the prevention of embolic stroke in hypertrophic cardiomyopathy
.
Circ Rep
 
2021
;
3
:
273
278
. https://doi.org/10.1253/circrep.CR-20-0101

463

Donnellan
 
E
,
Elshazly
 
MB
,
Vakamudi
 
S
,
Wazni
 
OM
,
Cohen
 
JA
,
Kanj
 
M
, et al.  
No association between CHADS-VASc score and left atrial appendage thrombus in patients with transthyretin amyloidosis
.
JACC Clin Electrophysiol
 
2019
;
5
:
1473
1474
. https://doi.org/10.1016/j.jacep.2019.10.013

464

El-Am
 
EA
,
Dispenzieri
 
A
,
Melduni
 
RM
,
Ammash
 
NM
,
White
 
RD
,
Hodge
 
DO
, et al.  
Direct current cardioversion of atrial arrhythmias in adults with cardiac amyloidosis
.
J Am Coll Cardiol
 
2019
;
73
:
589
597
. https://doi.org/10.1016/j.jacc.2018.10.079

465

Aguilar
 
MI
,
Hart
 
R
.
Oral anticoagulants for preventing stroke in patients with non-valvular atrial fibrillation and no previous history of stroke or transient ischemic attacks
.
Cochrane Database Syst Rev
 
2005
;
3
:
CD001927
. https://doi.org/10.1002/14651858.CD001927

466

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
867
. https://doi.org/10.7326/0003-4819-146-12-200706190-00007

467

Aguilar
 
MI
,
Hart
 
R
,
Pearce
 
LA
.
Oral anticoagulants versus antiplatelet therapy for preventing stroke in patients with non-valvular atrial fibrillation and no history of stroke or transient ischemic attacks
.
Cochrane Database Syst Rev
 
2007
;
3
:
CD006186
. https://doi.org/10.1002/14651858.CD006186.pub2

468

Andersen
 
LV
,
Vestergaard
 
P
,
Deichgraeber
 
P
,
Lindholt
 
JS
,
Mortensen
 
LS
,
Frost
 
L
.
Warfarin for the prevention of systemic embolism in patients with non-valvular atrial fibrillation: a meta-analysis
.
Heart
 
2008
;
94
:
1607
1613
. https://doi.org/10.1136/hrt.2007.135657

469

Dogliotti
 
A
,
Paolasso
 
E
,
Giugliano
 
RP
.
Current and new oral antithrombotics in non-valvular atrial fibrillation: a network meta-analysis of 79 808 patients
.
Heart
 
2014
;
100
:
396
405
. https://doi.org/10.1136/heartjnl-2013-304347

470

Joundi
 
RA
,
Cipriano
 
LE
,
Sposato
 
LA
,
Saposnik
 
G
;
Stroke Outcomes Research Working Group
.
Ischemic stroke risk in patients with atrial fibrillation and CHA2DS2-VASc score of 1: systematic review and meta-analysis
.
Stroke
 
2016
;
47
:
1364
1367
. https://doi.org/10.1161/STROKEAHA.115.012609

471

Nielsen
 
PB
,
Larsen
 
TB
,
Skjoth
 
F
,
Overvad
 
TF
,
Lip
 
GY
.
Stroke and thromboembolic event rates in atrial fibrillation according to different guideline treatment thresholds: a nationwide cohort study
.
Sci Rep
 
2016
;
6
:
27410
. https://doi.org/10.1038/srep27410

472

Shin
 
SY
,
Han
 
SJ
,
Kim
 
JS
,
Im
 
SI
,
Shim
 
J
,
Ahn
 
J
, et al.  
Identification of markers associated with development of stroke in “clinically low-risk” atrial fibrillation patients
.
J Am Heart Assoc
 
2019
;
8
:
e012697
. https://doi.org/10.1161/JAHA.119.012697

473

Krittayaphong
 
R
,
Raungrattanaamporn
 
O
,
Bhuripanyo
 
K
,
Sriratanasathavorn
 
C
,
Pooranawattanakul
 
S
,
Punlee
 
K
, et al.  
A randomized clinical trial of the efficacy of radiofrequency catheter ablation and amiodarone in the treatment of symptomatic atrial fibrillation
.
J Med Assoc Thai
 
2003
;
86
:
S8
16
.

474

Stabile
 
G
,
Bertaglia
 
E
,
Senatore
 
G
,
De Simone
 
A
,
Zoppo
 
F
,
Donnici
 
G
, et al.  
Catheter ablation treatment in patients with drug-refractory atrial fibrillation: a prospective, multi-centre, randomized, controlled study (Catheter Ablation For The Cure Of Atrial Fibrillation Study)
.
Eur Heart J
 
2006
;
27
:
216
221
. https://doi.org/10.1093/eurheartj/ehi583

475

Pappone
 
C
,
Augello
 
G
,
Sala
 
S
,
Gugliotta
 
F
,
Vicedomini
 
G
,
Gulletta
 
S
, et al.  
A randomized trial of circumferential pulmonary vein ablation versus antiarrhythmic drug therapy in paroxysmal atrial fibrillation: the APAF Study
.
J Am Coll Cardiol
 
2006
;
48
:
2340
2347
. https://doi.org/10.1016/j.jacc.2006.08.037

476

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
941
. https://doi.org/10.1056/NEJMoa050955

477

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
2505
. https://doi.org/10.1161/CIRCULATIONAHA.108.772582

478

Forleo
 
GB
,
Mantica
 
M
,
De Luca
 
L
,
Leo
 
R
,
Santini
 
L
,
Panigada
 
S
, et al.  
Catheter ablation of atrial fibrillation in patients with diabetes mellitus type 2: results from a randomized study comparing pulmonary vein isolation versus antiarrhythmic drug therapy
.
J Cardiovasc Electrophysiol
 
2009
;
20
:
22
28
. https://doi.org/10.1111/j.1540-8167.2008.01275.x

479

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
340
. https://doi.org/10.1001/jama.2009.2029

480

Pappone
 
C
,
Vicedomini
 
G
,
Augello
 
G
,
Manguso
 
F
,
Saviano
 
M
,
Baldi
 
M
, et al.  
Radiofrequency catheter ablation and antiarrhythmic drug therapy: a prospective, randomized, 4-year follow-up trial: the APAF study
.
Circ Arrhythm Electrophysiol
 
2011
;
4
:
808
814
. https://doi.org/10.1161/CIRCEP.111.966408

481

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
1723
. https://doi.org/10.1016/j.jacc.2012.11.064

482

Blandino
 
A
,
Toso
 
E
,
Scaglione
 
M
,
Anselmino
 
M
,
Ferraris
 
F
,
Sardi
 
D
, et al.  
Long-term efficacy and safety of two different rhythm control strategies in elderly patients with symptomatic persistent atrial fibrillation
.
J Cardiovasc Electrophysiol
 
2013
;
24
:
731
738
. https://doi.org/10.1111/jce.12126

483

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
507
. https://doi.org/10.1093/eurheartj/eht457

484

Hummel
 
J
,
Michaud
 
G
,
Hoyt
 
R
,
DeLurgio
 
D
,
Rasekh
 
A
,
Kusumoto
 
F
, et al.  
Phased RF ablation in persistent atrial fibrillation
.
Heart Rhythm
 
2014
;
11
:
202
209
. https://doi.org/10.1016/j.hrthm.2013.11.009

485

Verma
 
A
,
Jiang
 
CY
,
Betts
 
TR
,
Chen
 
J
,
Deisenhofer
 
I
,
Mantovan
 
R
, et al.  
Approaches to catheter ablation for persistent atrial fibrillation
.
N Engl J Med
 
2015
;
372
:
1812
1822
. https://doi.org/10.1056/NEJMoa1408288

486

Reddy
 
VY
,
Dukkipati
 
SR
,
Neuzil
 
P
,
Natale
 
A
,
Albenque
 
JP
,
Kautzner
 
J
, et al.  
Randomized, controlled trial of the safety and effectiveness of a contact force-sensing irrigated catheter for ablation of paroxysmal atrial fibrillation: results of the TactiCath Contact Force Ablation Catheter Study for Atrial Fibrillation (TOCCASTAR) study
.
Circulation
 
2015
;
132
:
907
915
. https://doi.org/10.1161/CIRCULATIONAHA.114.014092

487

Dukkipati
 
SR
,
Cuoco
 
F
,
Kutinsky
 
I
,
Aryana
 
A
,
Bahnson
 
TD
,
Lakkireddy
 
D
, et al.  
Pulmonary vein isolation using the visually guided laser balloon: a prospective, multicenter, and randomized comparison to standard radiofrequency ablation
.
J Am Coll Cardiol
 
2015
;
66
:
1350
1360
. https://doi.org/10.1016/j.jacc.2015.07.036

488

Sohara
 
H
,
Ohe
 
T
,
Okumura
 
K
,
Naito
 
S
,
Hirao
 
K
,
Shoda
 
M
, et al.  
Hot balloon ablation of the pulmonary veins for paroxysmal AF: a multicenter randomized trial in Japan
.
J Am Coll Cardiol
 
2016
;
68
:
2747
2757
. https://doi.org/10.1016/j.jacc.2016.10.037

489

Bertaglia
 
E
,
Senatore
 
G
,
De Michieli
 
L
,
De Simone
 
A
,
Amellone
 
C
,
Ferretto
 
S
, et al.  
Twelve-year follow-up of catheter ablation for atrial fibrillation: a prospective, multicenter, randomized study
.
Heart Rhythm
 
2017
;
14
:
486
492
. https://doi.org/10.1016/j.hrthm.2016.12.023

490

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
1285
. https://doi.org/10.1001/jama.2019.0692

491

Contreras-Valdes
 
FM
,
Buxton
 
AE
,
Josephson
 
ME
,
Anter
 
E
.
Atrial fibrillation ablation in patients with hypertrophic cardiomyopathy: long-term outcomes and clinical predictors
.
J Am Coll Cardiol
 
2015
;
65
:
1485
1487
. https://doi.org/10.1016/j.jacc.2014.12.063

492

Rozen
 
G
,
Elbaz-Greener
 
G
,
Marai
 
I
,
Andria
 
N
,
Hosseini
 
SM
,
Biton
 
Y
, et al.  
Utilization and complications of catheter ablation for atrial fibrillation in patients with hypertrophic cardiomyopathy
.
J Am Heart Assoc
 
2020
;
9
:
e015721
. https://doi.org/10.1161/JAHA.119.015721

493

Hodges
 
K
,
Tang
 
A
,
Rivas
 
CG
,
Umana-Pizano
 
J
,
Chemtob
 
R
,
Desai
 
MY
, et al.  
Surgical ablation of atrial fibrillation in hypertrophic obstructive cardiomyopathy: outcomes of a tailored surgical approach
.
J Card Surg
 
2020
;
35
:
2957
2964
. https://doi.org/10.1111/jocs.14946

494

Meng
 
Y
,
Zhang
 
Y
,
Liu
 
P
,
Zhu
 
C
,
Lu
 
T
,
Hu
 
E
, et al.  
Clinical efficacy and safety of Cox-Maze IV procedure for atrial fibrillation in patients with hypertrophic obstructive cardiomyopathy
.
Front Cardiovasc Med
 
2021
;
8
:
720950
. https://doi.org/10.3389/fcvm.2021.720950

495

Zhang
 
HD
,
Ding
 
L
,
Weng
 
SX
,
Zhou
 
B
,
Ding
 
XT
,
Hu
 
LX
, et al.  
Characteristics and long-term ablation outcomes of supraventricular arrhythmias in hypertrophic cardiomyopathy: a 10-year, single-center experience
.
Front Cardiovasc Med
 
2021
;
8
:
766571
. https://doi.org/10.3389/fcvm.2021.766571

496

Cardona-Guarache
 
R
,
Astrom-Aneq
 
M
,
Oesterle
 
A
,
Asirvatham
 
R
,
Svetlichnaya
 
J
,
Marcus
 
GM
, et al.  
Atrial arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy: prevalence, echocardiographic predictors, and treatment
.
J Cardiovasc Electrophysiol
 
2019
;
30
:
1801
1810
. https://doi.org/10.1111/jce.14069

497

Zhao
 
L
,
Xu
 
K
,
Jiang
 
W
,
Zhou
 
L
,
Wang
 
Y
,
Zhang
 
X
, et al.  
Long-term outcomes of catheter ablation of atrial fibrillation in dilated cardiomyopathy
.
Int J Cardiol
 
2015
;
190
:
227
232
. https://doi.org/10.1016/j.ijcard.2015.04.186

498

Stollberger
 
C
,
Gatterer
 
E
,
Finsterer
 
J
,
Kuck
 
KH
,
Tilz
 
RR
.
Repeated radiofrequency ablation of atrial tachycardia in restrictive cardiomyopathy secondary to myofibrillar myopathy
.
J Cardiovasc Electrophysiol
 
2014
;
25
:
905
907
. https://doi.org/10.1111/jce.12436

499

Briceno
 
DF
,
Markman
 
TM
,
Lupercio
 
F
,
Romero
 
J
,
Liang
 
JJ
,
Villablanca
 
PA
, et al.  
Catheter ablation versus conventional treatment of atrial fibrillation in patients with heart failure with reduced ejection fraction: a systematic review and meta-analysis of randomized controlled trials
.
J Interv Card Electrophysiol
 
2018
;
53
:
19
29
. https://doi.org/10.1007/s10840-018-0425-0

500

Prabhu
 
S
,
Costello
 
BT
,
Taylor
 
AJ
,
Gutman
 
SJ
,
Voskoboinik
 
A
,
McLellan
 
AJA
, et al.  
Regression of diffuse ventricular fibrosis following restoration of sinus rhythm with catheter ablation in patients with atrial fibrillation and systolic dysfunction: a substudy of the CAMERA MRI trial
.
JACC Clin Electrophysiol
 
2018
;
4
:
999
1007
. https://doi.org/10.1016/j.jacep.2018.04.013

501

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
. https://doi.org/10.1161/CIRCEP.119.007731

502

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
2640
. https://doi.org/10.1001/jama.293.21.2634

503

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
1595
. https://doi.org/10.1056/NEJMoa1113566

504

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
. https://doi.org/10.1001/jama.2014.467

505

Nielsen
 
JC
,
Johannessen
 
A
,
Raatikainen
 
P
,
Hindricks
 
G
,
Walfridsson
 
H
,
Pehrson
 
SM
, et al.  
Long-term efficacy of catheter ablation as first-line therapy for paroxysmal atrial fibrillation: 5-year outcome in a randomised clinical trial
.
Heart
 
2017
;
103
:
368
376
. https://doi.org/10.1136/heartjnl-2016-309781

506

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
1041
. https://doi.org/10.1093/europace/euab029

507

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
1704
. https://doi.org/10.1161/CIRCULATIONAHA.121.057095

508

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
2231
. https://doi.org/10.1016/j.jacc.2014.09.028

509

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
2169
. https://doi.org/10.1016/j.jacc.2015.03.002

510

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
996
. https://doi.org/10.1016/j.jacc.2015.06.488

511

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
152
. https://doi.org/10.1016/j.jacep.2015.04.004

512

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
1373
. https://doi.org/10.1111/pace.13763

513

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
28
. https://doi.org/10.1056/NEJMoa1817591

514

Meng
 
L
,
Tseng
 
CH
,
Shivkumar
 
K
,
Ajijola
 
O
.
Efficacy of stellate ganglion blockade in managing electrical storm: a systematic review
.
JACC Clin Electrophysiol
 
2017
;
3
:
942
949
. https://doi.org/10.1016/j.jacep.2017.06.006

515

Do
 
DH
,
Bradfield
 
J
,
Ajijola
 
OA
,
Vaseghi
 
M
,
Le
 
J
,
Rahman
 
S
, et al.  
Thoracic epidural anesthesia can be effective for the short-term management of ventricular tachycardia storm
.
J Am Heart Assoc
 
2017
;
6
:
e007080
. https://doi.org/10.1161/JAHA.117.007080

516

Richardson
 
T
,
Lugo
 
R
,
Saavedra
 
P
,
Crossley
 
G
,
Clair
 
W
,
Shen
 
S
, et al.  
Cardiac sympathectomy for the management of ventricular arrhythmias refractory to catheter ablation
.
Heart Rhythm
 
2018
;
15
:
56
62
. https://doi.org/10.1016/j.hrthm.2017.09.006

517

Price
 
J
,
Mah
 
DY
,
Fynn-Thompson
 
FL
,
Tsirka
 
AE
.
Successful bilateral thoracoscopic sympathectomy for recurrent ventricular arrhythmia in a pediatric patient with hypertrophic cardiomyopathy
.
HeartRhythm Case Rep
 
2020
;
6
:
23
26
. https://doi.org/10.1016/j.hrcr.2019.10.003

518

Vaseghi
 
M
,
Barwad
 
P
,
Malavassi Corrales
 
FJ
,
Tandri
 
H
,
Mathuria
 
N
,
Shah
 
R
, et al.  
Cardiac sympathetic denervation for refractory ventricular arrhythmias
.
J Am Coll Cardiol
 
2017
;
69
:
3070
3080
. https://doi.org/10.1016/j.jacc.2017.04.035

519

Dusi
 
V
,
Gornbein
 
J
,
Do
 
DH
,
Sorg
 
JM
,
Khakpour
 
H
,
Krokhaleva
 
Y
, et al.  
Arrhythmic risk profile and outcomes of patients undergoing cardiac sympathetic denervation for recurrent monomorphic ventricular tachycardia after ablation
.
J Am Heart Assoc
 
2021
;
10
:
e018371
. https://doi.org/10.1161/JAHA.120.018371

520

Krug
 
D
,
Blanck
 
O
,
Andratschke
 
N
,
Guckenberger
 
M
,
Jumeau
 
R
,
Mehrhof
 
F
, et al.  
Recommendations regarding cardiac stereotactic body radiotherapy for treatment refractory ventricular tachycardia
.
Heart Rhythm
 
2021
;
18
:
2137
2145
. https://doi.org/10.1016/j.hrthm.2021.08.004

521

Lin
 
G
,
Nishimura
 
RA
,
Gersh
 
BJ
,
Phil
 
D
,
Ommen
 
SR
,
Ackerman
 
MJ
, et al.  
Device complications and inappropriate implantable cardioverter defibrillator shocks in patients with hypertrophic cardiomyopathy
.
Heart
 
2009
;
95
:
709
714
. https://doi.org/10.1136/hrt.2008.150656

522

Orgeron
 
GM
,
James
 
CA
,
Te Riele
 
A
,
Tichnell
 
C
,
Murray
 
B
,
Bhonsale
 
A
, et al.  
Implantable cardioverter-defibrillator therapy in arrhythmogenic right ventricular dysplasia/cardiomyopathy: predictors of appropriate therapy, outcomes, and complications
.
J Am Heart Assoc
 
2017
;
6
:
e006242
. https://doi.org/10.1161/JAHA.117.006242

523

Corrado
 
D
,
Calkins
 
H
,
Link
 
MS
,
Leoni
 
L
,
Favale
 
S
,
Bevilacqua
 
M
, et al.  
Prophylactic implantable defibrillator in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia and no prior ventricular fibrillation or sustained ventricular tachycardia
.
Circulation
 
2010
;
122
:
1144
1152
. https://doi.org/10.1161/CIRCULATIONAHA.109.913871

524

Protonotarios
 
A
,
Bariani
 
R
,
Cappelletto
 
C
,
Pavlou
 
M
,
Garcia-Garcia
 
A
,
Cipriani
 
A
, et al.  
Importance of genotype for risk stratification in arrhythmogenic right ventricular cardiomyopathy using the 2019 ARVC risk calculator
.
Eur Heart J
 
2022
;
43
:
3053
3067
. https://doi.org/10.1093/eurheartj/ehac235

525

O’Mahony
 
C
,
Jichi
 
F
,
Pavlou
 
M
,
Monserrat
 
L
,
Anastasakis
 
A
,
Rapezzi
 
C
, et al.  
Hypertrophic cardiomyopathy outcomes I. A novel clinical risk prediction model for sudden cardiac death in hypertrophic cardiomyopathy (HCM risk-SCD)
.
Eur Heart J
 
2014
;
35
:
2010
2020
. https://doi.org/10.1093/eurheartj/eht439

526

Jorda
 
P
,
Bosman
 
LP
,
Gasperetti
 
A
,
Mazzanti
 
A
,
Gourraud
 
JB
,
Davies
 
B
, et al.  
Arrhythmic risk prediction in arrhythmogenic right ventricular cardiomyopathy: external validation of the arrhythmogenic right ventricular cardiomyopathy risk calculator
.
Eur Heart J
 
2022
;
43
:
3041
3052
. https://doi.org/10.1093/eurheartj/ehac289

527

Masri
 
A
,
Altibi
 
AM
,
Erqou
 
S
,
Zmaili
 
MA
,
Saleh
 
A
,
Al-Adham
 
R
, et al.  
Wearable cardioverter-defibrillator therapy for the prevention of sudden cardiac death: a systematic review and meta-analysis
.
JACC Clin Electrophysiol
 
2019
;
5
:
152
161
. https://doi.org/10.1016/j.jacep.2018.11.011

528

Connolly
 
SJ
,
Gent
 
M
,
Roberts
 
RS
,
Dorian
 
P
,
Roy
 
D
,
Sheldon
 
RS
, et al.  
Canadian implantable defibrillator study (CIDS): a randomized trial of the implantable cardioverter defibrillator against amiodarone
.
Circulation
 
2000
;
101
:
1297
1302
. https://doi.org/10.1161/01.CIR.101.11.1297

529

Antiarrhythmics versus Implantable Defibrillators Investigators
.
A comparison of antiarrhythmic-drug therapy with implantable defibrillators in patients resuscitated from near-fatal ventricular arrhythmias
.
N Engl J Med
 
1997
;
337
:
1576
1583
. https://doi.org/10.1056/NEJM199711273372202

530

Kuck
 
KH
,
Cappato
 
R
,
Siebels
 
J
,
Ruppel
 
R
.
Randomized comparison of antiarrhythmic drug therapy with implantable defibrillators in patients resuscitated from cardiac arrest: the Cardiac Arrest Study Hamburg (CASH)
.
Circulation
 
2000
;
102
:
748
754
. https://doi.org/10.1161/01.CIR.102.7.748

531

Connolly
 
SJ
,
Hallstrom
 
AP
,
Cappato
 
R
,
Schron
 
EB
,
Kuck
 
KH
,
Zipes
 
DP
, et al.  
Meta-analysis of the implantable cardioverter defibrillator secondary prevention trials
.
Eur Heart J
 
2000
;
21
:
2071
2078
. https://doi.org/10.1053/euhj.2000.2476

532

Elliott
 
PM
,
Sharma
 
S
,
Varnava
 
A
,
Poloniecki
 
J
,
Rowland
 
E
,
McKenna
 
WJ
.
Survival after cardiac arrest or sustained ventricular tachycardia in patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1999
;
33
:
1596
1601
. https://doi.org/10.1016/S0735-1097(99)00056-X

533

Cleland
 
JG
,
Daubert
 
JC
,
Erdmann
 
E
,
Freemantle
 
N
,
Gras
 
D
,
Kappenberger
 
L
, et al.  
The effect of cardiac resynchronization on morbidity and mortality in heart failure
.
N Engl J Med
 
2005
;
352
:
1539
1549
. https://doi.org/10.1056/NEJMoa050496

534

Cecchi
 
F
,
Maron
 
BJ
,
Epstein
 
SE
.
Long-term outcome of patients with hypertrophic cardiomyopathy successfully resuscitated after cardiac arrest
.
J Am Coll Cardiol
 
1989
;
13
:
1283
1288
. https://doi.org/10.1016/0735-1097(89)90302-1

535

Miron
 
A
,
Lafreniere-Roula
 
M
,
Steve Fan
 
CP
,
Armstrong
 
KR
,
Dragulescu
 
A
,
Papaz
 
T
, et al.  
A validated model for sudden cardiac death risk prediction in pediatric hypertrophic cardiomyopathy
.
Circulation
 
2020
;
142
:
217
229
. https://doi.org/10.1161/CIRCULATIONAHA.120.047235

536

Aquaro
 
GD
,
De Luca
 
A
,
Cappelletto
 
C
,
Raimondi
 
F
,
Bianco
 
F
,
Botto
 
N
, et al.  
Comparison of different prediction models for the indication of implanted cardioverter defibrillator in patients with arrhythmogenic right ventricular cardiomyopathy
.
ESC Heart Fail
 
2020
;
7
:
4080
4088
. https://doi.org/10.1002/ehf2.13019

537

Baudinaud
 
P
,
Laredo
 
M
,
Badenco
 
N
,
Rouanet
 
S
,
Waintraub
 
X
,
Duthoit
 
G
, et al.  
External validation of a risk prediction model for ventricular arrhythmias in arrhythmogenic right ventricular cardiomyopathy
.
Can J Cardiol
 
2021
;
37
:
1263
1266
. https://doi.org/10.1016/j.cjca.2021.02.018

538

Bosman
 
LP
,
Nielsen Gerlach
 
CL
,
Cadrin-Tourigny
 
J
,
Orgeron
 
G
,
Tichnell
 
C
,
Murray
 
B
, et al.  
Comparing clinical performance of current implantable cardioverter-defibrillator implantation recommendations in arrhythmogenic right ventricular cardiomyopathy
.
Europace
 
2022
;
24
:
296
305
. https://doi.org/10.1093/europace/euab162

539

Cadrin-Tourigny
 
J
,
Bosman
 
LP
,
Nozza
 
A
,
Wang
 
W
,
Tadros
 
R
,
Bhonsale
 
A
, et al.  
A new prediction model for ventricular arrhythmias in arrhythmogenic right ventricular cardiomyopathy
.
Eur Heart J
 
2022
;
43
:
e1
e9
. https://doi.org/10.1093/eurheartj/ehac180

540

Kayvanpour
 
E
,
Sammani
 
A
,
Sedaghat-Hamedani
 
F
,
Lehmann
 
DH
,
Broezel
 
A
,
Koelemenoglu
 
J
, et al.  
A novel risk model for predicting potentially life-threatening arrhythmias in non-ischemic dilated cardiomyopathy (DCM-SVA risk)
.
Int J Cardiol
 
2021
;
339
:
75
82
. https://doi.org/10.1016/j.ijcard.2021.07.002

541

Wahbi
 
K
,
Ben Yaou
 
R
,
Gandjbakhch
 
E
,
Anselme
 
F
,
Gossios
 
T
,
Lakdawala
 
NK
, et al.  
Development and validation of a new risk prediction score for life-threatening ventricular tachyarrhythmias in laminopathies
.
Circulation
 
2019
;
140
:
293
302
. https://doi.org/10.1161/CIRCULATIONAHA.118.039410

542

Verstraelen
 
TE
,
van Lint
 
FHM
,
Bosman
 
LP
,
de Brouwer
 
R
,
Proost
 
VM
,
Abeln
 
BGS
, et al.  
Prediction of ventricular arrhythmia in phospholamban p.Arg14del mutation carriers-reaching the frontiers of individual risk prediction
.
Eur Heart J
 
2021
;
42
:
2842
2850
. https://doi.org/10.1093/eurheartj/ehab294

543

Knops
 
RE
,
Olde Nordkamp
 
LRA
,
Delnoy
 
PHM
,
Boersma
 
LVA
,
Kuschyk
 
J
,
El-Chami
 
MF
, et al.  
Subcutaneous or transvenous defibrillator therapy
.
N Engl J Med
 
2020
;
383
:
526
536
. https://doi.org/10.1056/NEJMoa1915932

544

Cardim
 
N
,
Brito
 
D
,
Rocha Lopes
 
L
,
Freitas
 
A
,
Araujo
 
C
,
Belo
 
A
, et al.  
The Portuguese registry of hypertrophic cardiomyopathy: overall results
.
Rev Port Cardiol (Engl Ed)
 
2018
;
37
:
1
10
. https://doi.org/10.1016/j.repc.2017.08.005

545

McKenna
 
WJ
,
Judge
 
DP
.
Epidemiology of the inherited cardiomyopathies
.
Nat Rev Cardiol
 
2021
;
18
:
22
36
. https://doi.org/10.1038/s41569-020-0428-2

546

Pelliccia
 
F
,
Limongelli
 
G
,
Autore
 
C
,
Gimeno-Blanes
 
JR
,
Basso
 
C
,
Elliott
 
P
.
Sex-related differences in cardiomyopathies
.
Int J Cardiol
 
2019
;
286
:
239
243
. https://doi.org/10.1016/j.ijcard.2018.10.091

547

Perez-Sanchez
 
I
,
Romero-Puche
 
AJ
,
Garcia-Molina Saez
 
E
,
Sabater-Molina
 
M
,
Lopez-Ayala
 
JM
,
Munoz-Esparza
 
C
, et al.  
Factors influencing the phenotypic expression of hypertrophic cardiomyopathy in genetic carriers
.
Rev Esp Cardiol (Engl Ed)
 
2018
;
71
:
146
154
. https://doi.org/10.1016/j.rec.2017.06.002

548

Argiro
 
A
,
Ho
 
C
,
Day
 
SM
,
van der Velden
 
J
,
Cerbai
 
E
,
Saberi
 
S
, et al.  
Sex-related differences in genetic cardiomyopathies
.
J Am Heart Assoc
 
2022
;
11
:
e024947
. https://doi.org/10.1161/JAHA.121.024947

549

Shah
 
RA
,
Asatryan
 
B
,
Sharaf Dabbagh
 
G
,
Aung
 
N
,
Khanji
 
MY
,
Lopes
 
LR
, et al.  
Genotype-first approach I. Frequency, penetrance, and variable expressivity of dilated cardiomyopathy-associated putative pathogenic gene variants in UK Biobank participants
.
Circulation
 
2022
;
146
:
110
124
. https://doi.org/10.1161/CIRCULATIONAHA.121.058143

550

de Marvao
 
A
,
McGurk
 
KA
,
Zheng
 
SL
,
Thanaj
 
M
,
Bai
 
W
,
Duan
 
J
, et al.  
Phenotypic expression and outcomes in individuals with rare genetic variants of hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2021
;
78
:
1097
1110
. https://doi.org/10.1016/j.jacc.2021.07.017

551

McGurk
 
KA
,
Zheng
 
SL
,
Henry
 
A
,
Josephs
 
K
,
Edwards
 
M
,
de Marvao
 
A
, et al.  
Correspondence on “ACMG SF v3.0 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics (ACMG)” by Miller et al
.
Genet Med
 
2022
;
24
:
744
746
. https://doi.org/10.1016/j.gim.2021.10.020

552

Maron
 
BJ
,
Casey
 
SA
,
Olivotto
 
I
,
Sherrid
 
MV
,
Semsarian
 
C
,
Autore
 
C
, et al.  
Clinical course and quality of life in high-risk patients with hypertrophic cardiomyopathy and implantable cardioverter-defibrillators
.
Circ Arrhythm Electrophysiol
 
2018
;
11
:
e005820
. https://doi.org/10.1161/CIRCEP.117.005820

553

Ingles
 
J
,
Sarina
 
T
,
Kasparian
 
N
,
Semsarian
 
C
.
Psychological wellbeing and posttraumatic stress associated with implantable cardioverter defibrillator therapy in young adults with genetic heart disease
.
Int J Cardiol
 
2013
;
168
:
3779
3784
. https://doi.org/10.1016/j.ijcard.2013.06.006

554

James
 
CA
,
Tichnell
 
C
,
Murray
 
B
,
Daly
 
A
,
Sears
 
SF
,
Calkins
 
H
.
General and disease-specific psychosocial adjustment in patients with arrhythmogenic right ventricular dysplasia/cardiomyopathy with implantable cardioverter defibrillators: a large cohort study
.
Circ Cardiovasc Genet
 
2012
;
5
:
18
24
. https://doi.org/10.1161/CIRCGENETICS.111.960898

555

Rhodes
 
AC
,
Murray
 
B
,
Tichnell
 
C
,
James
 
CA
,
Calkins
 
H
,
Sears
 
SF
.
Quality of life metrics in arrhythmogenic right ventricular cardiomyopathy patients: the impact of age, shock and sex
.
Int J Cardiol
 
2017
;
248
:
216
220
. https://doi.org/10.1016/j.ijcard.2017.08.026

556

Sweeting
 
J
,
Ball
 
K
,
McGaughran
 
J
,
Atherton
 
J
,
Semsarian
 
C
,
Ingles
 
J
.
Impact of the implantable cardioverter defibrillator on confidence to undertake physical activity in inherited heart disease: a cross-sectional study
.
Eur J Cardiovasc Nurs
 
2017
;
16
:
742
752
. https://doi.org/10.1177/1474515117715760

557

Sears
 
SF
 Jr,
Conti
 
JB
.
Quality of life and psychological functioning of icd patients
.
Heart
 
2002
;
87
:
488
493
. https://doi.org/10.1136/heart.87.5.488

558

Ingles
 
J
,
Spinks
 
C
,
Yeates
 
L
,
McGeechan
 
K
,
Kasparian
 
N
,
Semsarian
 
C
.
Posttraumatic stress and prolonged grief after the sudden cardiac death of a young relative
.
JAMA Intern Med
 
2016
;
176
:
402
405
. https://doi.org/10.1001/jamainternmed.2015.7808

559

Stiles
 
MK
,
Wilde
 
AAM
,
Abrams
 
DJ
,
Ackerman
 
MJ
,
Albert
 
CM
,
Behr
 
ER
, et al.  
2020 APHRS/HRS expert consensus statement on the investigation of decedents with sudden unexplained death and patients with sudden cardiac arrest, and of their families
.
Heart Rhythm
 
2021
;
18
:
e1
e50
. https://doi.org/10.1016/j.hrthm.2020.10.010

560

van den Heuvel
 
LM
,
Sarina
 
T
,
Sweeting
 
J
,
Yeates
 
L
,
Bates
 
K
,
Spinks
 
C
, et al.  
A prospective longitudinal study of health-related quality of life and psychological wellbeing after an implantable cardioverter-defibrillator in patients with genetic heart diseases
.
Heart Rhythm O2
 
2022
;
3
:
143
151
. https://doi.org/10.1016/j.hroo.2022.02.003

561

Passman
 
R
,
Subacius
 
H
,
Ruo
 
B
,
Schaechter
 
A
,
Howard
 
A
,
Sears
 
SF
, et al.  
Implantable cardioverter defibrillators and quality of life: results from the defibrillators in nonischemic cardiomyopathy treatment evaluation study
.
Arch Intern Med
 
2007
;
167
:
2226
2232
. https://doi.org/10.1001/archinte.167.20.2226

562

Ni
 
SQ
,
Ni
 
J
,
Yang
 
N
,
Wang
 
J
.
Effect of magnetic nanoparticles on the performance of activated sludge treatment system
.
Bioresour Technol
 
2013
;
143
:
555
561
. https://doi.org/10.1016/j.biortech.2013.06.041

563

von Kanel
 
R
,
Baumert
 
J
,
Kolb
 
C
,
Cho
 
E-Y
,
Ladwig
 
K-H
.
Chronic posttraumatic stress and its predictors in patients living with an implantable cardioverter defibrillator
.
J Affect Disord
 
2011
;
131
:
344
352
. https://doi.org/10.1016/j.jad.2010.12.002

564

Lewis
 
KB
,
Carroll
 
SL
,
Birnie
 
D
,
Stacey
 
D
,
Matlock
 
DD
.
Incorporating patients’ preference diagnosis in implantable cardioverter defibrillator decision-making: a review of recent literature
.
Curr Opin Cardiol
 
2018
;
33
:
42
49
. https://doi.org/10.1097/HCO.0000000000000464

565

Luiten
 
RC
,
Ormond
 
K
,
Post
 
L
,
Asif
 
IM
,
Wheeler
 
MT
,
Caleshu
 
C
.
Exercise restrictions trigger psychological difficulty in active and athletic adults with hypertrophic cardiomyopathy
.
Open Heart
 
2016
;
3
:
e000488
. https://doi.org/10.1136/openhrt-2016-000488

566

Subas
 
T
,
Luiten
 
R
,
Hanson-Kahn
 
A
,
Wheeler
 
M
,
Caleshu
 
C
.
Evolving decisions: perspectives of active and athletic individuals with inherited heart disease who exercise against recommendations
.
J Genet Couns
 
2019
;
28
:
119
129
. https://doi.org/10.1007/s10897-018-0297-6

567

Alpert
 
C
,
Day
 
SM
,
Saberi
 
S
.
Sports and exercise in athletes with hypertrophic cardiomyopathy
.
Clin Sports Med
 
2015
;
34
:
489
505
. https://doi.org/10.1016/j.csm.2015.03.005

568

Day
 
SM
.
Exercise in hypertrophic cardiomyopathy
.
J Cardiovasc Transl Res
 
2009
;
2
:
407
414
. https://doi.org/10.1007/s12265-009-9134-5

569

Simon
 
NM
.
Treating complicated grief
.
JAMA
 
2013
;
310
:
416
423
. https://doi.org/10.1001/jama.2013.8614

570

McDonald
 
K
,
Sharpe
 
L
,
Yeates
 
L
,
Semsarian
 
C
,
Ingles
 
J
.
Needs analysis of parents following sudden cardiac death in the young
.
Open Heart
 
2020
;
7
:
e001120
. https://doi.org/10.1136/openhrt-2019-001120

571

Wisten
 
A
,
Zingmark
 
K
.
Supportive needs of parents confronted with sudden cardiac death–a qualitative study
.
Resuscitation
 
2007
;
74
:
68
74
. https://doi.org/10.1016/j.resuscitation.2006.11.014

572

O’Donovan
 
CE
,
Waddell-Smith
 
KE
,
Skinner
 
JR
,
Broadbent
 
E
.
Predictors of β-blocker adherence in cardiac inherited disease
.
Open Heart
 
2018
;
5
:
e000877
. https://doi.org/10.1136/openhrt-2018-000877

573

Cupples
 
S
,
Dew
 
MA
,
Grady
 
KL
,
De Geest
 
S
,
Dobbels
 
F
,
Lanuza
 
D
, et al.  
Report of the Psychosocial Outcomes Workgroup of the Nursing and Social Sciences Council of the International Society for Heart and Lung Transplantation: present status of research on psychosocial outcomes in cardiothoracic transplantation: review and recommendations for the field
.
J Heart Lung Transplant
 
2006
;
25
:
716
725
. https://doi.org/10.1016/j.healun.2006.02.005

574

Aatre
 
RD
,
Day
 
SM
.
Psychological issues in genetic testing for inherited cardiovascular diseases
.
Circ Cardiovasc Genet
 
2011
;
4
:
81
90
. https://doi.org/10.1161/CIRCGENETICS.110.957365

575

Burns
 
C
,
James
 
C
,
Ingles
 
J
.
Communication of genetic information to families with inherited rhythm disorders
.
Heart Rhythm
 
2018
;
15
:
780
786
. https://doi.org/10.1016/j.hrthm.2017.11.024

576

Yeates
 
L
,
Hunt
 
L
,
Saleh
 
M
,
Semsarian
 
C
,
Ingles
 
J
.
Poor psychological wellbeing particularly in mothers following sudden cardiac death in the young
.
Eur J Cardiovasc Nurs
 
2013
;
12
:
484
491
. https://doi.org/10.1177/1474515113485510

577

Karam
 
N
,
Jabre
 
P
,
Narayanan
 
K
,
Sharifzadehgan
 
A
,
Perier
 
MC
,
Tennenbaum
 
J
, et al.  
Psychological support and medical screening of first-degree relatives of sudden cardiac arrest victims
.
JACC Clin Electrophysiol
 
2020
;
6
:
586
587
. https://doi.org/10.1016/j.jacep.2020.02.002

578

Kampmann
 
C
,
Wiethoff
 
CM
,
Wenzel
 
A
,
Stolz
 
G
,
Betancor
 
M
,
Wippermann
 
CF
, et al.  
Normal values of M mode echocardiographic measurements of more than 2000 healthy infants and children in central Europe
.
Heart
 
2000
;
83
:
667
672
. https://doi.org/10.1136/heart.83.6.667

579

Cardim
 
N
,
Galderisi
 
M
,
Edvardsen
 
T
,
Plein
 
S
,
Popescu
 
BA
,
D’Andrea
 
A
, et al.  
Role of multimodality cardiac imaging in the management of patients with hypertrophic cardiomyopathy: an expert consensus of the European Association of Cardiovascular Imaging Endorsed by the Saudi Heart Association
.
Eur Heart J Cardiovasc Imaging
 
2015
;
16
:
280
. https://doi.org/10.1093/ehjci/jeu291

580

Maron
 
MS
,
Finley
 
JJ
,
Bos
 
JM
,
Hauser
 
TH
,
Manning
 
WJ
,
Haas
 
TS
, et al.  
Prevalence, clinical significance, and natural history of left ventricular apical aneurysms in hypertrophic cardiomyopathy
.
Circulation
 
2008
;
118
:
1541
1549
. https://doi.org/10.1161/CIRCULATIONAHA.108.781401

581

Weinsaft
 
JW
,
Kim
 
HW
,
Crowley
 
AL
,
Klem
 
I
,
Shenoy
 
C
,
Van Assche
 
L
, et al.  
LV thrombus detection by routine echocardiography: insights into performance characteristics using delayed enhancement CMR
.
JACC Cardiovasc Imaging
 
2011
;
4
:
702
712
. https://doi.org/10.1016/j.jcmg.2011.03.017

582

Brouwer
 
WP
,
Germans
 
T
,
Head
 
MC
,
van der Velden
 
J
,
Heymans
 
MW
,
Christiaans
 
I
, et al.  
Multiple myocardial crypts on modified long-axis view are a specific finding in pre-hypertrophic HCM mutation carriers
.
Eur Heart J Cardiovasc Imaging
 
2012
;
13
:
292
297
. https://doi.org/10.1093/ehjci/jes005

583

Maron
 
MS
,
Rowin
 
EJ
,
Lin
 
D
,
Appelbaum
 
E
,
Chan
 
RH
,
Gibson
 
CM
, et al.  
Prevalence and clinical profile of myocardial crypts in hypertrophic cardiomyopathy
.
Circ Cardiovasc Imaging
 
2012
;
5
:
441
447
. https://doi.org/10.1161/CIRCIMAGING.112.972760

584

Spirito
 
P
,
Autore
 
C
,
Rapezzi
 
C
,
Bernabo
 
P
,
Badagliacca
 
R
,
Maron
 
MS
, et al.  
Syncope and risk of sudden death in hypertrophic cardiomyopathy
.
Circulation
 
2009
;
119
:
1703
1710
. https://doi.org/10.1161/CIRCULATIONAHA.108.798314

585

Vogelsberg
 
H
,
Mahrholdt
 
H
,
Deluigi
 
CC
,
Yilmaz
 
A
,
Kispert
 
EM
,
Greulich
 
S
, et al.  
Cardiovascular magnetic resonance in clinically suspected cardiac amyloidosis: noninvasive imaging compared to endomyocardial biopsy
.
J Am Coll Cardiol
 
2008
;
51
:
1022
1030
. https://doi.org/10.1016/j.jacc.2007.10.049

586

Syed
 
IS
,
Glockner
 
JF
,
Feng
 
D
,
Araoz
 
PA
,
Martinez
 
MW
,
Edwards
 
WD
, et al.  
Role of cardiac magnetic resonance imaging in the detection of cardiac amyloidosis
.
JACC Cardiovasc Imaging
 
2010
;
3
:
155
164
. https://doi.org/10.1016/j.jcmg.2009.09.023

587

Wigle
 
ED
,
Sasson
 
Z
,
Henderson
 
MA
,
Ruddy
 
TD
,
Fulop
 
J
,
Rakowski
 
H
, et al.  
Hypertrophic cardiomyopathy. The importance of the site and the extent of hypertrophy. A review
.
Prog Cardiovasc Dis
 
1985
;
28
:
1
83
. https://doi.org/10.1016/0033-0620(85)90024-6

588

Dimitrow
 
PP
,
Bober
 
M
,
Michalowska
 
J
,
Sorysz
 
D
.
Left ventricular outflow tract gradient provoked by upright position or exercise in treated patients with hypertrophic cardiomyopathy without obstruction at rest
.
Echocardiography
 
2009
;
26
:
513
520
. https://doi.org/10.1111/j.1540-8175.2008.00851.x

589

Maron
 
BJ
,
Gottdiener
 
JS
,
Bonow
 
RO
,
Epstein
 
SE
.
Hypertrophic cardiomyopathy with unusual locations of left ventricular hypertrophy undetectable by M-mode echocardiography. Identification by wide-angle two-dimensional echocardiography
.
Circulation
 
1981
;
63
:
409
418
. https://doi.org/10.1161/01.CIR.63.2.409

590

Elliott
 
P
,
Gimeno
 
J
,
Tome
 
M
,
McKenna
 
W
.
Left ventricular outflow tract obstruction and sudden death in hypertrophic cardiomyopathy
.
Eur Heart J
 
2006
;
27
:
3073
; author reply 3073-4
. https://doi.org/10.1093/eurheartj/ehl383

591

Wigle
 
ED
,
Rakowski
 
H
,
Kimball
 
BP
,
Williams
 
WG
.
Hypertrophic cardiomyopathy. Clinical spectrum and treatment
.
Circulation
 
1995
;
92
:
1680
1692
. https://doi.org/10.1161/01.CIR.92.7.1680

592

Spirito
 
P
,
Bellone
 
P
,
Harris
 
KM
,
Bernabo
 
P
,
Bruzzi
 
P
,
Maron
 
BJ
.
Magnitude of left ventricular hypertrophy and risk of sudden death in hypertrophic cardiomyopathy
.
N Engl J Med
 
2000
;
342
:
1778
1785
. https://doi.org/10.1056/NEJM200006153422403

593

Elliott
 
PM
,
Gimeno Blanes
 
JR
,
Mahon
 
NG
,
Poloniecki
 
JD
,
McKenna
 
WJ
.
Relation between severity of left-ventricular hypertrophy and prognosis in patients with hypertrophic cardiomyopathy
.
Lancet
 
2001
;
357
:
420
424
. https://doi.org/10.1016/S0140-6736(00)04005-8

594

Kumar
 
S
,
Van Ness
 
G
,
Bender
 
A
,
Yadava
 
M
,
Minnier
 
J
,
Ravi
 
S
, et al.  
Standardized goal-directed valsalva maneuver for assessment of inducible left ventricular outflow tract obstruction in hypertrophic cardiomyopathy
.
J Am Soc Echocardiogr
 
2018
;
31
:
791
798
. https://doi.org/10.1016/j.echo.2018.01.022

595

Maron
 
MS
,
Olivotto
 
I
,
Zenovich
 
AG
,
Link
 
MS
,
Pandian
 
NG
,
Kuvin
 
JT
, et al.  
Hypertrophic cardiomyopathy is predominantly a disease of left ventricular outflow tract obstruction
.
Circulation
 
2006
;
114
:
2232
2239
. https://doi.org/10.1161/CIRCULATIONAHA.106.644682

596

Shah
 
JS
,
Esteban
 
MT
,
Thaman
 
R
,
Sharma
 
R
,
Mist
 
B
,
Pantazis
 
A
, et al.  
Prevalence of exercise-induced left ventricular outflow tract obstruction in symptomatic patients with non-obstructive hypertrophic cardiomyopathy
.
Heart
 
2008
;
94
:
1288
1294
. https://doi.org/10.1136/hrt.2007.126003

597

Marwick
 
TH
,
Nakatani
 
S
,
Haluska
 
B
,
Thomas
 
JD
,
Lever
 
HM
.
Provocation of latent left ventricular outflow tract gradients with amyl nitrite and exercise in hypertrophic cardiomyopathy
.
Am J Cardiol
 
1995
;
75
:
805
809
. https://doi.org/10.1016/S0002-9149(99)80416-0

598

Reant
 
P
,
Dufour
 
M
,
Peyrou
 
J
,
Reynaud
 
A
,
Rooryck
 
C
,
Dijos
 
M
, et al.  
Upright treadmill vs. semi-supine bicycle exercise echocardiography to provoke obstruction in symptomatic hypertrophic cardiomyopathy: a pilot study
.
Eur Heart J Cardiovasc Imaging
 
2018
;
19
:
31
38
. https://doi.org/10.1093/ehjci/jew313

599

Yu
 
EH
,
Omran
 
AS
,
Wigle
 
ED
,
Williams
 
WG
,
Siu
 
SC
,
Rakowski
 
H
.
Mitral regurgitation in hypertrophic obstructive cardiomyopathy: relationship to obstruction and relief with myectomy
.
J Am Coll Cardiol
 
2000
;
36
:
2219
2225
. https://doi.org/10.1016/S0735-1097(00)01019-6

600

Oki
 
T
,
Fukuda
 
N
,
Iuchi
 
A
,
Tabata
 
T
,
Tanimoto
 
M
,
Manabe
 
K
, et al.  
Transesophageal echocardiographic evaluation of mitral regurgitation in hypertrophic cardiomyopathy: contributions of eccentric left ventricular hypertrophy and related abnormalities of the mitral complex
.
J Am Soc Echocardiogr
 
1995
;
8
:
503
510
. https://doi.org/10.1016/S0894-7317(05)80338-4

601

Grigg
 
LE
,
Wigle
 
ED
,
Williams
 
WG
,
Daniel
 
LB
,
Rakowski
 
H
.
Transesophageal Doppler echocardiography in obstructive hypertrophic cardiomyopathy: clarification of pathophysiology and importance in intraoperative decision making
.
J Am Coll Cardiol
 
1992
;
20
:
42
52
. https://doi.org/10.1016/0735-1097(92)90135-A

602

Marwick
 
TH
,
Stewart
 
WJ
,
Lever
 
HM
,
Lytle
 
BW
,
Rosenkranz
 
ER
,
Duffy
 
CI
, et al.  
Benefits of intraoperative echocardiography in the surgical management of hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1992
;
20
:
1066
1072
. https://doi.org/10.1016/0735-1097(92)90359-U

603

Geske
 
JB
,
Cullen
 
MW
,
Sorajja
 
P
,
Ommen
 
SR
,
Nishimura
 
RA
.
Assessment of left ventricular outflow gradient: hypertrophic cardiomyopathy versus aortic valvular stenosis
.
JACC Cardiovasc Interv
 
2012
;
5
:
675
681
. https://doi.org/10.1016/j.jcin.2012.01.026

604

Rudolph
 
A
,
Abdel-Aty
 
H
,
Bohl
 
S
,
Boye
 
P
,
Zagrosek
 
A
,
Dietz
 
R
, et al.  
Noninvasive detection of fibrosis applying contrast-enhanced cardiac magnetic resonance in different forms of left ventricular hypertrophy relation to remodeling
.
J Am Coll Cardiol
 
2009
;
53
:
284
291
. https://doi.org/10.1016/j.jacc.2008.08.064

605

Moon
 
JCC
,
Reed
 
E
,
Sheppard
 
MN
,
Elkington
 
AG
,
Ho
 
SY
,
Burke
 
M
, et al.  
The histologic basis of late gadolinium enhancement cardiovascular magnetic resonance in hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2004
;
43
:
2260
2264
. https://doi.org/10.1016/j.jacc.2004.03.035

606

Kwon
 
DH
,
Smedira
 
NG
,
Rodriguez
 
ER
,
Tan
 
C
,
Setser
 
R
,
Thamilarasan
 
M
, et al.  
Cardiac magnetic resonance detection of myocardial scarring in hypertrophic cardiomyopathy: correlation with histopathology and prevalence of ventricular tachycardia
.
J Am Coll Cardiol
 
2009
;
54
:
242
249
. https://doi.org/10.1016/j.jacc.2009.04.026

607

White
 
RD
,
Obuchowski
 
NA
,
Gunawardena
 
S
,
Lipchik
 
EO
,
Lever
 
HM
,
Van Dyke
 
CW
, et al.  
Left ventricular outflow tract obstruction in hypertrophic cardiomyopathy: presurgical and postsurgical evaluation by computed tomography magnetic resonance imaging
.
Am J Card Imaging
 
1996
;
10
:
1
13
.

608

Richard
 
P
,
Charron
 
P
,
Carrier
 
L
,
Ledeuil
 
C
,
Cheav
 
T
,
Pichereau
 
C
, et al.  
Hypertrophic cardiomyopathy: distribution of disease genes, spectrum of mutations, and implications for a molecular diagnosis strategy
.
Circulation
 
2003
;
107
:
2227
2232
. https://doi.org/10.1161/01.CIR.0000066323.15244.54

609

Murphy
 
SL
,
Anderson
 
JH
,
Kapplinger
 
JD
,
Kruisselbrink
 
TM
,
Gersh
 
BJ
,
Ommen
 
SR
, et al.  
Evaluation of the Mayo Clinic phenotype-based genotype predictor score in patients with clinically diagnosed hypertrophic cardiomyopathy
.
J Cardiovasc Transl Res
 
2016
;
9
:
153
161
. https://doi.org/10.1007/s12265-016-9681-5

610

Gruner
 
C
,
Ivanov
 
J
,
Care
 
M
,
Williams
 
L
,
Moravsky
 
G
,
Yang
 
H
, et al.  
Toronto hypertrophic cardiomyopathy genotype score for prediction of a positive genotype in hypertrophic cardiomyopathy
.
Circ Cardiovasc Genet
 
2013
;
6
:
19
26
. https://doi.org/10.1161/CIRCGENETICS.112.963363

611

Ingles
 
J
,
Burns
 
C
,
Bagnall
 
RD
,
Lam
 
L
,
Yeates
 
L
,
Sarina
 
T
, et al.  
Nonfamilial hypertrophic cardiomyopathy: prevalence, natural history, and clinical implications
.
Circ Cardiovasc Genet
 
2017
;
10
:
e001620
. https://doi.org/10.1161/CIRCGENETICS.116.001620

612

Ko
 
C
,
Arscott
 
P
,
Concannon
 
M
,
Saberi
 
S
,
Day
 
SM
,
Yashar
 
BM
, et al.  
Genetic testing impacts the utility of prospective familial screening in hypertrophic cardiomyopathy through identification of a nonfamilial subgroup
.
Genet Med
 
2018
;
20
:
69
75
. https://doi.org/10.1038/gim.2017.79

613

Anan
 
R
,
Shono
 
H
,
Kisanuki
 
A
,
Arima
 
S
,
Nakao
 
S
,
Tanaka
 
H
.
Patients with familial hypertrophic cardiomyopathy caused by a Phe110Ile missense mutation in the cardiac troponin T gene have variable cardiac morphologies and a favorable prognosis
.
Circulation
 
1998
;
98
:
391
397
. https://doi.org/10.1161/01.CIR.98.5.391

614

Elliott
 
P
,
Baker
 
R
,
Pasquale
 
F
,
Quarta
 
G
,
Ebrahim
 
H
,
Mehta
 
AB
, et al.  
Prevalence of Anderson–Fabry disease in patients with hypertrophic cardiomyopathy: the European Anderson–Fabry Disease survey
.
Heart
 
2011
;
97
:
1957
1960
. https://doi.org/10.1136/heartjnl-2011-300364

615

Nagueh
 
SF
,
Appleton
 
CP
,
Gillebert
 
TC
,
Marino
 
PN
,
Oh
 
JK
,
Smiseth
 
OA
, et al.  
Recommendations for the evaluation of left ventricular diastolic function by echocardiography
.
Eur J Echocardiogr
 
2009
;
10
:
165
193
. https://doi.org/10.1093/ejechocard/jep007

616

Kubo
 
T
,
Gimeno
 
JR
,
Bahl
 
A
,
Steffensen
 
U
,
Steffensen
 
M
,
Osman
 
E
, et al.  
Prevalence, clinical significance, and genetic basis of hypertrophic cardiomyopathy with restrictive phenotype
.
J Am Coll Cardiol
 
2007
;
49
:
2419
2426
. https://doi.org/10.1016/j.jacc.2007.02.061

617

Biagini
 
E
,
Spirito
 
P
,
Rocchi
 
G
,
Ferlito
 
M
,
Rosmini
 
S
,
Lai
 
F
, et al.  
Prognostic implications of the Doppler restrictive filling pattern in hypertrophic cardiomyopathy
.
Am J Cardiol
 
2009
;
104
:
1727
1731
. https://doi.org/10.1016/j.amjcard.2009.07.057

618

Geske
 
JB
,
Sorajja
 
P
,
Nishimura
 
RA
,
Ommen
 
SR
.
Evaluation of left ventricular filling pressures by Doppler echocardiography in patients with hypertrophic cardiomyopathy: correlation with direct left atrial pressure measurement at cardiac catheterization
.
Circulation
 
2007
;
116
:
2702
2708
. https://doi.org/10.1161/CIRCULATIONAHA.107.698985

619

Kitaoka
 
H
,
Kubo
 
T
,
Okawa
 
M
,
Takenaka
 
N
,
Sakamoto
 
C
,
Baba
 
Y
, et al.  
Tissue doppler imaging and plasma BNP levels to assess the prognosis in patients with hypertrophic cardiomyopathy
.
J Am Soc Echocardiogr
 
2011
;
24
:
1020
1025
. https://doi.org/10.1016/j.echo.2011.05.009

620

Ha
 
J-W
,
Cho
 
J-R
,
Kim
 
J-M
,
Ahn
 
J-A
,
Choi
 
E-Y
,
Kang
 
S-M
, et al.  
Tissue Doppler-derived indices predict exercise capacity in patients with apical hypertrophic cardiomyopathy
.
Chest
 
2005
;
128
:
3428
3433
. https://doi.org/10.1378/chest.128.5.3428

621

Spoladore
 
R
,
Maron
 
MS
,
D’Amato
 
R
,
Camici
 
PG
,
Olivotto
 
I
.
Pharmacological treatment options for hypertrophic cardiomyopathy: high time for evidence
.
Eur Heart J
 
2012
;
33
:
1724
1733
. https://doi.org/10.1093/eurheartj/ehs150

622

Olivotto
 
I
,
Oreziak
 
A
,
Barriales-Villa
 
R
,
Abraham
 
TP
,
Masri
 
A
,
Garcia-Pavia
 
P
, et al.  
Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
 
2020
;
396
:
759
769
. https://doi.org/10.1016/S0140-6736(20)31792-X

623

Dybro
 
AM
,
Rasmussen
 
TB
,
Nielsen
 
RR
,
Ladefoged
 
BT
,
Andersen
 
MJ
,
Jensen
 
MK
, et al.  
Effects of metoprolol on exercise hemodynamics in patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2022
;
79
:
1565
1575
. https://doi.org/10.1016/j.jacc.2022.02.024

624

Wigle
 
ED
,
Auger
 
P
,
Marquis
 
Y
.
Muscular subaortic stenosis. The direct relation between the intraventricular pressure difference and the left ventricular ejection time
.
Circulation
 
1967
;
36
:
36
44
. https://doi.org/10.1161/01.CIR.36.1.36

625

Wigle
 
ED
,
Henderson
 
M
,
Rakowski
 
H
,
Wilansky
 
S
.
Muscular (hypertrophic) subaortic stenosis (hypertrophic obstructive cardiomyopathy): the evidence for true obstruction to left ventricular outflow
.
Postgrad Med J
 
1986
;
62
:
531
536
. https://doi.org/10.1136/pgmj.62.728.531

626

Stauffer
 
JC
,
Ruiz
 
V
,
Morard
 
JD
.
Subaortic obstruction after sildenafil in a patient with hypertrophic cardiomyopathy
.
N Engl J Med
 
1999
;
341
:
700
701
. https://doi.org/10.1056/NEJM199908263410916

627

Braunwald
 
E
,
Lambrew
 
CT
,
Rockoff
 
SD
,
Ross
 
J
 Jr
,
Morrow
 
AG
.
Idiopathic hypertrophic subaortic stenosis. I. A description of the disease based upon an analysis of 64 patients
.
Circulation
 
1964
;
30
(
Suppl 4
):
3
119
. https://doi.org/10.1161/01.cir.29.5s4.iv-3

628

Olivotto
 
I
,
Cecchi
 
F
,
Casey
 
SA
,
Dolara
 
A
,
Traverse
 
JH
,
Maron
 
BJ
.
Impact of atrial fibrillation on the clinical course of hypertrophic cardiomyopathy
.
Circulation
 
2001
;
104
:
2517
2524
. https://doi.org/10.1161/hc4601.097997

629

Camm
 
AJ
,
Lip
 
GY
,
De Caterina
 
R
,
Savelieva
 
I
,
Atar
 
D
,
Hohnloser
 
SH
, et al.  
2012 focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation. Developed with the special contribution of the European Heart Rhythm Association
.
Eur Heart J
 
2012
;
33
:
2719
2747
. https://doi.org/10.1093/eurheartj/ehs253

630

European Heart Rhythm Association, European Association for Cardio-Thoracic Surgery
;
Camm
 
AJ
,
Kirchhof
 
P
,
Lip
 
GYH
,
Schotten
 
U
,
Savelieva
 
I
, 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
2429
. https://doi.org/10.1093/eurheartj/ehq278

631

Dybro
 
AM
,
Rasmussen
 
TB
,
Nielsen
 
RR
,
Andersen
 
MJ
,
Jensen
 
MK
,
Poulsen
 
SH
.
Randomized trial of metoprolol in patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2021
;
78
:
2505
2517
. https://doi.org/10.1016/j.jacc.2021.07.065

632

Sherrid
 
MV
,
Barac
 
I
,
McKenna
 
WJ
,
Elliott
 
PM
,
Dickie
 
S
,
Chojnowska
 
L
, et al.  
Multicenter study of the efficacy and safety of disopyramide in obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2005
;
45
:
1251
1258
. https://doi.org/10.1016/j.jacc.2005.01.012

633

Sherrid
 
MV
,
Shetty
 
A
,
Winson
 
G
,
Kim
 
B
,
Musat
 
D
,
Alviar
 
CL
, et al.  
Treatment of obstructive hypertrophic cardiomyopathy symptoms and gradient resistant to first-line therapy with beta-blockade or verapamil
.
Circ Heart Fail
 
2013
;
6
:
694
702
. https://doi.org/10.1161/CIRCHEARTFAILURE.112.000122

634

O’Connor
 
MJ
,
Miller
 
K
,
Shaddy
 
RE
,
Lin
 
KY
,
Hanna
 
BD
,
Ravishankar
 
C
, et al.  
Disopyramide use in infants and children with hypertrophic cardiomyopathy
.
Cardiol Young
 
2018
;
28
:
530
535
. https://doi.org/10.1017/S1047951117002384

635

Adler
 
A
,
Fourey
 
D
,
Weissler-Snir
 
A
,
Hindieh
 
W
,
Chan
 
RH
,
Gollob
 
MH
, et al.  
Safety of outpatient initiation of disopyramide for obstructive hypertrophic cardiomyopathy patients
.
J Am Heart Assoc
 
2017
;
6
:
e005152
. https://doi.org/10.1161/JAHA.116.005152

636

Epstein
 
SE
,
Rosing
 
DR
.
Verapamil: its potential for causing serious complications in patients with hypertrophic cardiomyopathy
.
Circulation
 
1981
;
64
:
437
441
. https://doi.org/10.1161/01.CIR.64.3.437

637

Rosing
 
DR
,
Kent
 
KM
,
Maron
 
BJ
,
Epstein
 
SE
.
Verapamil therapy: a new approach to the pharmacologic treatment of hypertrophic cardiomyopathy. II. Effects on exercise capacity and symptomatic status
.
Circulation
 
1979
;
60
:
1208
1213
. https://doi.org/10.1161/01.CIR.60.6.1208

638

Bonow
 
RO
,
Rosing
 
DR
,
Epstein
 
SE
.
The acute and chronic effects of verapamil on left ventricular function in patients with hypertrophic cardiomyopathy
.
Eur Heart J
 
1983
;
4
(
Suppl F
):
57
65
. https://doi.org/10.1093/eurheartj/4.suppl_F.57

639

Spicer
 
RL
,
Rocchini
 
AP
,
Crowley
 
DC
,
Vasiliades
 
J
,
Rosenthal
 
A
.
Hemodynamic effects of verapamil in children and adolescents with hypertrophic cardiomyopathy
.
Circulation
 
1983
;
67
:
413
420
. https://doi.org/10.1161/01.CIR.67.2.413

640

Rosing
 
DR
,
Idanpaan-Heikkila
 
U
,
Maron
 
BJ
,
Bonow
 
RO
,
Epstein
 
SE
.
Use of calcium-channel blocking drugs in hypertrophic cardiomyopathy
.
Am J Cardiol
 
1985
;
55
:
185B
195B
. https://doi.org/10.1016/0002-9149(85)90630-7

641

Toshima
 
H
,
Koga
 
Y
,
Nagata
 
H
,
Toyomasu
 
K
,
Itaya
 
K
,
Matoba
 
T
.
Comparable effects of oral diltiazem and verapamil in the treatment of hypertrophic cardiomyopathy. Double-blind crossover study
.
Jpn Heart J
 
1986
;
27
:
701
715
. https://doi.org/10.1536/ihj.27.701

642

Desai
 
MY
,
Owens
 
A
,
Geske
 
JB
,
Wolski
 
K
,
Naidu
 
SS
,
Smedira
 
NG
, et al.  
Myosin inhibition in patients with obstructive hypertrophic cardiomyopathy referred for septal reduction therapy
.
J Am Coll Cardiol
 
2022
;
80
:
95
108
. https://doi.org/10.1016/j.jacc.2022.04.048

643

Desai
 
MY
,
Owens
 
A
,
Geske
 
JB
,
Wolski
 
K
,
Saberi
 
S
,
Wang
 
A
, et al.  
Dose-blinded myosin inhibition in patients with obstructive hypertrophic cardiomyopathy referred for septal reduction therapy: outcomes through 32 weeks
.
Circulation
 
2023
;
147
:
850
863
. https://doi.org/10.1161/CIRCULATIONAHA.122.062534

644

Spertus
 
JA
,
Fine
 
JT
,
Elliott
 
P
,
Ho
 
CY
,
Olivotto
 
I
,
Saberi
 
S
, et al.  
Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): health status analysis of a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
 
2021
;
397
:
2467
2475
. https://doi.org/10.1016/S0140-6736(21)00763-7

645

Saberi
 
S
,
Cardim
 
N
,
Yamani
 
M
,
Schulz-Menger
 
J
,
Li
 
W
,
Florea
 
V
, et al.  
Mavacamten favorably impacts cardiac structure in obstructive hypertrophic cardiomyopathy: EXPLORER-HCM cardiac magnetic resonance substudy analysis
.
Circulation
 
2021
;
143
:
606
608
. https://doi.org/10.1161/CIRCULATIONAHA.120.052359

646

Chuang
 
C
,
Collibee
 
S
,
Ashcraft
 
L
,
Wang
 
W
,
Vander Wal
 
M
,
Wang
 
X
, et al.  
Discovery of Aficamten (CK-274), a next-generation cardiac myosin inhibitor for the treatment of hypertrophic cardiomyopathy
.
J Med Chem
 
2021
;
64
:
14142
14152
. https://doi.org/10.1021/acs.jmedchem.1c01290

647

Maron
 
MS
,
Masri
 
A
,
Choudhury
 
L
,
Olivotto
 
I
,
Saberi
 
S
,
Wang
 
A
, et al.  
Phase 2 study of aficamten in patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2023
;
81
:
34
45
. https://doi.org/10.1016/j.jacc.2022.10.020

648

Adelman
 
AG
,
Shah
 
PM
,
Gramiak
 
R
,
Wigle
 
ED
.
Long-term propranolol therapy in muscular subaortic stenosis
.
Br Heart J
 
1970
;
32
:
804
811
. https://doi.org/10.1136/hrt.32.6.804

649

Flamm
 
MD
,
Harrison
 
DC
,
Hancock
 
EW
.
Muscular subaortic stenosis. Prevention of outflow obstruction with propranolol
.
Circulation
 
1968
;
38
:
846
858
. https://doi.org/10.1161/01.CIR.38.5.846

650

Monda
 
E
,
Lioncino
 
M
,
Palmiero
 
G
,
Franco
 
F
,
Rubino
 
M
,
Cirillo
 
A
, et al.  
Bisoprolol for treatment of symptomatic patients with obstructive hypertrophic cardiomyopathy. The BASIC (bisoprolol AS therapy in hypertrophic cardiomyopathy) study
.
Int J Cardiol
 
2022
;
354
:
22
28
. https://doi.org/10.1016/j.ijcard.2022.03.013

651

Sorajja
 
P
,
Nishimura
 
RA
,
Gersh
 
BJ
,
Dearani
 
JA
,
Hodge
 
DO
,
Wiste
 
HJ
, et al.  
Outcome of mildly symptomatic or asymptomatic obstructive hypertrophic cardiomyopathy: a long-term follow-up study
.
J Am Coll Cardiol
 
2009
;
54
:
234
241
. https://doi.org/10.1016/j.jacc.2009.01.079

652

Cavigli
 
L
,
Fumagalli
 
C
,
Maurizi
 
N
,
Rossi
 
A
,
Arretini
 
A
,
Targetti
 
M
, et al.  
Timing of invasive septal reduction therapies and outcome of patients with obstructive hypertrophic cardiomyopathy
.
Int J Cardiol
 
2018
;
273
:
155
161
. https://doi.org/10.1016/j.ijcard.2018.09.004

653

Menon
 
SC
,
Ackerman
 
MJ
,
Ommen
 
SR
,
Cabalka
 
AK
,
Hagler
 
DJ
,
O’Leary
 
PW
, et al.  
Impact of septal myectomy on left atrial volume and left ventricular diastolic filling patterns: an echocardiographic study of young patients with obstructive hypertrophic cardiomyopathy
.
J Am Soc Echocardiogr
 
2008
;
21
:
684
688
. https://doi.org/10.1016/j.echo.2007.11.006

654

Morrow
 
AG
,
Reitz
 
BA
,
Epstein
 
SE
,
Henry
 
WL
,
Conkle
 
DM
,
Itscoitz
 
SB
, et al.  
Operative treatment in hypertrophic subaortic stenosis. Techniques, and the results of pre and postoperative assessments in 83 patients
.
Circulation
 
1975
;
52
:
88
102
. https://doi.org/10.1161/01.CIR.52.1.88

655

Krajcer
 
Z
,
Leachman
 
RD
,
Cooley
 
DA
,
Coronado
 
R
.
Septal myotomy-myomectomy versus mitral valve replacement in hypertrophic cardiomyopathy. Ten-year follow-up in 185 patients
.
Circulation
 
1989
;
80
:
I57
I64
.

656

Heric
 
B
,
Lytle
 
BW
,
Miller
 
DP
,
Rosenkranz
 
ER
,
Lever
 
HM
,
Cosgrove
 
DM
.
Surgical management of hypertrophic obstructive cardiomyopathy. Early and late results
.
J Thorac Cardiovasc Surg
 
1995
;
110
:
195
206
;
discussion 206-8
. https://doi.org/10.1016/S0022-5223(05)80026-1

657

Robbins
 
RC
,
Stinson
 
EB
.
Long-term results of left ventricular myotomy and myectomy for obstructive hypertrophic cardiomyopathy
.
J Thorac Cardiovasc Surg
 
1996
;
111
:
586
594
. https://doi.org/10.1016/S0022-5223(96)70310-0

658

Schonbeck
 
MH
,
Brunner-La Rocca
 
HP
,
Vogt
 
PR
,
Lachat
 
ML
,
Jenni
 
R
,
Hess
 
OM
, et al.  
Long-term follow-up in hypertrophic obstructive cardiomyopathy after septal myectomy
.
Ann Thorac Surg
 
1998
;
65
:
1207
1214
. https://doi.org/10.1016/S0003-4975(98)00187-8

659

Schulte
 
HD
,
Borisov
 
K
,
Gams
 
E
,
Gramsch-Zabel
 
H
,
Losse
 
B
,
Schwartzkopff
 
B
.
Management of symptomatic hypertrophic obstructive cardiomyopathy–long-term results after surgical therapy
.
Thorac Cardiovasc Surg
 
1999
;
47
:
213
218
. https://doi.org/10.1055/s-2007-1013146

660

Ommen
 
SR
,
Maron
 
BJ
,
Olivotto
 
I
,
Maron
 
MS
,
Cecchi
 
F
,
Betocchi
 
S
, et al.  
Long-term effects of surgical septal myectomy on survival in patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2005
;
46
:
470
476
. https://doi.org/10.1016/j.jacc.2005.02.090

661

Woo
 
A
,
Williams
 
WG
,
Choi
 
R
,
Wigle
 
ED
,
Rozenblyum
 
E
,
Fedwick
 
K
, et al.  
Clinical and echocardiographic determinants of long-term survival after surgical myectomy in obstructive hypertrophic cardiomyopathy
.
Circulation
 
2005
;
111
:
2033
2041
. https://doi.org/10.1161/01.CIR.0000162460.36735.71

662

Smedira
 
NG
,
Lytle
 
BW
,
Lever
 
HM
,
Rajeswaran
 
J
,
Krishnaswamy
 
G
,
Kaple
 
RK
, et al.  
Current effectiveness and risks of isolated septal myectomy for hypertrophic obstructive cardiomyopathy
.
Ann Thorac Surg
 
2008
;
85
:
127
133
. https://doi.org/10.1016/j.athoracsur.2007.07.063

663

Desai
 
MY
,
Bhonsale
 
A
,
Smedira
 
NG
,
Naji
 
P
,
Thamilarasan
 
M
,
Lytle
 
BW
, et al.  
Predictors of long-term outcomes in symptomatic hypertrophic obstructive cardiomyopathy patients undergoing surgical relief of left ventricular outflow tract obstruction
.
Circulation
 
2013
;
128
:
209
216
. https://doi.org/10.1161/CIRCULATIONAHA.112.000849

664

Hodges
 
K
,
Rivas
 
CG
,
Aguilera
 
J
,
Borden
 
R
,
Alashi
 
A
,
Blackstone
 
EH
, et al.  
Surgical management of left ventricular outflow tract obstruction in a specialized hypertrophic obstructive cardiomyopathy center
.
J Thorac Cardiovasc Surg
 
2019
;
157
:
2289
2299
. https://doi.org/10.1016/j.jtcvs.2018.11.148

665

Nguyen
 
A
,
Schaff
 
HV
,
Nishimura
 
RA
,
Dearani
 
JA
,
Geske
 
JB
,
Lahr
 
BD
, et al.  
Does septal thickness influence outcome of myectomy for hypertrophic obstructive cardiomyopathy?
 
Eur J Cardiothorac Surg
 
2018
;
53
:
582
589
. https://doi.org/10.1093/ejcts/ezx398

666

Altarabsheh
 
SE
,
Dearani
 
JA
,
Burkhart
 
HM
,
Schaff
 
HV
,
Deo
 
SV
,
Eidem
 
BW
, et al.  
Outcome of septal myectomy for obstructive hypertrophic cardiomyopathy in children and young adults
.
Ann Thorac Surg
 
2013
;
95
:
663
669
;
discussion 669
. https://doi.org/10.1016/j.athoracsur.2012.08.011

667

Iacovoni
 
A
,
Spirito
 
P
,
Simon
 
C
,
Iascone
 
M
,
Di Dedda
 
G
,
De Filippo
 
P
, et al.  
A contemporary European experience with surgical septal myectomy in hypertrophic cardiomyopathy
.
Eur Heart J
 
2012
;
33
:
2080
2087
. https://doi.org/10.1093/eurheartj/ehs064

668

Dearani
 
JA
,
Ommen
 
SR
,
Gersh
 
BJ
,
Schaff
 
HV
,
Danielson
 
GK
.
Surgery insight: septal myectomy for obstructive hypertrophic cardiomyopathy–the Mayo Clinic experience
.
Nat Clin Pract Cardiovasc Med
 
2007
;
4
:
503
512
. https://doi.org/10.1038/ncpcardio0965

669

Kofflard
 
MJ
,
van Herwerden
 
LA
,
Waldstein
 
DJ
,
Ruygrok
 
P
,
Boersma
 
E
,
Taams
 
MA
, et al.  
Initial results of combined anterior mitral leaflet extension and myectomy in patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1996
;
28
:
197
202
. https://doi.org/10.1016/0735-1097(96)00103-9

670

McIntosh
 
CL
,
Maron
 
BJ
,
Cannon
 
RO
 III
,
Klues
 
HG
.
Initial results of combined anterior mitral leaflet plication and ventricular septal myotomy-myectomy for relief of left ventricular outflow tract obstruction in patients with hypertrophic cardiomyopathy
.
Circulation
 
1992
;
86
(
5 Suppl
):
II60
II7
.

671

Reis
 
RL
,
Bolton
 
MR
,
King
 
JF
,
Pugh
 
DM
,
Dunn
 
MI
,
Mason
 
DT
.
Anterion-superior displacement of papillary muscles producing obstruction and mitral regurgitation in idiopathic hypertrophic subaortic stenosis. Operative relief by posterior-superior realignment of papillary muscles following ventricular septal myectomy
.
Circulation
 
1974
;
50
(
2 Suppl
):
II181
II188
.

672

Schoendube
 
FA
,
Klues
 
HG
,
Reith
 
S
,
Flachskampf
 
FA
,
Hanrath
 
P
,
Messmer
 
BJ
.
Long-term clinical and echocardiographic follow-up after surgical correction of hypertrophic obstructive cardiomyopathy with extended myectomy and reconstruction of the subvalvular mitral apparatus
.
Circulation
 
1995
;
92
(
9 Suppl
):
II122
II127
. https://doi.org/10.1161/01.CIR.92.9.122

673

Kaple
 
RK
,
Murphy
 
RT
,
DiPaola
 
LM
,
Houghtaling
 
PL
,
Lever
 
HM
,
Lytle
 
BW
, et al.  
Mitral valve abnormalities in hypertrophic cardiomyopathy: echocardiographic features and surgical outcomes
.
Ann Thorac Surg
 
2008
;
85
:
1527
1535
,
1535.e1-2
. https://doi.org/10.1016/j.athoracsur.2008.01.061

674

Stassano
 
P
,
Di Tommaso
 
L
,
Triggiani
 
D
,
Contaldo
 
A
,
Gagliardi
 
C
,
Spampinato
 
N
.
Mitral valve replacement and limited myectomy for hypertrophic obstructive cardiomyopathy: a 25-year follow-up
.
Tex Heart Inst J
 
2004
;
31
:
137
142
.

675

Minakata
 
K
,
Dearani
 
JA
,
Nishimura
 
RA
,
Maron
 
BJ
,
Danielson
 
GK
.
Extended septal myectomy for hypertrophic obstructive cardiomyopathy with anomalous mitral papillary muscles or chordae
.
J Thorac Cardiovasc Surg
 
2004
;
127
:
481
489
. https://doi.org/10.1016/j.jtcvs.2003.09.040

676

Boll
 
G
,
Rowin
 
EJ
,
Maron
 
BJ
,
Wang
 
W
,
Rastegar
 
H
,
Maron
 
MS
.
Efficacy of combined Cox-Maze IV and ventricular septal myectomy for treatment of atrial fibrillation in patients with obstructive hypertrophic cardiomyopathy
.
Am J Cardiol
 
2020
;
125
:
120
126
. https://doi.org/10.1016/j.amjcard.2019.09.029

677

Laredo
 
M
,
Khraiche
 
D
,
Raisky
 
O
,
Gaudin
 
R
,
Bajolle
 
F
,
Maltret
 
A
, et al.  
Long-term results of the modified Konno procedure in high-risk children with obstructive hypertrophic cardiomyopathy
.
J Thorac Cardiovasc Surg
 
2018
;
156
:
2285
2294.e2
. https://doi.org/10.1016/j.jtcvs.2018.06.040

678

Sigwart
 
U
.
Non-surgical myocardial reduction for hypertrophic obstructive cardiomyopathy
.
Lancet
 
1995
;
346
:
211
214
. https://doi.org/10.1016/S0140-6736(95)91267-3

679

Faber
 
L
,
Welge
 
D
,
Fassbender
 
D
,
Schmidt
 
HK
,
Horstkotte
 
D
,
Seggewiss
 
H
.
One-year follow-up of percutaneous septal ablation for symptomatic hypertrophic obstructive cardiomyopathy in 312 patients: predictors of hemodynamic and clinical response
.
Clin Res Cardiol
 
2007
;
96
:
864
873
. https://doi.org/10.1007/s00392-007-0578-9

680

Fernandes
 
VL
,
Nielsen
 
C
,
Nagueh
 
SF
,
Herrin
 
AE
,
Slifka
 
C
,
Franklin
 
J
, et al.  
Follow-up of alcohol septal ablation for symptomatic hypertrophic obstructive cardiomyopathy the Baylor and Medical University of South Carolina experience 1996 to 2007
.
JACC Cardiovasc Interv
 
2008
;
1
:
561
570
. https://doi.org/10.1016/j.jcin.2008.07.005

681

Kuhn
 
H
,
Lawrenz
 
T
,
Lieder
 
F
,
Leuner
 
C
,
Strunk-Mueller
 
C
,
Obergassel
 
L
, et al.  
Survival after transcoronary ablation of septal hypertrophy in hypertrophic obstructive cardiomyopathy (TASH): a 10 year experience
.
Clin Res Cardiol
 
2008
;
97
:
234
243
. https://doi.org/10.1007/s00392-007-0616-7

682

Sorajja
 
P
,
Valeti
 
U
,
Nishimura
 
RA
,
Ommen
 
SR
,
Rihal
 
CS
,
Gersh
 
BJ
, et al.  
Outcome of alcohol septal ablation for obstructive hypertrophic cardiomyopathy
.
Circulation
 
2008
;
118
:
131
139
. https://doi.org/10.1161/CIRCULATIONAHA.107.738740

683

Sorajja
 
P
,
Ommen
 
SR
,
Holmes
 
DR
 Jr
,
Dearani
 
JA
,
Rihal
 
CS
,
Gersh
 
BJ
, et al.  
Survival after alcohol septal ablation for obstructive hypertrophic cardiomyopathy
.
Circulation
 
2012
;
126
:
2374
2380
. https://doi.org/10.1161/CIRCULATIONAHA.111.076257

684

Veselka
 
J
,
Krejci
 
J
,
Tomasov
 
P
,
Zemanek
 
D
.
Long-term survival after alcohol septal ablation for hypertrophic obstructive cardiomyopathy: a comparison with general population
.
Eur Heart J
 
2014
;
35
:
2040
2045
. https://doi.org/10.1093/eurheartj/eht495

685

Liebregts
 
M
,
Faber
 
L
,
Jensen
 
MK
,
Vriesendorp
 
PA
,
Januska
 
J
,
Krejci
 
J
, et al.  
Outcomes of alcohol septal ablation in younger patients with obstructive hypertrophic cardiomyopathy
.
JACC Cardiovasc Interv
 
2017
;
10
:
1134
1143
. https://doi.org/10.1016/j.jcin.2017.03.030

686

Veselka
 
J
,
Jensen
 
MK
,
Liebregts
 
M
,
Januska
 
J
,
Krejci
 
J
,
Bartel
 
T
, et al.  
Long-term clinical outcome after alcohol septal ablation for obstructive hypertrophic cardiomyopathy: results from the Euro-ASA registry
.
Eur Heart J
 
2016
;
37
:
1517
1523
. https://doi.org/10.1093/eurheartj/ehv693

687

Kim
 
LK
,
Swaminathan
 
RV
,
Looser
 
P
,
Minutello
 
RM
,
Wong
 
SC
,
Bergman
 
G
, et al.  
Hospital volume outcomes after septal myectomy and alcohol septal ablation for treatment of obstructive hypertrophic cardiomyopathy: US Nationwide Inpatient Database, 2003–2011
.
JAMA Cardiol
 
2016
;
1
:
324
332
. https://doi.org/10.1001/jamacardio.2016.0252

688

ten Cate
 
FJ
,
Soliman
 
OI
,
Michels
 
M
,
Theuns
 
DA
,
de Jong
 
PL
,
Geleijnse
 
ML
, et al.  
Long-term outcome of alcohol septal ablation in patients with obstructive hypertrophic cardiomyopathy: a word of caution
.
Circ Heart Fail
 
2010
;
3
:
362
369
. https://doi.org/10.1161/CIRCHEARTFAILURE.109.862359

689

Durand
 
E
,
Mousseaux
 
E
,
Coste
 
P
,
Pilliere
 
R
,
Dubourg
 
O
,
Trinquart
 
L
, et al.  
Non-surgical septal myocardial reduction by coil embolization for hypertrophic obstructive cardiomyopathy: early and 6 months follow-up
.
Eur Heart J
 
2008
;
29
:
348
355
. https://doi.org/10.1093/eurheartj/ehm632

690

Iacob
 
M
,
Pinte
 
F
,
Tintoiu
 
I
,
Cotuna
 
L
,
Caroescu
 
M
,
Popa
 
A
, et al.  
Microcoil embolisation for ablation of septal hypertrophy in hypertrophic obstructive cardiomyopathy
.
Kardiol Pol
 
2004
;
61
:
350
355
.

691

Gross
 
CM
,
Schulz-Menger
 
J
,
Kramer
 
J
,
Siegel
 
I
,
Pilz
 
B
,
Waigand
 
J
, et al.  
Percutaneous transluminal septal artery ablation using polyvinyl alcohol foam particles for septal hypertrophy in patients with hypertrophic obstructive cardiomyopathy: acute and 3-year outcomes
.
J Endovasc Ther
 
2004
;
11
:
705
711
. https://doi.org/10.1583/03-1171MR.1

692

Oto
 
A
,
Aytemir
 
K
,
Okutucu
 
S
,
Kaya
 
EB
,
Deniz
 
A
,
Cil
 
B
, et al.  
Cyanoacrylate for septal ablation in hypertrophic cardiomyopathy
.
J Interv Cardiol
 
2011
;
24
:
77
84
. https://doi.org/10.1111/j.1540-8183.2010.00605.x

693

Lawrenz
 
T
,
Borchert
 
B
,
Leuner
 
C
,
Bartelsmeier
 
M
,
Reinhardt
 
J
,
Strunk-Mueller
 
C
.
Endocardial radiofrequency ablation for hypertrophic obstructive cardiomyopathy: acute results and 6 months’ follow-up in 19 patients
.
J Am Coll Cardiol
 
2011
;
57
:
572
576
. https://doi.org/10.1016/j.jacc.2010.07.055

694

Keane
 
D
,
Hynes
 
B
,
King
 
G
,
Shiels
 
P
,
Brown
 
A
.
Feasibility study of percutaneous transvalvular endomyocardial cryoablation for the treatment of hypertrophic obstructive cardiomyopathy
.
J Invasive Cardiol
 
2007
;
19
:
247
251
.

695

Panaich
 
SS
,
Badheka
 
AO
,
Chothani
 
A
,
Mehta
 
K
,
Patel
 
NJ
,
Deshmukh
 
A
, et al.  
Results of ventricular septal myectomy and hypertrophic cardiomyopathy (from Nationwide Inpatient Sample [1998–2010])
.
Am J Cardiol
 
2014
;
114
:
1390
1395
. https://doi.org/10.1016/j.amjcard.2014.07.075

696

Bourque
 
C
,
Reant
 
P
,
Bernard
 
A
,
Leroux
 
L
,
Bonnet
 
G
,
Pernot
 
M
, et al.  
Comparison of surgical ventricular septal reduction to alcohol septal ablation therapy in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2022
;
172
:
109
114
. https://doi.org/10.1016/j.amjcard.2022.02.033

697

Agarwal
 
S
,
Tuzcu
 
EM
,
Desai
 
MY
,
Smedira
 
N
,
Lever
 
HM
,
Lytle
 
BW
, et al.  
Updated meta-analysis of septal alcohol ablation versus myectomy for hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2010
;
55
:
823
834
. https://doi.org/10.1016/j.jacc.2009.09.047

698

Alam
 
M
,
Dokainish
 
H
,
Lakkis
 
NM
.
Hypertrophic obstructive cardiomyopathy–alcohol septal ablation vs. myectomy: a meta-analysis
.
Eur Heart J
 
2009
;
30
:
1080
1087
. https://doi.org/10.1093/eurheartj/ehp016

699

Zeng
 
Z
,
Wang
 
F
,
Dou
 
X
,
Zhang
 
S
,
Pu
 
J
.
Comparison of percutaneous transluminal septal myocardial ablation versus septal myectomy for the treatment of patients with hypertrophic obstructive cardiomyopathy–a meta analysis
.
Int J Cardiol
 
2006
;
112
:
80
84
. https://doi.org/10.1016/j.ijcard.2005.10.009

700

Leonardi
 
RA
,
Kransdorf
 
EP
,
Simel
 
DL
,
Wang
 
A
.
Meta-analyses of septal reduction therapies for obstructive hypertrophic cardiomyopathy: comparative rates of overall mortality and sudden cardiac death after treatment
.
Circ Cardiovasc Interv
 
2010
;
3
:
97
104
. https://doi.org/10.1161/CIRCINTERVENTIONS.109.916676

701

Batzner
 
A
,
Pfeiffer
 
B
,
Neugebauer
 
A
,
Aicha
 
D
,
Blank
 
C
,
Seggewiss
 
H
.
Survival after alcohol septal ablation in patients with hypertrophic obstructive cardiomyopathy
.
J Am Coll Cardiol
 
2018
;
72
:
3087
3094
. https://doi.org/10.1016/j.jacc.2018.09.064

702

Nguyen
 
A
,
Schaff
 
HV
,
Hang
 
D
,
Nishimura
 
RA
,
Geske
 
JB
,
Dearani
 
JA
, et al.  
Surgical myectomy versus alcohol septal ablation for obstructive hypertrophic cardiomyopathy: a propensity score-matched cohort
.
J Thorac Cardiovasc Surg
 
2019
;
157
:
306
315.e3
. https://doi.org/10.1016/j.jtcvs.2018.08.062

703

Bytyci
 
I
,
Nistri
 
S
,
Morner
 
S
,
Henein
 
MY
.
Alcohol septal ablation versus septal myectomy treatment of obstructive hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
J Clin Med
 
2020
;
9
:
3062
. https://doi.org/10.3390/jcm9103062

704

Faber
 
L
,
Welge
 
D
,
Fassbender
 
D
,
Schmidt
 
HK
,
Horstkotte
 
D
,
Seggewiss
 
H
.
Percutaneous septal ablation for symptomatic hypertrophic obstructive cardiomyopathy: managing the risk of procedure-related AV conduction disturbances
.
Int J Cardiol
 
2007
;
119
:
163
167
. https://doi.org/10.1016/j.ijcard.2006.07.179

705

Veselka
 
J
,
Liebregts
 
M
,
Cooper
 
R
,
Faber
 
L
,
Januska
 
J
,
Kashtanov
 
M
, et al.  
Outcomes of patients with hypertrophic obstructive cardiomyopathy and pacemaker implanted after alcohol septal ablation
.
JACC Cardiovasc Interv
 
2022
;
15
:
1910
1917
. https://doi.org/10.1016/j.jcin.2022.06.034

706

Veselka
 
J
,
Jensen
 
M
,
Liebregts
 
M
,
Cooper
 
RM
,
Januska
 
J
,
Kashtanov
 
M
, et al.  
Alcohol septal ablation in patients with severe septal hypertrophy
.
Heart
 
2020
;
106
:
462
466
. https://doi.org/10.1136/heartjnl-2019-315422

707

Veselka
 
J
,
Faber
 
L
,
Liebregts
 
M
,
Cooper
 
R
,
Januska
 
J
,
Kashtanov
 
M
, et al.  
Short- and long-term outcomes of alcohol septal ablation for hypertrophic obstructive cardiomyopathy in patients with mild left ventricular hypertrophy: a propensity score matching analysis
.
Eur Heart J
 
2019
;
40
:
1681
1687
. https://doi.org/10.1093/eurheartj/ehz110

708

Tumiene
 
B
,
Graessner
 
H
,
Mathijssen
 
IM
,
Pereira
 
AM
,
Schaefer
 
F
,
Scarpa
 
M
, et al.  
European reference networks: challenges and opportunities
.
J Community Genet
 
2021
;
12
:
217
229
. https://doi.org/10.1007/s12687-021-00521-8

709

Veselka
 
J
,
Faber
 
L
,
Jensen
 
MK
,
Cooper
 
R
,
Januska
 
J
,
Krejci
 
J
, et al.  
Effect of institutional experience on outcomes of alcohol septal ablation for hypertrophic obstructive cardiomyopathy
.
Can J Cardiol
 
2018
;
34
:
16
22
. https://doi.org/10.1016/j.cjca.2017.10.020

710

Cui
 
H
,
Schaff
 
HV
,
Wang
 
S
,
Lahr
 
BD
,
Rowin
 
EJ
,
Rastegar
 
H
, et al.  
Survival following alcohol septal ablation or septal myectomy for patients with obstructive hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2022
;
79
:
1647
1655
. https://doi.org/10.1016/j.jacc.2022.02.032

711

McCully
 
RB
,
Nishimura
 
RA
,
Tajik
 
AJ
,
Schaff
 
HV
,
Danielson
 
GK
.
Extent of clinical improvement after surgical treatment of hypertrophic obstructive cardiomyopathy
.
Circulation
 
1996
;
94
:
467
471
. https://doi.org/10.1161/01.CIR.94.3.467

712

Orme
 
NM
,
Sorajja
 
P
,
Dearani
 
JA
,
Schaff
 
HV
,
Gersh
 
BJ
,
Ommen
 
SR
.
Comparison of surgical septal myectomy to medical therapy alone in patients with hypertrophic cardiomyopathy and syncope
.
Am J Cardiol
 
2013
;
111
:
388
392
. https://doi.org/10.1016/j.amjcard.2012.10.014

713

Geske
 
JB
,
Driver
 
CN
,
Yogeswaran
 
V
,
Ommen
 
SR
,
Schaff
 
HV
.
Comparison of expected and observed outcomes for septal myectomy in hypertrophic obstructive cardiomyopathy
.
Am Heart J
 
2020
;
221
:
159
164
. https://doi.org/10.1016/j.ahj.2019.11.020

714

Ferrazzi
 
P
,
Spirito
 
P
,
Iacovoni
 
A
,
Calabrese
 
A
,
Migliorati
 
K
,
Simon
 
C
, et al.  
Transaortic chordal cutting: mitral valve repair for obstructive hypertrophic cardiomyopathy with mild septal hypertrophy
.
J Am Coll Cardiol
 
2015
;
66
:
1687
1696
. https://doi.org/10.1016/j.jacc.2015.07.069

715

Veselka
 
J
,
Faber
 
L
,
Liebregts
 
M
,
Cooper
 
R
,
Januska
 
J
,
Krejci
 
J
, et al.  
Outcome of alcohol septal ablation in mildly symptomatic patients with hypertrophic obstructive cardiomyopathy: a long-term follow-up study based on the Euro-alcohol septal ablation registry
.
J Am Heart Assoc
 
2017
;
6
:
e005735
. https://doi.org/10.1161/JAHA.117.005735

716

Cooley
 
DA
,
Wukasch
 
DC
,
Leachman
 
RD
.
Mitral valve replacement for idiopathic hypertrophic subaortic stenosis. Results in 27 patients
.
J Cardiovasc Surg (Torino)
 
1976
;
17
:
380
387
.

717

Chen
 
MS
,
McCarthy
 
PM
,
Lever
 
HM
,
Smedira
 
NG
,
Lytle
 
BL
.
Effectiveness of atrial fibrillation surgery in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2004
;
93
:
373
375
. https://doi.org/10.1016/j.amjcard.2003.10.025

718

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
2091
. https://doi.org/10.1056/NEJMoa2101897

719

Slade
 
AK
,
Sadoul
 
N
,
Shapiro
 
L
,
Chojnowska
 
L
,
Simon
 
JP
,
Saumarez
 
RC
, et al.  
DDD pacing in hypertrophic cardiomyopathy: a multicentre clinical experience
.
Heart
 
1996
;
75
:
44
49
. https://doi.org/10.1136/hrt.75.1.44

720

Nishimura
 
RA
,
Trusty
 
JM
,
Hayes
 
DL
,
Ilstrup
 
DM
,
Larson
 
DR
,
Hayes
 
SN
, et al.  
Dual-chamber pacing for hypertrophic cardiomyopathy: a randomized, double-blind, crossover trial
.
J Am Coll Cardiol
 
1997
;
29
:
435
441
. https://doi.org/10.1016/S0735-1097(96)00473-1

721

Kappenberger
 
L
,
Linde
 
C
,
Daubert
 
C
,
McKenna
 
W
,
Meisel
 
E
,
Sadoul
 
N
, et al.  
Pacing in hypertrophic obstructive cardiomyopathy. A randomized crossover study
.
Eur Heart J
 
1997
;
18
:
1249
1256
. https://doi.org/10.1093/oxfordjournals.eurheartj.a015435

722

Maron
 
BJ
,
Nishimura
 
RA
,
McKenna
 
WJ
,
Rakowski
 
H
,
Josephson
 
ME
,
Kieval
 
RS
.
Assessment of permanent dual-chamber pacing as a treatment for drug-refractory symptomatic patients with obstructive hypertrophic cardiomyopathy. A randomized, double-blind, crossover study (M-PATHY)
.
Circulation
 
1999
;
99
:
2927
2933
. https://doi.org/10.1161/01.CIR.99.22.2927

723

Mickelsen
 
S
,
Bathina
 
M
,
Hsu
 
P
,
Holmes
 
J
,
Kusumoto
 
FM
.
Doppler evaluation of the descending aorta in patients with hypertrophic cardiomyopathy: potential for assessing the functional significance of outflow tract gradients and for optimizing pacemaker function
.
J Interv Card Electrophysiol
 
2004
;
11
:
47
53
. https://doi.org/10.1023/B:JICE.0000035929.84238.2f

724

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
3520
. https://doi.org/10.1093/eurheartj/ehab364

725

Qintar
 
M
,
Morad
 
A
,
Alhawasli
 
H
,
Shorbaji
 
K
,
Firwana
 
B
,
Essali
 
A
, et al.  
Pacing for drug-refractory or drug-intolerant hypertrophic cardiomyopathy
.
Cochrane Database Syst Rev
 
2012
;
2012
:
CD008523
. https://doi.org/10.1002/14651858.CD008523.pub2

726

O’Mahony
 
C
,
Lambiase
 
PD
,
Quarta
 
G
,
Cardona
 
M
,
Calcagnino
 
M
,
Tsovolas
 
K
, et al.  
The long-term survival and the risks and benefits of implantable cardioverter defibrillators in patients with hypertrophic cardiomyopathy
.
Heart
 
2012
;
98
:
116
125
. https://doi.org/10.1136/hrt.2010.217182

727

Minami
 
Y
,
Kajimoto
 
K
,
Terajima
 
Y
,
Yashiro
 
B
,
Okayama
 
D
,
Haruki
 
S
, et al.  
Clinical implications of midventricular obstruction in patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2011
;
57
:
2346
2355
. https://doi.org/10.1016/j.jacc.2011.02.033

728

Efthimiadis
 
GK
,
Pagourelias
 
ED
,
Parcharidou
 
D
,
Gossios
 
T
,
Kamperidis
 
V
,
Theofilogiannakos
 
EK
, et al.  
Clinical characteristics and natural history of hypertrophic cardiomyopathy with midventricular obstruction
.
Circ J
 
2013
;
77
:
2366
2374
. https://doi.org/10.1253/circj.CJ-12-1561

729

Shah
 
A
,
Duncan
 
K
,
Winson
 
G
,
Chaudhry
 
FA
,
Sherrid
 
MV
.
Severe symptoms in mid and apical hypertrophic cardiomyopathy
.
Echocardiography
 
2009
;
26
:
922
933
. https://doi.org/10.1111/j.1540-8175.2009.00905.x

730

Alfonso
 
F
,
Frenneaux
 
MP
,
McKenna
 
WJ
.
Clinical sustained uniform ventricular tachycardia in hypertrophic cardiomyopathy: association with left ventricular apical aneurysm
.
Br Heart J
 
1989
;
61
:
178
181
. https://doi.org/10.1136/hrt.61.2.178

731

Said
 
SM
,
Schaff
 
HV
,
Abel
 
MD
,
Dearani
 
JA
.
Transapical approach for apical myectomy and relief of midventricular obstruction in hypertrophic cardiomyopathy
.
J Card Surg
 
2012
;
27
:
443
448
. https://doi.org/10.1111/j.1540-8191.2012.01475.x

732

Kunkala
 
MR
,
Schaff
 
HV
,
Nishimura
 
RA
,
Abel
 
MD
,
Sorajja
 
P
,
Dearani
 
JA
, et al.  
Transapical approach to myectomy for midventricular obstruction in hypertrophic cardiomyopathy
.
Ann Thorac Surg
 
2013
;
96
:
564
570
. https://doi.org/10.1016/j.athoracsur.2013.04.073

733

Gao
 
X-J
,
Kang
 
L-M
,
Zhang
 
J
,
Dou
 
K-F
,
Yuan
 
J-S
,
Yang
 
Y-J
.
Mid-ventricular obstructive hypertrophic cardiomyopathy with apical aneurysm and sustained ventricular tachycardia: a case report and literature review
.
Chin Med J (Engl)
 
2011
;
124
:
1754
1757
.

734

Takeda
 
I
,
Sekine
 
M
,
Matsushima
 
H
,
Hosomi
 
N
,
Nakamura
 
T
,
Ohtsuki
 
T
, et al.  
Two cases of cerebral embolism caused by apical thrombi in midventricular obstructive cardiomyopathy
.
Intern Med
 
2011
;
50
:
1059
1060
. https://doi.org/10.2169/internalmedicine.50.5079

735

Sato
 
Y
,
Matsumoto
 
N
,
Matsuo
 
S
,
Yoda
 
S
,
Tani
 
S
,
Kasamaki
 
Y
, et al.  
Mid-ventricular obstructive hypertrophic cardiomyopathy associated with an apical aneurysm: evaluation of possible causes of aneurysm formation
.
Yonsei Med J
 
2007
;
48
:
879
882
. https://doi.org/10.3349/ymj.2007.48.5.879

736

Papanastasiou
 
CA
,
Zegkos
 
T
,
Kokkinidis
 
DG
,
Parcharidou
 
D
,
Karamitsos
 
TD
,
Efthimiadis
 
GK
.
Prognostic role of left ventricular apical aneurysm in hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
Int J Cardiol
 
2021
;
339
:
108
. https://doi.org/10.1016/j.ijcard.2021.07.025

737

Rowin
 
EJ
,
Maron
 
BJ
,
Haas
 
TS
,
Garberich
 
RF
,
Wang
 
W
,
Link
 
MS
, et al.  
Hypertrophic cardiomyopathy with left ventricular apical aneurysm: implications for risk stratification and management
.
J Am Coll Cardiol
 
2017
;
69
:
761
773
. https://doi.org/10.1016/j.jacc.2016.11.063

738

Ammirati
 
E
,
Contri
 
R
,
Coppini
 
R
,
Cecchi
 
F
,
Frigerio
 
M
,
Olivotto
 
I
.
Pharmacological treatment of hypertrophic cardiomyopathy: current practice and novel perspectives
.
Eur J Heart Fail
 
2016
;
18
:
1106
1118
. https://doi.org/10.1002/ejhf.541

739

Olivotto
 
I
,
Camici
 
PG
,
Merlini
 
PA
,
Rapezzi
 
C
,
Patten
 
M
,
Climent
 
V
, et al.  
Efficacy of ranolazine in patients with symptomatic hypertrophic cardiomyopathy: the RESTYLE-HCM randomized, double-blind, placebo-controlled study
.
Circ Heart Fail
 
2018
;
11
:
e004124
. https://doi.org/10.1161/CIRCHEARTFAILURE.117.004124

740

Bourmayan
 
C
,
Razavi
 
A
,
Fournier
 
C
,
Dussaule
 
JC
,
Baragan
 
J
,
Gerbaux
 
A
, et al.  
Effect of propranolol on left ventricular relaxation in hypertrophic cardiomyopathy: an echographic study
.
Am Heart J
 
1985
;
109
:
1311
1316
. https://doi.org/10.1016/0002-8703(85)90357-6

741

Alvares
 
RF
,
Goodwin
 
JF
.
Non-invasive assessment of diastolic function in hypertrophic cardiomyopathy on and off beta adrenergic blocking drugs
.
Br Heart J
 
1982
;
48
:
204
212
. https://doi.org/10.1136/hrt.48.3.204

742

Wilmshurst
 
PT
,
Thompson
 
DS
,
Juul
 
SM
,
Jenkins
 
BS
,
Webb-Peploe
 
MM
.
Effects of verapamil on haemodynamic function and myocardial metabolism in patients with hypertrophic cardiomyopathy
.
Br Heart J
 
1986
;
56
:
544
553
. https://doi.org/10.1136/hrt.56.6.544

743

Udelson
 
JE
,
Bonow
 
RO
,
O’Gara
 
PT
,
Maron
 
BJ
,
Van Lingen
 
A
,
Bacharach
 
SL
, et al.  
Verapamil prevents silent myocardial perfusion abnormalities during exercise in asymptomatic patients with hypertrophic cardiomyopathy
.
Circulation
 
1989
;
79
:
1052
1060
. https://doi.org/10.1161/01.CIR.79.5.1052

744

Pacileo
 
G
,
De Cristofaro
 
M
,
Russo
 
MG
,
Sarubbi
 
B
,
Pisacane
 
C
,
Calabro
 
R
.
Hypertrophic cardiomyopathy in pediatric patients: effect of verapamil on regional and global left ventricular diastolic function
.
Can J Cardiol
 
2000
;
16
:
146
152
.

745

Cappelli
 
F
,
Morini
 
S
,
Pieragnoli
 
P
,
Targetti
 
M
,
Stefano
 
P
,
Marchionni
 
N
, et al.  
Cardiac resynchronization therapy for end-stage hypertrophic cardiomyopathy: the need for disease-specific criteria
.
J Am Coll Cardiol
 
2018
;
71
:
464
466
. https://doi.org/10.1016/j.jacc.2017.11.040

746

Killu
 
AM
,
Park
 
J-Y
,
Sara
 
JD
,
Hodge
 
DO
,
Gersh
 
BJ
,
Nishimura
 
RA
, et al.  
Cardiac resynchronization therapy in patients with end-stage hypertrophic cardiomyopathy
.
Europace
 
2018
;
20
:
82
88
. https://doi.org/10.1093/europace/euw327

747

Gu
 
M
,
Jin
 
H
,
Hua
 
W
,
Fan
 
X-H
,
Niu
 
H-X
,
Tian
 
T
, et al.  
Clinical outcome of cardiac resynchronization therapy in dilated-phase hypertrophic cardiomyopathy
.
J Geriatr Cardiol
 
2017
;
14
:
238
244
. https://doi.org/10.11909/j.issn.1671-5411.2017.04.002

748

Ahmed
 
I
,
Loudon
 
BL
,
Abozguia
 
K
,
Cameron
 
D
,
Shivu
 
GN
,
Phan
 
TT
, et al.  
Biventricular pacemaker therapy improves exercise capacity in patients with non-obstructive hypertrophic cardiomyopathy via augmented diastolic filling on exercise
.
Eur J Heart Fail
 
2020
;
22
:
1263
1272
. https://doi.org/10.1002/ejhf.1722

749

Elliott
 
PM
,
Gimeno
 
JR
,
Thaman
 
R
,
Shah
 
J
,
Ward
 
D
,
Dickie
 
S
, et al.  
Historical trends in reported survival rates in patients with hypertrophic cardiomyopathy
.
Heart
 
2006
;
92
:
785
791
. https://doi.org/10.1136/hrt.2005.068577

750

Barriales-Villa
 
R
,
Centurion-Inda
 
R
,
Fernandez-Fernandez
 
X
,
Ortiz
 
MF
,
Perez-Alvarez
 
L
,
Rodriguez Garcia
 
I
, et al.  
Severe cardiac conduction disturbances and pacemaker implantation in patients with hypertrophic cardiomyopathy
.
Rev Esp Cardiol
 
2010
;
63
:
985
988
. https://doi.org/10.1016/S0300-8932(10)70210-4

751

Nicod
 
P
,
Polikar
 
R
,
Peterson
 
KL
.
Hypertrophic cardiomyopathy and sudden death
.
N Engl J Med
 
1988
;
318
:
1255
1257
. https://doi.org/10.1056/NEJM198805123181907

752

Stafford
 
WJ
,
Trohman
 
RG
,
Bilsker
 
M
,
Zaman
 
L
,
Castellanos
 
A
,
Myerburg
 
RJ
.
Cardiac arrest in an adolescent with atrial fibrillation and hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1986
;
7
:
701
704
. https://doi.org/10.1016/S0735-1097(86)80484-3

753

Krikler
 
DM
,
Davies
 
MJ
,
Rowland
 
E
,
Goodwin
 
JF
,
Evans
 
RC
,
Shaw
 
DB
.
Sudden death in hypertrophic cardiomyopathy: associated accessory atrioventricular pathways
.
Br Heart J
 
1980
;
43
:
245
251
. https://doi.org/10.1136/hrt.43.3.245

754

Joseph
 
S
,
Balcon
 
R
,
McDonald
 
L
.
Syncope in hypertrophic obstructive cardiomyopathy due to asystole
.
Br Heart J
 
1972
;
34
:
974
976
. https://doi.org/10.1136/hrt.34.9.974

755

McKenna
 
WJ
,
Deanfield
 
JE
.
Hypertrophic cardiomyopathy: an important cause of sudden death
.
Arch Dis Child
 
1984
;
59
:
971
975
. https://doi.org/10.1136/adc.59.10.971

756

McKenna
 
WJ
,
Franklin
 
RC
,
Nihoyannopoulos
 
P
,
Robinson
 
KC
,
Deanfield
 
JE
.
Arrhythmia and prognosis in infants, children and adolescents with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1988
;
11
:
147
153
. https://doi.org/10.1016/0735-1097(88)90181-7

757

Ostman-Smith
 
I
,
Wettrell
 
G
,
Keeton
 
B
,
Holmgren
 
D
,
Ergander
 
U
,
Gould
 
S
, et al.  
Age- and gender-specific mortality rates in childhood hypertrophic cardiomyopathy
.
Eur Heart J
 
2008
;
29
:
1160
1167
. https://doi.org/10.1093/eurheartj/ehn122

758

Lipshultz
 
SE
,
Orav
 
EJ
,
Wilkinson
 
JD
,
Towbin
 
JA
,
Messere
 
JE
,
Lowe
 
AM
, et al.  
Risk stratification at diagnosis for children with hypertrophic cardiomyopathy: an analysis of data from the Pediatric Cardiomyopathy Registry
.
Lancet
 
2013
;
382
:
1889
1897
. https://doi.org/10.1016/S0140-6736(13)61685-2

759

Marston
 
NA
,
Han
 
L
,
Olivotto
 
I
,
Day
 
SM
,
Ashley
 
EA
,
Michels
 
M
, et al.  
Clinical characteristics and outcomes in childhood-onset hypertrophic cardiomyopathy
.
Eur Heart J
 
2021
;
42
:
1988
1996
. https://doi.org/10.1093/eurheartj/ehab148

760

Sorajja
 
P
,
Ommen
 
SR
,
Nishimura
 
RA
,
Gersh
 
BJ
,
Berger
 
PB
,
Tajik
 
AJ
.
Adverse prognosis of patients with hypertrophic cardiomyopathy who have epicardial coronary artery disease
.
Circulation
 
2003
;
108
:
2342
2348
. https://doi.org/10.1161/01.CIR.0000097110.55312.BF

761

Kofflard
 
MJ
,
Ten Cate
 
FJ
,
van der Lee
 
C
,
van Domburg
 
RT
.
Hypertrophic cardiomyopathy in a large community-based population: clinical outcome and identification of risk factors for sudden cardiac death and clinical deterioration
.
J Am Coll Cardiol
 
2003
;
41
:
987
993
. https://doi.org/10.1016/S0735-1097(02)03004-8

762

Maki
 
S
,
Ikeda
 
H
,
Muro
 
A
,
Yoshida
 
N
,
Shibata
 
A
,
Koga
 
Y
, et al.  
Predictors of sudden cardiac death in hypertrophic cardiomyopathy
.
Am J Cardiol
 
1998
;
82
:
774
778
. https://doi.org/10.1016/S0002-9149(98)00455-X

763

Autore
 
C
,
Bernabo
 
P
,
Barilla
 
CS
,
Bruzzi
 
P
,
Spirito
 
P
.
The prognostic importance of left ventricular outflow obstruction in hypertrophic cardiomyopathy varies in relation to the severity of symptoms
.
J Am Coll Cardiol
 
2005
;
45
:
1076
1080
. https://doi.org/10.1016/j.jacc.2004.12.067

764

D’Andrea
 
A
,
Caso
 
P
,
Severino
 
S
,
Cuomo
 
S
,
Capozzi
 
G
,
Calabro
 
P
, et al.  
Prognostic value of intra-left ventricular electromechanical asynchrony in patients with hypertrophic cardiomyopathy
.
Eur Heart J
 
2006
;
27
:
1311
1318
. https://doi.org/10.1093/eurheartj/ehi688

765

Monserrat
 
L
,
Elliott
 
PM
,
Gimeno
 
JR
,
Sharma
 
S
,
Penas-Lado
 
M
,
McKenna
 
WJ
.
Non-sustained ventricular tachycardia in hypertrophic cardiomyopathy: an independent marker of sudden death risk in young patients
.
J Am Coll Cardiol
 
2003
;
42
:
873
879
. https://doi.org/10.1016/S0735-1097(03)00827-1

766

Maron
 
BJ
,
Casey
 
SA
,
Hurrell
 
DG
,
Aeppli
 
DM
.
Relation of left ventricular thickness to age and gender in hypertrophic cardiomyopathy
.
Am J Cardiol
 
2003
;
91
:
1195
1198
. https://doi.org/10.1016/S0002-9149(03)00266-2

767

Norrish
 
G
,
Cleary
 
A
,
Field
 
E
,
Cervi
 
E
,
Boleti
 
O
,
Ziolkowska
 
L
, et al.  
Clinical features and natural history of preadolescent nonsyndromic hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2022
;
79
:
1986
1997
. https://doi.org/10.1016/j.jacc.2022.03.347

768

Gimeno
 
JR
,
Tome-Esteban
 
M
,
Lofiego
 
C
,
Hurtado
 
J
,
Pantazis
 
A
,
Mist
 
B
, et al.  
Exercise-induced ventricular arrhythmias and risk of sudden cardiac death in patients with hypertrophic cardiomyopathy
.
Eur Heart J
 
2009
;
30
:
2599
2605
. https://doi.org/10.1093/eurheartj/ehp327

769

Dimitrow
 
PP
,
Chojnowska
 
L
,
Rudzinski
 
T
,
Piotrowski
 
W
,
Ziolkowska
 
L
,
Wojtarowicz
 
A
, et al.  
Sudden death in hypertrophic cardiomyopathy: old risk factors re-assessed in a new model of maximalized follow-up
.
Eur Heart J
 
2010
;
31
:
3084
3093
. https://doi.org/10.1093/eurheartj/ehq308

770

Maron
 
BJ
,
Spirito
 
P
,
Ackerman
 
MJ
,
Casey
 
SA
,
Semsarian
 
C
,
Estes
 
NA
 III
, et al.  
Prevention of sudden cardiac death with implantable cardioverter-defibrillators in children and adolescents with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2013
;
61
:
1527
1535
. https://doi.org/10.1016/j.jacc.2013.01.037

771

Ostman-Smith
 
I
,
Sjoberg
 
G
,
Rydberg
 
A
,
Larsson
 
P
,
Fernlund
 
E
.
Predictors of risk for sudden death in childhood hypertrophic cardiomyopathy: the importance of the ECG risk score
.
Open Heart
 
2017
;
4
:
e000658
. https://doi.org/10.1136/openhrt-2017-000658

772

Ziolkowska
 
L
,
Turska-Kmiec
 
A
,
Petryka
 
J
,
Kawalec
 
W
.
Predictors of long-term outcome in children with hypertrophic cardiomyopathy
.
Pediatr Cardiol
 
2016
;
37
:
448
458
. https://doi.org/10.1007/s00246-015-1298-y

773

Yetman
 
AT
,
Hamilton
 
RM
,
Benson
 
LN
,
McCrindle
 
BW
.
Long-term outcome and prognostic determinants in children with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
1998
;
32
:
1943
1950
. https://doi.org/10.1016/S0735-1097(98)00493-8

774

Cecchi
 
F
,
Olivotto
 
I
,
Montereggi
 
A
,
Squillatini
 
G
,
Dolara
 
A
,
Maron
 
BJ
.
Prognostic value of non-sustained ventricular tachycardia and the potential role of amiodarone treatment in hypertrophic cardiomyopathy: assessment in an unselected non-referral based patient population
.
Heart
 
1998
;
79
:
331
336
. https://doi.org/10.1136/hrt.79.4.331

775

Jensen
 
MK
,
Jacobsson
 
L
,
Almaas
 
V
,
van Buuren
 
F
,
Hansen
 
PR
,
Hansen
 
TF
, et al.  
Influence of septal thickness on the clinical outcome after alcohol septal alation in hypertrophic cardiomyopathy
.
Circ Cardiovasc Interv
 
2016
;
9
:
e003214
. https://doi.org/10.1161/CIRCINTERVENTIONS.115.003214

776

Nakajima
 
S
,
Morioka
 
S
.
[Effects of plasminogen activator on epidermal cell migration]
.
Nihon Hifuka Gakkai Zasshi
 
1990
;
100
:
1199
1201
.

777

Balaji
 
S
,
DiLorenzo
 
MP
,
Fish
 
FA
,
Etheridge
 
SP
,
Aziz
 
PF
,
Russell
 
MW
, et al.  
Risk factors for lethal arrhythmic events in children and adolescents with hypertrophic cardiomyopathy and an implantable defibrillator: an international multicenter study
.
Heart Rhythm
 
2019
;
16
:
1462
1467
. https://doi.org/10.1016/j.hrthm.2019.04.040

778

Bharucha
 
T
,
Lee
 
KJ
,
Daubeney
 
PE
,
Nugent
 
AW
,
Turner
 
C
,
Sholler
 
GF
, et al.  
Sudden death in childhood cardiomyopathy: results from a long-term national population-based study
.
J Am Coll Cardiol
 
2015
;
65
:
2302
2310
. https://doi.org/10.1016/j.jacc.2015.03.552

779

McMahon
 
CJ
,
Nagueh
 
SF
,
Pignatelli
 
RH
,
Denfield
 
SW
,
Dreyer
 
WJ
,
Price
 
JF
, et al.  
Characterization of left ventricular diastolic function by tissue Doppler imaging and clinical status in children with hypertrophic cardiomyopathy
.
Circulation
 
2004
;
109
:
1756
1762
. https://doi.org/10.1161/01.CIR.0000124723.16433.31

780

Ostman-Smith
 
I
,
Wettrell
 
G
,
Keeton
 
B
,
Riesenfeld
 
T
,
Holmgren
 
D
,
Ergander
 
U
.
Echocardiographic and electrocardiographic identification of those children with hypertrophic cardiomyopathy who should be considered at high-risk of dying suddenly
.
Cardiol Young
 
2005
;
15
:
632
642
. https://doi.org/10.1017/S1047951105001824

781

Efthimiadis
 
GK
,
Parcharidou
 
DG
,
Giannakoulas
 
G
,
Pagourelias
 
ED
,
Charalampidis
 
P
,
Savvopoulos
 
G
, et al.  
Left ventricular outflow tract obstruction as a risk factor for sudden cardiac death in hypertrophic cardiomyopathy
.
Am J Cardiol
 
2009
;
104
:
695
699
. https://doi.org/10.1016/j.amjcard.2009.04.039

782

Elliott
 
PM
,
Poloniecki
 
J
,
Dickie
 
S
,
Sharma
 
S
,
Monserrat
 
L
,
Varnava
 
A
, et al.  
Sudden death in hypertrophic cardiomyopathy: identification of high risk patients
.
J Am Coll Cardiol
 
2000
;
36
:
2212
2218
. https://doi.org/10.1016/S0735-1097(00)01003-2

783

Louie
 
EK
,
Maron
 
BJ
.
Hypertrophic cardiomyopathy with extreme increase in left ventricular wall thickness: functional and morphologic features and clinical significance
.
J Am Coll Cardiol
 
1986
;
8
:
57
65
. https://doi.org/10.1016/S0735-1097(86)80092-4

784

Norrish
 
G
,
Ding
 
T
,
Field
 
E
,
Cervi
 
E
,
Ziolkowska
 
L
,
Olivotto
 
I
, et al.  
Relationship between maximal left ventricular wall thickness and sudden cardiac death in childhood onset hypertrophic cardiomyopathy
.
Circ Arrhythm Electrophysiol
 
2022
;
15
:
e010075
. https://doi.org/10.1161/CIRCEP.121.010075

785

Williams
 
L
,
Frenneaux
 
M
.
Syncope in hypertrophic cardiomyopathy: mechanisms and consequences for treatment
.
Europace
 
2007
;
9
:
817
822
. https://doi.org/10.1093/europace/eum093

786

Sediva
 
H
,
Hnat
 
T
,
Bonaventura
 
J
,
Slesarenko
 
J
,
Veselka
 
J
.
Head-up tilt test in risk stratification of patients with hypertrophic cardiomyopathy
.
Int J Angiol
 
2019
;
28
:
245
248
. https://doi.org/10.1055/s-0039-1688983

787

Moak
 
JP
,
Leifer
 
ES
,
Tripodi
 
D
,
Mohiddin
 
SA
,
Fananapazir
 
L
.
Long-term follow-up of children and adolescents diagnosed with hypertrophic cardiomyopathy: risk factors for adverse arrhythmic events
.
Pediatr Cardiol
 
2011
;
32
:
1096
1105
. https://doi.org/10.1007/s00246-011-9967-y

788

Romeo
 
F
,
Cianfrocca
 
C
,
Pelliccia
 
F
,
Colloridi
 
V
,
Cristofani
 
R
,
Reale
 
A
.
Long-term prognosis in children with hypertrophic cardiomyopathy: an analysis of 37 patients aged less than or equal to 14 years at diagnosis
.
Clin Cardiol
 
1990
;
13
:
101
107
. https://doi.org/10.1002/clc.4960130208

789

Maskatia
 
SA
,
Decker
 
JA
,
Spinner
 
JA
,
Kim
 
JJ
,
Price
 
JF
,
Jefferies
 
JL
, et al.  
Restrictive physiology is associated with poor outcomes in children with hypertrophic cardiomyopathy
.
Pediatr Cardiol
 
2012
;
33
:
141
149
. https://doi.org/10.1007/s00246-011-0106-6

790

Olivotto
 
I
,
Maron
 
MS
,
Adabag
 
AS
,
Casey
 
SA
,
Vargiu
 
D
,
Link
 
MS
, et al.  
Gender-related differences in the clinical presentation and outcome of hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2005
;
46
:
480
487
. https://doi.org/10.1016/j.jacc.2005.04.043

791

Yan
 
L-R
,
Zhao
 
S-H
,
Wang
 
H-Y
,
Duan
 
F-J
,
Wang
 
Z-M
,
Yang
 
Y-J
, et al.  
Clinical characteristics and prognosis of 60 patients with midventricular obstructive hypertrophic cardiomyopathy
.
J Cardiovasc Med (Hagerstown)
 
2015
;
16
:
751
760
. https://doi.org/10.2459/JCM.0000000000000163

792

Minami
 
Y
,
Haruki
 
S
,
Hagiwara
 
N
.
Phenotypic overlap in hypertrophic cardiomyopathy: apical hypertrophy, midventricular obstruction, and apical aneurysm
.
J Cardiol
 
2014
;
64
:
463
469
. https://doi.org/10.1016/j.jjcc.2014.03.003

793

Ommen
 
SR
,
Mital
 
S
,
Burke
 
MA
,
Day
 
SM
,
Deswal
 
A
,
Elliott
 
P
, et al.  
2020 AHA/ACC Guideline for the diagnosis and treatment of patients with hypertrophic cardiomyopathy: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines
.
Circulation
 
2020
;
142
:
e558
e631
. https://doi.org/10.1161/CIR.0000000000000937

794

Kamp
 
NJ
,
Chery
 
G
,
Kosinski
 
AS
,
Desai
 
MY
,
Wazni
 
O
,
Schmidler
 
GS
, et al.  
Risk stratification using late gadolinium enhancement on cardiac magnetic resonance imaging in patients with hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
Prog Cardiovasc Dis
 
2021
;
66
:
10
16
. https://doi.org/10.1016/j.pcad.2020.11.001

795

Kramer
 
CM
,
DiMarco
 
JP
,
Kolm
 
P
,
Ho
 
CY
,
Desai
 
MY
,
Kwong
 
RY
, et al.  
Predictors of major atrial fibrillation endpoints in the National Heart, Lung, and Blood Institute HCMR
.
JACC Clin Electrophysiol
 
2021
;
7
:
1376
1386
. https://doi.org/10.1016/j.jacep.2021.04.004

796

Raja
 
AA
,
Farhad
 
H
,
Valente
 
AM
,
Couce
 
JP
,
Jefferies
 
JL
,
Bundgaard
 
H
, et al.  
Prevalence and progression of late gadolinium enhancement in children and adolescents with hypertrophic cardiomyopathy
.
Circulation
 
2018
;
138
:
782
792
. https://doi.org/10.1161/CIRCULATIONAHA.117.032966

797

Petryka-Mazurkiewicz
 
J
,
Ziolkowska
 
L
,
Kowalczyk-Domagala
 
M
,
Mazurkiewicz
 
L
,
Boruc
 
A
,
Spiewak
 
M
, et al.  
LGE for risk stratification in primary prevention in children with HCM
.
JACC Cardiovasc Imaging
 
2020
;
13
:
2684
2686
. https://doi.org/10.1016/j.jcmg.2020.06.009

798

Frenneaux
 
MP
,
Counihan
 
PJ
,
Caforio
 
AL
,
Chikamori
 
T
,
McKenna
 
WJ
.
Abnormal blood pressure response during exercise in hypertrophic cardiomyopathy
.
Circulation
 
1990
;
82
:
1995
2002
. https://doi.org/10.1161/01.CIR.82.6.1995

799

Counihan
 
PJ
,
Frenneaux
 
MP
,
Webb
 
DJ
,
McKenna
 
WJ
.
Abnormal vascular responses to supine exercise in hypertrophic cardiomyopathy
.
Circulation
 
1991
;
84
:
686
696
. https://doi.org/10.1161/01.CIR.84.2.686

800

Sadoul
 
N
,
Prasad
 
K
,
Elliott
 
PM
,
Bannerjee
 
S
,
Frenneaux
 
MP
,
McKenna
 
WJ
.
Prospective prognostic assessment of blood pressure response during exercise in patients with hypertrophic cardiomyopathy
.
Circulation
 
1997
;
96
:
2987
2991
. https://doi.org/10.1161/01.CIR.96.9.2987

801

Smith
 
ED
,
Tome
 
J
,
McGrath
 
R
,
Kumar
 
S
,
Concannon
 
M
,
Day
 
SM
, et al.  
Exercise hemodynamics in hypertrophic cardiomyopathy identify risk of incident heart failure but not ventricular arrhythmias or sudden cardiac death
.
Int J Cardiol
 
2019
;
274
:
226
231
. https://doi.org/10.1016/j.ijcard.2018.07.110

802

Norrish
 
G
,
Cantarutti
 
N
,
Pissaridou
 
E
,
Ridout
 
DA
,
Limongelli
 
G
,
Elliott
 
PM
, et al.  
Risk factors for sudden cardiac death in childhood hypertrophic cardiomyopathy: a systematic review and meta-analysis
.
Eur J Prev Cardiol
 
2017
;
24
:
1220
1230
. https://doi.org/10.1177/2047487317702519

803

Watkins
 
H
,
Rosenzweig
 
A
,
Hwang
 
DS
,
Levi
 
T
,
McKenna
 
W
,
Seidman
 
CE
, et al.  
Characteristics and prognostic implications of myosin missense mutations in familial hypertrophic cardiomyopathy
.
N Engl J Med
 
1992
;
326
:
1108
1114
. https://doi.org/10.1056/NEJM199204233261703

804

Anan
 
R
,
Greve
 
G
,
Thierfelder
 
L
,
Watkins
 
H
,
McKenna
 
WJ
,
Solomon
 
S
, et al.  
Prognostic implications of novel beta cardiac myosin heavy chain gene mutations that cause familial hypertrophic cardiomyopathy
.
J Clin Invest
 
1994
;
93
:
280
285
. https://doi.org/10.1172/JCI116957

805

Moolman
 
JC
,
Corfield
 
VA
,
Posen
 
B
,
Ngumbela
 
K
,
Seidman
 
C
,
Brink
 
PA
, et al.  
Sudden death due to troponin T mutations
.
J Am Coll Cardiol
 
1997
;
29
:
549
555
. https://doi.org/10.1016/S0735-1097(96)00530-X

806

Ackerman
 
MJ
,
VanDriest
 
SL
,
Ommen
 
SR
,
Will
 
ML
,
Nishimura
 
RA
,
Tajik
 
AJ
, et al.  
Prevalence and age-dependence of malignant mutations in the beta-myosin heavy chain and troponin T genes in hypertrophic cardiomyopathy: a comprehensive outpatient perspective
.
J Am Coll Cardiol
 
2002
;
39
:
2042
2048
. https://doi.org/10.1016/S0735-1097(02)01900-9

807

Garcia-Giustiniani
 
D
,
Arad
 
M
,
Ortiz-Genga
 
M
,
Barriales-Villa
 
R
,
Fernandez
 
X
,
Rodriguez-Garcia
 
I
, et al.  
Phenotype and prognostic correlations of the converter region mutations affecting the beta myosin heavy chain
.
Heart
 
2015
;
101
:
1047
1053
. https://doi.org/10.1136/heartjnl-2014-307205

808

Van Driest
 
SL
,
Maron
 
BJ
,
Ackerman
 
MJ
.
From malignant mutations to malignant domains: the continuing search for prognostic significance in the mutant genes causing hypertrophic cardiomyopathy
.
Heart
 
2004
;
90
:
7
8
. https://doi.org/10.1136/heart.90.1.7

809

Fananapazir
 
L
,
Epstein
 
ND
.
Genotype-phenotype correlations in hypertrophic cardiomyopathy. Insights provided by comparisons of kindreds with distinct and identical beta-myosin heavy chain gene mutations
.
Circulation
 
1994
;
89
:
22
32
. https://doi.org/10.1161/01.CIR.89.1.22

810

Landstrom
 
AP
,
Ackerman
 
MJ
.
Mutation type is not clinically useful in predicting prognosis in hypertrophic cardiomyopathy
.
Circulation
 
2010
;
122
:
2441
2449
;
discussion 2450
. https://doi.org/10.1161/CIRCULATIONAHA.110.954446

811

Richard
 
P
,
Charron
 
P
,
Leclercq
 
C
,
Ledeuil
 
C
,
Carrier
 
L
,
Dubourg
 
O
, et al.  
Homozygotes for a R869G mutation in the beta-myosin heavy chain gene have a severe form of familial hypertrophic cardiomyopathy
.
J Mol Cell Cardiol
 
2000
;
32
:
1575
1583
. https://doi.org/10.1006/jmcc.2000.1193

812

Richard
 
P
,
Isnard
 
R
,
Carrier
 
L
,
Dubourg
 
O
,
Donatien
 
Y
,
Mathieu
 
B
, et al.  
Double heterozygosity for mutations in the beta-myosin heavy chain and in the cardiac myosin binding protein C genes in a family with hypertrophic cardiomyopathy
.
J Med Genet
 
1999
;
36
:
542
545
. https://doi.org/10.1136/jmg.36.7.542

813

Jeschke
 
B
,
Uhl
 
K
,
Weist
 
B
,
Schroder
 
D
,
Meitinger
 
T
,
Dohlemann
 
C
, et al.  
A high risk phenotype of hypertrophic cardiomyopathy associated with a compound genotype of two mutated beta-myosin heavy chain genes
.
Hum Genet
 
1998
;
102
:
299
304
. https://doi.org/10.1007/s004390050695

814

Kaski
 
JP
,
Syrris
 
P
,
Esteban
 
MT
,
Jenkins
 
S
,
Pantazis
 
A
,
Deanfield
 
JE
, et al.  
Prevalence of sarcomere protein gene mutations in preadolescent children with hypertrophic cardiomyopathy
.
Circ Cardiovasc Genet
 
2009
;
2
:
436
441
. https://doi.org/10.1161/CIRCGENETICS.108.821314

815

Morita
 
H
,
Rehm
 
HL
,
Menesses
 
A
,
McDonough
 
B
,
Roberts
 
AE
,
Kucherlapati
 
R
, et al.  
Shared genetic causes of cardiac hypertrophy in children and adults
.
N Engl J Med
 
2008
;
358
:
1899
1908
. https://doi.org/10.1056/NEJMoa075463

816

Lopes
 
LR
,
Syrris
 
P
,
Guttmann
 
OP
,
O’Mahony
 
C
,
Tang
 
HC
,
Dalageorgou
 
C
, et al.  
Novel genotype-phenotype associations demonstrated by high-throughput sequencing in patients with hypertrophic cardiomyopathy
.
Heart
 
2015
;
101
:
294
301
. https://doi.org/10.1136/heartjnl-2014-306387

817

van Velzen
 
HG
,
Vriesendorp
 
PA
,
Oldenburg
 
RA
,
van Slegtenhorst
 
MA
,
van der Velden
 
J
,
Schinkel
 
AFL
, et al.  
Value of genetic testing for the prediction of long-term outcome in patients with hypertrophic cardiomyopathy
.
Am J Cardiol
 
2016
;
118
:
881
887
. https://doi.org/10.1016/j.amjcard.2016.06.038

818

McKenna
 
WJ
,
Oakley
 
CM
,
Krikler
 
DM
,
Goodwin
 
JF
.
Improved survival with amiodarone in patients with hypertrophic cardiomyopathy and ventricular tachycardia
.
Br Heart J
 
1985
;
53
:
412
416
. https://doi.org/10.1136/hrt.53.4.412

819

Melacini
 
P
,
Maron
 
BJ
,
Bobbo
 
F
,
Basso
 
C
,
Tokajuk
 
B
,
Zucchetto
 
M
, et al.  
Evidence that pharmacological strategies lack efficacy for the prevention of sudden death in hypertrophic cardiomyopathy
.
Heart
 
2007
;
93
:
708
710
. https://doi.org/10.1136/hrt.2006.099416

820

Zipes
 
DP
,
Camm
 
AJ
,
Borggrefe
 
M
,
Buxton
 
AE
,
Chaitman
 
B
,
Fromer
 
M
, et al.  
ACC/AHA/ESC 2006 guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: a report of the American College of Cardiology/American Heart Association Task Force and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death) developed in collaboration with the European Heart Rhythm Association and the Heart Rhythm Society
.
Europace
 
2006
;
8
:
746
837
. https://doi.org/10.1093/europace/eul108

821

Vriesendorp
 
PA
,
Schinkel
 
AF
,
Liebregts
 
M
,
Theuns
 
DA
,
van Cleemput
 
J
,
Ten Cate
 
FJ
, et al.  
Validation of the 2014 European Society of Cardiology guidelines risk prediction model for the primary prevention of sudden cardiac death in hypertrophic cardiomyopathy
.
Circ Arrhythm Electrophysiol
 
2015
;
8
:
829
835
. https://doi.org/10.1161/CIRCEP.114.002553

822

Choi
 
Y-J
,
Kim
 
H-K
,
Lee
 
SC
,
Park
 
J-B
,
Moon
 
I
,
Park
 
J
, et al.  
Validation of the hypertrophic cardiomyopathy risk-sudden cardiac death calculator in Asians
.
Heart
 
2019
;
105
:
1892
1897
. https://doi.org/10.1136/heartjnl-2019-315160

823

O’Mahony
 
C
,
Jichi
 
F
,
Ommen
 
SR
,
Christiaans
 
I
,
Arbustini
 
E
,
Garcia-Pavia
 
P
, et al.  
International external validation study of the 2014 European Society of Cardiology Guidelines on sudden cardiac death prevention in hypertrophic cardiomyopathy (EVIDENCE-HCM)
.
Circulation
 
2018
;
137
:
1015
1023
. https://doi.org/10.1161/CIRCULATIONAHA.117.030437

824

Fernandez
 
A
,
Quiroga
 
A
,
Ochoa
 
JP
,
Mysuta
 
M
,
Casabe
 
JH
,
Biagetti
 
M
, et al.  
Validation of the 2014 European Society of Cardiology sudden cardiac death risk prediction model in hypertrophic cardiomyopathy in a reference center in South America
.
Am J Cardiol
 
2016
;
118
:
121
126
. https://doi.org/10.1016/j.amjcard.2016.04.021

825

Norrish
 
G
,
Ding
 
T
,
Field
 
E
,
McLeod
 
K
,
Ilina
 
M
,
Stuart
 
G
, et al.  
A validation study of the European Society of Cardiology guidelines for risk stratification of sudden cardiac death in childhood hypertrophic cardiomyopathy
.
Europace
 
2019
;
21
:
1559
1565
. https://doi.org/10.1093/europace/euz118

826

Ostman-Smith
 
I
,
Sjoberg
 
G
,
Alenius Dahlqvist
 
J
,
Larsson
 
P
,
Fernlund
 
E
.
Sudden cardiac death in childhood hypertrophic cardiomyopathy is best predicted by a combination of electrocardiogram risk-score and HCMRisk-Kids score
.
Acta Paediatr
 
2021
;
110
:
3105
3115
. https://doi.org/10.1111/apa.16045

827

Magnusson
 
P
,
Gadler
 
F
,
Liv
 
P
,
Morner
 
S
.
Hypertrophic cardiomyopathy and implantable defibrillators in Sweden: inappropriate shocks and complications requiring surgery
.
J Cardiovasc Electrophysiol
 
2015
;
26
:
1088
1094
. https://doi.org/10.1111/jce.12750

828

Norrish
 
G
,
Chubb
 
H
,
Field
 
E
,
McLeod
 
K
,
Ilina
 
M
,
Spentzou
 
G
, et al.  
Clinical outcomes and programming strategies of implantable cardioverter-defibrillator devices in paediatric hypertrophic cardiomyopathy: a UK National Cohort Study
.
Europace
 
2021
;
23
:
400
408
. https://doi.org/10.1093/europace/euaa307

829

Liebregts
 
M
,
Faber
 
L
,
Jensen
 
MK
,
Vriesendorp
 
PA
,
Hansen
 
PR
,
Seggewiss
 
H
, et al.  
Validation of the HCM Risk-SCD model in patients with hypertrophic cardiomyopathy following alcohol septal ablation
.
Europace
 
2018
;
20
:
f198
f203
. https://doi.org/10.1093/europace/eux251

830

Veselka
 
J
,
Liebregts
 
M
,
Cooper
 
R
,
Faber
 
L
,
Januska
 
J
,
Kashtanov
 
M
, et al.  
Prediction of sudden cardiac arrest after alcohol septal ablation for hypertrophic obstructive cardiomyopathy: ASA-SCARRE risk score
.
Am J Cardiol
 
2022
;
184
:
120
126
. https://doi.org/10.1016/j.amjcard.2022.08.028

831

Maron
 
BJ
,
Spirito
 
P
,
Shen
 
WK
,
Haas
 
TS
,
Formisano
 
F
,
Link
 
MS
, et al.  
Implantable cardioverter-defibrillators and prevention of sudden cardiac death in hypertrophic cardiomyopathy
.
JAMA
 
2007
;
298
:
405
412
. https://doi.org/10.1001/jama.298.4.405

832

Syska
 
P
,
Przybylski
 
A
,
Chojnowska
 
L
,
Lewandowski
 
M
,
Sterlinski
 
M
,
Maciag
 
A
, et al.  
Implantable cardioverter-defibrillator in patients with hypertrophic cardiomyopathy: efficacy and complications of the therapy in long-term follow-up
.
J Cardiovasc Electrophysiol
 
2010
;
21
:
883
889
. https://doi.org/10.1111/j.1540-8167.2009.01716.x

833

Norrish
 
G
,
Qu
 
C
,
Field
 
E
,
Cervi
 
E
,
Khraiche
 
D
,
Klaassen
 
S
, et al.  
External validation of the HCM Risk-Kids model for predicting sudden cardiac death in childhood hypertrophic cardiomyopathy
.
Eur J Prev Cardiol
 
2022
;
29
:
678
686
. https://doi.org/10.1093/eurjpc/zwab181

834

Maron
 
MS
,
Appelbaum
 
E
,
Harrigan
 
CJ
,
Buros
 
J
,
Gibson
 
CM
,
Hanna
 
C
, et al.  
Clinical profile and significance of delayed enhancement in hypertrophic cardiomyopathy
.
Circ Heart Fail
 
2008
;
1
:
184
191
. https://doi.org/10.1161/CIRCHEARTFAILURE.108.768119

835

Bruder
 
O
,
Wagner
 
A
,
Jensen
 
CJ
,
Schneider
 
S
,
Ong
 
P
,
Kispert
 
EM
, et al.  
Myocardial scar visualized by cardiovascular magnetic resonance imaging predicts major adverse events in patients with hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2010
;
56
:
875
887
. https://doi.org/10.1016/j.jacc.2010.05.007

836

O’Hanlon
 
R
,
Grasso
 
A
,
Roughton
 
M
,
Moon
 
JC
,
Clark
 
S
,
Wage
 
R
, et al.  
Prognostic significance of myocardial fibrosis in hypertrophic cardiomyopathy
.
J Am Coll Cardiol
 
2010
;
56
:
867
874
. https://doi.org/10.1016/j.jacc.2010.05.010

837

Green
 
JJ
,
Berger
 
JS
,
Kramer
 
CM
,
Salerno
 
M
.
Prognostic value of late gadolinium enhancement in clinical outcomes for hypertrophic cardiomyopathy
.
JACC Cardiovasc Imaging
 
2012
;
5
:
370
377
. https://doi.org/10.1016/j.jcmg.2011.11.021

838

Ismail
 
TF
,
Jabbour
 
A
,
Gulati
 
A
,
Mallorie
 
A
,
Raza
 
S
,
Cowling
 
TE
, et al.  
Role of late gadolinium enhancement cardiovascular magnetic resonance in the risk stratification of hypertrophic cardiomyopathy
.
Heart
 
2014
;
100
:
1851
1858
. https://doi.org/10.1136/heartjnl-2013-305471

839

Briasoulis
 
A
,
Mallikethi-Reddy
 
S
,
Palla
 
M
,
Alesh
 
I
,
Afonso
 
L
.
Myocardial fibrosis on cardiac magnetic resonance and cardiac outcomes in hypertrophic cardiomyopathy: a meta-analysis
.
Heart
 
2015
;
101
:
1406
1411
. https://doi.org/10.1136/heartjnl-2015-307682

840

Mentias
 
A
,
Raeisi-Giglou
 
P
,
Smedira
 
NG
,
Feng
 
K
,
Sato
 
K
,
Wazni
 
O
, et al.  
Late gadolinium enhancement in patients with hypertrophic cardiomyopathy and preserved systolic function
.
J Am Coll Cardiol
 
2018
;
72
:
857
870
. https://doi.org/10.1016/j.jacc.2018.05.060

841

Rowin
 
EJ
,
Maron
 
MS
,
Adler
 
A
,
Albano
 
AJ
,
Varnava
 
AM
,
Spears
 
D
, et al.  
Importance of newer cardiac magnetic resonance-based risk markers for sudden death prevention in hypertrophic cardiomyopathy: an international multicenter study
.
Heart Rhythm
 
2022
;
19
:
782
789
. https://doi.org/10.1016/j.hrthm.2021.12.017

842

Rowin
 
EJ
,
Maron
 
BJ
,
Carrick
 
RT
,
Patel
 
PP
,
Koethe
 
B
,
Wells
 
S
, et al.  
Outcomes in patients with hypertrophic cardiomyopathy and left ventricular systolic dysfunction
.
J Am Coll Cardiol
 
2020
;
75
:
3033
3043
. https://doi.org/10.1016/j.jacc.2020.04.045

843

Thaman
 
R
,
Gimeno
 
JR
,
Murphy
 
RT
,
Kubo
 
T
,
Sachdev
 
B
,
Mogensen
 
J
, et al.  
Prevalence and clinical significance of systolic impairment in hypertrophic cardiomyopathy
.
Heart
 
2005
;
91
:
920
925
. https://doi.org/10.1136/hrt.2003.031161

844

Kawarai
 
H
,
Kajimoto
 
K
,
Minami
 
Y
,
Hagiwara
 
N
,
Kasanuki
 
H
.
Risk of sudden death in end-stage hypertrophic cardiomyopathy
.
J Card Fail
 
2011
;
17
:
459
464
. https://doi.org/10.1016/j.cardfail.2011.01.015

845

Pettersen
 
MD
,
Du
 
W
,
Skeens
 
ME
,
Humes
 
RA
.
Regression equations for calculation of z scores of cardiac structures in a large cohort of healthy infants, children, and adolescents: an echocardiographic study
.
J Am Soc Echocardiogr
 
2008
;
21
:
922
934
. https://doi.org/10.1016/j.echo.2008.02.006

846

Chubb
 
H
,
Simpson
 
JM
.
The use of Z-scores in paediatric cardiology
.
Ann Pediatr Cardiol
 
2012
;
5
:
179
184
. https://doi.org/10.4103/0974-2069.99622

847

Sepehrkhouy
 
S
,
Gho
 
J
,
van Es
 
R
,
Harakalova
 
M
,
de Jonge
 
N
,
Dooijes
 
D
, et al.  
Distinct fibrosis pattern in desmosomal and phospholamban mutation carriers in hereditary cardiomyopathies
.
Heart Rhythm
 
2017
;
14
:
1024
1032
. https://doi.org/10.1016/j.hrthm.2017.03.034

848

Chen
 
W
,
Qian
 
W
,
Zhang
 
X
,
Li
 
D
,
Qian
 
Z
,
Xu
 
H
, et al.  
Ring-like late gadolinium enhancement for predicting ventricular tachyarrhythmias in non-ischaemic dilated cardiomyopathy
.
Eur Heart J Cardiovasc Imaging
 
2021
;
22
:
1130
1138
. https://doi.org/10.1093/ehjci/jeab117

849

Writing Group, Document Reading Group, EACVI Reviewers: this document was reviewed by members of the EACVI Scientific Documents Committee for 2014–2016 and 2016–2018
.
A joint procedural position statement on imaging in cardiac sarcoidosis: from the Cardiovascular and Inflammation & Infection Committees of the European Association of Nuclear Medicine, the European Association of Cardiovascular Imaging, and the American Society of Nuclear Cardiology
.
Eur Heart J Cardiovasc Imaging
 
2017
;
18
:
1073
1089
. https://doi.org/10.1093/ehjci/jex146

850

Fatkin
 
D
,
Huttner
 
IG
,
Kovacic
 
JC
,
Seidman
 
JG
,
Seidman
 
CE
.
Precision medicine in the management of dilated cardiomyopathy: JACC state-of-the-art review
.
J Am Coll Cardiol
 
2019
;
74
:
2921
2938
. https://doi.org/10.1016/j.jacc.2019.10.011

851

Merlo
 
M
,
Cannata
 
A
,
Sinagra
 
G
.
Dilated cardiomyopathy: a paradigm of revolution in medicine
.
J Clin Med
 
2020
;
9
:
3385
. https://doi.org/10.3390/jcm9113385

852

Seferovic
 
PM
,
Polovina
 
M
,
Bauersachs
 
J
,
Arad
 
M
,
Gal
 
TB
,
Lund
 
LH
, et al.  
Heart failure in cardiomyopathies: a position paper from the Heart Failure Association of the European Society of Cardiology
.
Eur J Heart Fail
 
2019
;
21
:
553
576
. https://doi.org/10.1002/ejhf.1461

853

Verdonschot
 
JAJ
,
Hazebroek
 
MR
,
Krapels
 
IPC
,
Henkens
 
M
,
Raafs
 
A
,
Wang
 
P
, et al.  
Implications of genetic testing in dilated cardiomyopathy
.
Circ Genom Precis Med
 
2020
;
13
:
476
487
. https://doi.org/10.1161/CIRCGEN.120.003031

854

Hey
 
TM
,
Rasmussen
 
TB
,
Madsen
 
T
,
Aagaard
 
MM
,
Harbo
 
M
,
Molgaard
 
H
, et al.  
Clinical and genetic investigations of 109 index patients with dilated cardiomyopathy and 445 of their relatives
.
Circ Heart Fail
 
2020
;
13
:
e006701
. https://doi.org/10.1161/CIRCHEARTFAILURE.119.006701

855

Cuenca
 
S
,
Ruiz-Cano
 
MJ
,
Gimeno-Blanes
 
JR
,
Jurado
 
A
,
Salas
 
C
,
Gomez-Diaz
 
I
, et al.  
Genetic basis of familial dilated cardiomyopathy patients undergoing heart transplantation
.
J Heart Lung Transplant
 
2016
;
35
:
625
635
. https://doi.org/10.1016/j.healun.2015.12.014

856

van der Meulen
 
MH
,
Herkert
 
JC
,
den Boer
 
SL
,
du Marchie Sarvaas
 
GJ
,
Blom
 
N
,
Ten Harkel
 
ADJ
, et al.  
Genetic evaluation of a nation-wide Dutch pediatric DCM cohort: the use of genetic testing in risk stratification
.
Circ Genom Precis Med
 
2022
;
15
:
e002981
. https://doi.org/10.1161/CIRCGEN.120.002981

857

Ware
 
SM
,
Wilkinson
 
JD
,
Tariq
 
M
,
Schubert
 
JA
,
Sridhar
 
A
,
Colan
 
SD
, et al.  
Genetic causes of cardiomyopathy in children: first results from the pediatric cardiomyopathy genes study
.
J Am Heart Assoc
 
2021
;
10
:
e017731
. https://doi.org/10.1161/JAHA.120.017731

858

Escobar-Lopez
 
L
,
Ochoa
 
JP
,
Royuela
 
A
,
Verdonschot
 
JAJ
,
Dal Ferro
 
M
,
Espinosa
 
MA
, et al.  
Clinical risk score to predict pathogenic genotypes in patients with dilated cardiomyopathy
.
J Am Coll Cardiol
 
2022
;
80
:
1115
1126
. https://doi.org/10.1016/j.jacc.2022.06.040

859

Cannata
 
A
,
Merlo
 
M
,
Dal Ferro
 
M
,
Barbati
 
G
,
Manca
 
P
,
Paldino
 
A
, et al.  
Association of titin variations with late-onset dilated cardiomyopathy
.
JAMA Cardiol
 
2022
;
7
:
371
377
. https://doi.org/10.1001/jamacardio.2021.5890

860

Bardy
 
GH
,
Lee
 
KL
,
Mark
 
DB
,
Poole
 
JE
,
Packer
 
DL
,
Boineau
 
R
, et al.  
Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure
.
N Engl J Med
 
2005
;
352
:
225
237
. https://doi.org/10.1056/NEJMoa043399

861

Kober
 
L
,
Thune
 
JJ
,
Nielsen
 
JC
,
Haarbo
 
J
,
Videbaek
 
L
,
Korup
 
E
, et al.  
Defibrillator implantation in patients with nonischemic systolic heart failure
.
N Engl J Med
 
2016
;
375
:
1221
1230
. https://doi.org/10.1056/NEJMoa1608029

862

Elming
 
MB
,
Nielsen
 
JC
,
Haarbo
 
J
,
Videbaek
 
L
,
Korup
 
E
,
Signorovitch
 
J
, et al.  
Age and outcomes of primary prevention implantable cardioverter-defibrillators in patients with nonischemic systolic heart failure
.
Circulation
 
2017
;
136
:
1772
1780
. https://doi.org/10.1161/CIRCULATIONAHA.117.028829

863

Alba
 
AC
,
Foroutan
 
F
,
Duero Posada
 
J
,
Battioni
 
L
,
Schofield
 
T
,
Alhussein
 
M
, et al.  
Implantable cardiac defibrillator and mortality in non-ischaemic cardiomyopathy: an updated meta-analysis
.
Heart
 
2018
;
104
:
230
236
. https://doi.org/10.1136/heartjnl-2017-311430

864

Smith
 
ED
,
Lakdawala
 
NK
,
Papoutsidakis
 
N
,
Aubert
 
G
,
Mazzanti
 
A
,
McCanta
 
AC
, et al.  
Desmoplakin cardiomyopathy, a fibrotic and inflammatory form of cardiomyopathy distinct from typical dilated or arrhythmogenic right ventricular cardiomyopathy
.
Circulation
 
2020
;
141
:
1872
1884
. https://doi.org/10.1161/CIRCULATIONAHA.119.044934

865

Barriales-Villa
 
R
,
Ochoa
 
JP
,
Larranaga-Moreira
 
JM
,
Salazar-Mendiguchia
 
J
,
Diez-Lopez
 
C
,
Restrepo-Cordoba
 
MA
, et al.  
Risk predictors in a Spanish cohort with cardiac laminopathies. The REDLAMINA registry
.
Rev Esp Cardiol (Engl Ed)
 
2021
;
74
:
216
224
. https://doi.org/10.1016/j.recesp.2020.03.002

866

Gigli
 
M
,
Stolfo
 
D
,
Graw
 
SL
,
Merlo
 
M
,
Gregorio
 
C
,
Nee Chen
 
S
, et al.  
Phenotypic expression, natural history, and risk stratification of cardiomyopathy caused by filamin C truncating variants
.
Circulation
 
2021
;
144
:
1600
1611
. https://doi.org/10.1161/CIRCULATIONAHA.121.053521

867

Akhtar
 
MM
,
Lorenzini
 
M
,
Pavlou
 
M
,
Ochoa
 
JP
,
O’Mahony
 
C
,
Restrepo-Cordoba
 
MA
, et al.  
Association of left ventricular systolic dysfunction among carriers of truncating variants in filamin C with frequent ventricular arrhythmia and end-stage heart failure
.
JAMA Cardiol
 
2021
;
6
:
891
901
. https://doi.org/10.1001/jamacardio.2021.1106

868

Hodgkinson
 
KA
,
Howes
 
AJ
,
Boland
 
P
,
Shen
 
XS
,
Stuckless
 
S
,
Young
 
T-L
, et al.  
Long-term clinical outcome of arrhythmogenic right ventricular cardiomyopathy in individuals with a p.S358L mutation in TMEM43 following implantable cardioverter defibrillator therapy
.
Circ Arrhythm Electrophysiol
 
2016
;
9
:
e003589
. https://doi.org/10.1161/CIRCEP.115.003589

869

Hey
 
TM
,
Rasmussen
 
TB
,
Madsen
 
T
,
Aagaard
 
MM
,
Harbo
 
M
,
Molgaard
 
H
, et al.  
Pathogenic RBM20-variants are associated with a severe disease expression in male patients with dilated cardiomyopathy
.
Circ Heart Fail
 
2019
;
12
:
e005700
. https://doi.org/10.1161/CIRCHEARTFAILURE.118.005700

870

Ebert
 
M
,
Wijnmaalen
 
AP
,
de Riva
 
M
,
Trines
 
SA
,
Androulakis
 
AFA
,
Glashan
 
CA
, et al.  
Prevalence and prognostic impact of pathogenic variants in patients with dilated cardiomyopathy referred for ventricular tachycardia ablation
.
JACC Clin Electrophysiol
 
2020
;
6
:
1103
1114
. https://doi.org/10.1016/j.jacep.2020.04.025

871

Rootwelt-Norberg
 
C
,
Christensen
 
AH
,
Skjolsvik
 
ET
,
Chivulescu
 
M
,
Vissing
 
CR
,
Bundgaard
 
H
, et al.  
Timing of cardioverter-defibrillator implantation in patients with cardiac laminopathies–external validation of the LMNA-risk ventricular tachyarrhythmia calculator
.
Heart Rhythm
 
2023
;
20
:
423
429
. https://doi.org/10.1016/j.hrthm.2022.11.024

872

Akhtar
 
MM
,
Lorenzini
 
M
,
Cicerchia
 
M
,
Ochoa
 
JP
,
Hey
 
TM
,
Sabater Molina
 
M
, et al.  
Clinical phenotypes and prognosis of dilated cardiomyopathy caused by truncating variants in the TTN gene
.
Circ Heart Fail
 
2020
;
13
:
e006832
. https://doi.org/10.1161/CIRCHEARTFAILURE.119.006832

873

Mirelis
 
JG
,
Escobar-Lopez
 
L
,
Ochoa
 
JP
,
Espinosa
 
MA
,
Villacorta
 
E
,
Navarro
 
M
, et al.  
Combination of late gadolinium enhancement and genotype improves prediction of prognosis in non-ischaemic dilated cardiomyopathy
.
Eur J Heart Fail
 
2022
;
24
:
1183
1196
. https://doi.org/10.1002/ejhf.2514

874

Di Marco
 
A
,
Brown
 
PF
,
Bradley
 
J
,
Nucifora
 
G
,
Claver
 
E
,
de Frutos
 
F
, et al.  
Improved risk stratification for ventricular arrhythmias and sudden death in patients with nonischemic dilated cardiomyopathy
.
J Am Coll Cardiol
 
2021
;
77
:
2890
2905
. https://doi.org/10.1016/j.jacc.2021.04.030

875

Brilakis
 
ES
,
Shen
 
WK
,
Hammill
 
SC
,
Hodge
 
DO
,
Rea
 
RF
,
Lexvold
 
NY
, et al.  
Role of programmed ventricular stimulation and implantable cardioverter defibrillators in patients with idiopathic dilated cardiomyopathy and syncope
.
Pacing Clin Electrophysiol
 
2001
;
24
:
1623
1630
. https://doi.org/10.1046/j.1460-9592.2001.01623.x

876

Merino
 
JL
,
Carmona
 
JR
,
Fernandez-Lozano
 
I
,
Peinado
 
R
,
Basterra
 
N
,
Sobrino
 
JA
.
Mechanisms of sustained ventricular tachycardia in myotonic dystrophy: implications for catheter ablation
.
Circulation
 
1998
;
98
:
541
546
. https://doi.org/10.1161/01.CIR.98.6.541

877

Russo
 
V
,
Papa
 
AA
,
Rago
 
A
,
Ciardiello
 
C
,
Martino
 
AM
,
Stazi
 
A
, et al.  
Arrhythmic CArdiac DEath in MYotonic dystrophy type 1 patients (ACADEMY 1) study: the predictive role of programmed ventricular stimulation
.
Europace
 
2022
;
24
:
1148
1155
. https://doi.org/10.1093/europace/euab282

878

van Rijsingen
 
IA
,
Arbustini
 
E
,
Elliott
 
PM
,
Mogensen
 
J
,
Hermans-van Ast
 
JF
,
van der Kooi
 
AJ
, et al.  
Risk factors for malignant ventricular arrhythmias in lamin a/c mutation carriers a European cohort study
.
J Am Coll Cardiol
 
2012
;
59
:
493
500
. https://doi.org/10.1016/j.jacc.2011.08.078

879

Thuillot
 
M
,
Maupain
 
C
,
Gandjbakhch
 
E
,
Waintraub
 
X
,
Hidden-Lucet
 
F
,
Isnard
 
R
, et al.  
External validation of risk factors for malignant ventricular arrhythmias in lamin A/C mutation carriers
.
Eur J Heart Fail
 
2019
;
21
:
253
254
. https://doi.org/10.1002/ejhf.1384

880

Ader
 
F
,
De Groote
 
P
,
Reant
 
P
,
Rooryck-Thambo
 
C
,
Dupin-Deguine
 
D
,
Rambaud
 
C
, et al.  
FLNC pathogenic variants in patients with cardiomyopathies: prevalence and genotype-phenotype correlations
.
Clin Genet
 
2019
;
96
:
317
329
. https://doi.org/10.1111/cge.13594

881

Hodgkinson
 
KA
,
Connors
 
SP
,
Merner
 
N
,
Haywood
 
A
,
Young
 
T-L
,
McKenna
 
WJ
, et al.  
The natural history of a genetic subtype of arrhythmogenic right ventricular cardiomyopathy caused by a p.S358L mutation in TMEM43
.
Clin Genet
 
2013
;
83
:
321
331
. https://doi.org/10.1111/j.1399-0004.2012.01919.x

882

van der Zwaag
 
PA
,
van Rijsingen
 
IA
,
Asimaki
 
A
,
Jongbloed
 
JD
,
van Veldhuisen
 
DJ
,
Wiesfeld
 
AC
, et al.  
Phospholamban R14del mutation in patients diagnosed with dilated cardiomyopathy or arrhythmogenic right ventricular cardiomyopathy: evidence supporting the concept of arrhythmogenic cardiomyopathy
.
Eur J Heart Fail
 
2012
;
14
:
1199
1207
. https://doi.org/10.1093/eurjhf/hfs119

883

van Rijsingen
 
IA
,
van der Zwaag
 
PA
,
Groeneweg
 
JA
,
Nannenberg
 
EA
,
Jongbloed
 
JD
,
Zwinderman
 
AH
, et al.  
Outcome in phospholamban R14del carriers: results of a large multicentre cohort study
.
Circ Cardiovasc Genet
 
2014
;
7
:
455
465
. https://doi.org/10.1161/CIRCGENETICS.113.000374

884

Hallstrom
 
AP
,
Greene
 
HL
,
Wyse
 
DG
,
Zipes
 
D
,
Epstein
 
AE
,
Domanski
 
MJ
, et al.  
Antiarrhythmics Versus Implantable Defibrillators (AVID)–rationale, design, and methods
.
Am J Cardiol
 
1995
;
75
:
470
475
. https://doi.org/10.1016/S0002-9149(99)80583-9

885

Beggs
 
SAS
,
Jhund
 
PS
,
Jackson
 
CE
,
McMurray
 
JJV
,
Gardner
 
RS
.
Non-ischaemic cardiomyopathy, sudden death and implantable defibrillators: a review and meta-analysis
.
Heart
 
2018
;
104
:
144
150
. https://doi.org/10.1136/heartjnl-2016-310850

886

Jorda
 
P
,
Toro
 
R
,
Diez
 
C
,
Salazar-Mendiguchia
 
J
,
Fernandez-Falgueras
 
A
,
Perez-Serra
 
A
, et al.  
Malignant arrhythmogenic role associated with RBM20: a comprehensive interpretation focused on a personalized approach
.
J Pers Med
 
2021
;
11
:
130
. https://doi.org/10.3390/jpm11020130

887

Hasselberg
 
NE
,
Edvardsen
 
T
,
Petri
 
H
,
Berge
 
KE
,
Leren
 
TP
,
Bundgaard
 
H
, et al.  
Risk prediction of ventricular arrhythmias and myocardial function in Lamin A/C mutation positive subjects
.
Europace
 
2014
;
16
:
563
571
. https://doi.org/10.1093/europace/eut291

888

Protonotarios
 
A
,
Wicks
 
E
,
Ashworth
 
M
,
Stephenson
 
E
,
Guttmann
 
O
,
Savvatis
 
K
, et al.  
Prevalence of (18)F-fluorodeoxyglucose positron emission tomography abnormalities in patients with arrhythmogenic right ventricular cardiomyopathy
.
Int J Cardiol
 
2019
;
284
:
99
104
. https://doi.org/10.1016/j.ijcard.2018.10.083

889

Casella
 
M
,
Gasperetti
 
A
,
Sicuso
 
R
,
Conte
 
E
,
Catto
 
V
,
Sommariva
 
E
, et al.  
Characteristics of patients with arrhythmogenic left ventricular cardiomyopathy: combining genetic and histopathologic findings
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e009005
. https://doi.org/10.1161/CIRCEP.120.009005

890

Thiene
 
G
,
Nava
 
A
,
Corrado
 
D
,
Rossi
 
L
,
Pennelli
 
N
.
Right ventricular cardiomyopathy and sudden death in young people
.
N Engl J Med
 
1988
;
318
:
129
133
. https://doi.org/10.1056/NEJM198801213180301

891

Protonotarios
 
A
,
Anastasakis
 
A
,
Panagiotakos
 
DB
,
Antoniades
 
L
,
Syrris
 
P
,
Vouliotis
 
A
, et al.  
Arrhythmic risk assessment in genotyped families with arrhythmogenic right ventricular cardiomyopathy
.
Europace
 
2016
;
18
:
610
616
. https://doi.org/10.1093/europace/euv061

892

Corrado
 
D
,
van Tintelen
 
PJ
,
McKenna
 
WJ
,
Hauer
 
RNW
,
Anastastakis
 
A
,
Asimaki
 
A
, et al.  
Arrhythmogenic right ventricular cardiomyopathy: evaluation of the current diagnostic criteria and differential diagnosis
.
Eur Heart J
 
2020
;
41
:
1414
1429
. https://doi.org/10.1093/eurheartj/ehz669

893

Cipriani
 
A
,
Bauce
 
B
,
De Lazzari
 
M
,
Rigato
 
I
,
Bariani
 
R
,
Meneghin
 
S
, et al.  
Arrhythmogenic right ventricular cardiomyopathy: characterization of left ventricular phenotype and differential diagnosis with dilated cardiomyopathy
.
J Am Heart Assoc
 
2020
;
9
:
e014628
. https://doi.org/10.1161/JAHA.119.014628

894

Sen-Chowdhry
 
S
,
Syrris
 
P
,
Prasad
 
SK
,
Hughes
 
SE
,
Merrifield
 
R
,
Ward
 
D
, et al.  
Left-dominant arrhythmogenic cardiomyopathy: an under-recognized clinical entity
.
J Am Coll Cardiol
 
2008
;
52
:
2175
2187
. https://doi.org/10.1016/j.jacc.2008.09.019

895

Bhonsale
 
A
,
Groeneweg
 
JA
,
James
 
CA
,
Dooijes
 
D
,
Tichnell
 
C
,
Jongbloed
 
JD
, et al.  
Impact of genotype on clinical course in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated mutation carriers
.
Eur Heart J
 
2015
;
36
:
847
855
. https://doi.org/10.1093/eurheartj/ehu509

896

Hermida
 
A
,
Fressart
 
V
,
Hidden-Lucet
 
F
,
Donal
 
E
,
Probst
 
V
,
Deharo
 
JC
, et al.  
High risk of heart failure associated with desmoglein-2 mutations compared to plakophilin-2 mutations in arrhythmogenic right ventricular cardiomyopathy/dysplasia
.
Eur J Heart Fail
 
2019
;
21
:
792
800
. https://doi.org/10.1002/ejhf.1423

897

DeWitt
 
ES
,
Chandler
 
SF
,
Hylind
 
RJ
,
Beausejour Ladouceur
 
V
,
Blume
 
ED
,
VanderPluym
 
C
, et al.  
Phenotypic manifestations of arrhythmogenic cardiomyopathy in children and adolescents
.
J Am Coll Cardiol
 
2019
;
74
:
346
358
. https://doi.org/10.1016/j.jacc.2019.05.022

898

Protonotarios
 
A
,
Elliott
 
PM
.
Arrhythmogenic right ventricular cardiomyopathy as a hidden cause of paediatric myocarditis presentation
.
Int J Cardiol
 
2018
;
271
:
113
114
. https://doi.org/10.1016/j.ijcard.2018.06.117

899

Bosman
 
LP
,
Cadrin-Tourigny
 
J
,
Bourfiss
 
M
,
Aliyari Ghasabeh
 
M
,
Sharma
 
A
,
Tichnell
 
C
, et al.  
Diagnosing arrhythmogenic right ventricular cardiomyopathy by 2010 Task Force Criteria: clinical performance and simplified practical implementation
.
Europace
 
2020
;
22
:
787
796
. https://doi.org/10.1093/europace/euaa039

900

Platonov
 
PG
,
Calkins
 
H
,
Hauer
 
RN
,
Corrado
 
D
,
Svendsen
 
JH
,
Wichter
 
T
, et al.  
High interobserver variability in the assessment of epsilon waves: implications for diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia
.
Heart Rhythm
 
2016
;
13
:
208
216
. https://doi.org/10.1016/j.hrthm.2015.08.031

901

Protonotarios
 
A
,
Anastasakis
 
A
,
Tsatsopoulou
 
A
,
Antoniades
 
L
,
Prappa
 
E
,
Syrris
 
P
, et al.  
Clinical significance of epsilon waves in arrhythmogenic cardiomyopathy
.
J Cardiovasc Electrophysiol
 
2015
;
26
:
1204
1210
. https://doi.org/10.1111/jce.12755

902

Gasperetti
 
A
,
Cappelletto
 
C
,
Carrick
 
R
,
Targetti
 
M
,
Tichnell
 
C
,
Martino
 
A
, et al.  
Association of premature ventricular contraction burden on serial Holter monitoring with arrhythmic risk in patients with arrhythmogenic right ventricular cardiomyopathy
.
JAMA Cardiol
 
2022
;
7
:
378
385
. https://doi.org/10.1001/jamacardio.2021.6016

903

Corrado
 
D
,
Basso
 
C
,
Leoni
 
L
,
Tokajuk
 
B
,
Bauce
 
B
,
Frigo
 
G
, et al.  
Three-dimensional electroanatomic voltage mapping increases accuracy of diagnosing arrhythmogenic right ventricular cardiomyopathy/dysplasia
.
Circulation
 
2005
;
111
:
3042
3050
. https://doi.org/10.1161/CIRCULATIONAHA.104.486977

904

Hoogendoorn
 
JC
,
Sramko
 
M
,
Venlet
 
J
,
Siontis
 
KC
,
Kumar
 
S
,
Singh
 
R
, et al.  
Electroanatomical voltage mapping to distinguish right-sided cardiac sarcoidosis from arrhythmogenic right ventricular cardiomyopathy
.
JACC Clin Electrophysiol
 
2020
;
6
:
696
707
. https://doi.org/10.1016/j.jacep.2020.02.008

905

Avella
 
A
,
d’Amati
 
G
,
Pappalardo
 
A
,
Re
 
F
,
Silenzi
 
PF
,
Laurenzi
 
F
, et al.  
Diagnostic value of endomyocardial biopsy guided by electroanatomic voltage mapping in arrhythmogenic right ventricular cardiomyopathy/dysplasia
.
J Cardiovasc Electrophysiol
 
2008
;
19
:
1127
1134
. https://doi.org/10.1111/j.1540-8167.2008.01228.x

906

Casella
 
M
,
Dello Russo
 
A
,
Bergonti
 
M
,
Catto
 
V
,
Conte
 
E
,
Sommariva
 
E
, et al.  
Diagnostic yield of electroanatomic voltage mapping in guiding endomyocardial biopsies
.
Circulation
 
2020
;
142
:
1249
1260
. https://doi.org/10.1161/CIRCULATIONAHA.120.046900

907

Tessier
 
R
,
Marteau
 
L
,
Vivien
 
M
,
Guyomarch
 
B
,
Thollet
 
A
,
Fellah
 
I
, et al.  
18F-Fluorodeoxyglucose positron emission tomography for the detection of myocardial inflammation in arrhythmogenic left ventricular cardiomyopathy
.
Circ Cardiovasc Imaging
 
2022
;
15
:
e014065
. https://doi.org/10.1161/CIRCIMAGING.122.014065

908

Gasperetti
 
A
,
Rossi
 
VA
,
Chiodini
 
A
,
Casella
 
M
,
Costa
 
S
,
Akdis
 
D
, et al.  
Differentiating hereditary arrhythmogenic right ventricular cardiomyopathy from cardiac sarcoidosis fulfilling 2010 ARVC Task Force Criteria
.
Heart Rhythm
 
2021
;
18
:
231
238
. https://doi.org/10.1016/j.hrthm.2020.09.015

909

Laredo
 
M
,
Duthoit
 
G
,
Gandjbakhch
 
E
,
Redheuil
 
A
,
Hebert
 
J-L
.
Total pericardium agenesis mistaken for arrhythmogenic right ventricular cardiomyopathy
.
Eur Heart J Cardiovasc Imaging
 
2018
;
19
:
120
. https://doi.org/10.1093/ehjci/jex251

910

Castelletti
 
S
,
Crotti
 
L
,
Dagradi
 
F
,
Rella
 
V
,
Salerno
 
S
,
Parati
 
G
, et al.  
Partial pericardial agenesis mimicking arrhythmogenic right ventricular cardiomyopathy
.
Clin J Sport Med
 
2020
;
30
:
e159
e162
. https://doi.org/10.1097/JSM.0000000000000733

911

Brosnan
 
MJ
,
Te Riele
 
A
,
Bosman
 
LP
,
Hoorntje
 
ET
,
van den Berg
 
MP
,
Hauer
 
RNW
, et al.  
Electrocardiographic features differentiating arrhythmogenic right ventricular cardiomyopathy from an athlete’s heart
.
JACC Clin Electrophysiol
 
2018
;
4
:
1613
1625
. https://doi.org/10.1016/j.jacep.2018.09.008

912

D’Ascenzi
 
F
,
Solari
 
M
,
Corrado
 
D
,
Zorzi
 
A
,
Mondillo
 
S
.
Diagnostic differentiation between arrhythmogenic cardiomyopathy and athlete’s heart by using imaging
.
JACC Cardiovasc Imaging
 
2018
;
11
:
1327
1339
. https://doi.org/10.1016/j.jcmg.2018.04.031

913

Rossi
 
VA
,
Niederseer
 
D
,
Sokolska
 
JM
,
Kovacs
 
B
,
Costa
 
S
,
Gasperetti
 
A
, et al.  
A novel diagnostic score integrating atrial dimensions to differentiate between the athlete’s heart and arrhythmogenic right ventricular cardiomyopathy
.
J Clin Med
 
2021
;
10
:
4094
. https://doi.org/10.3390/jcm10184094

914

van Tintelen
 
JP
,
Van Gelder
 
IC
,
Asimaki
 
A
,
Suurmeijer
 
AJH
,
Wiesfeld
 
ACP
,
Jongbloed
 
JDH
, et al.  
Severe cardiac phenotype with right ventricular predominance in a large cohort of patients with a single missense mutation in the DES gene
.
Heart Rhythm
 
2009
;
6
:
1574
1583
. https://doi.org/10.1016/j.hrthm.2009.07.041

915

Merner
 
ND
,
Hodgkinson
 
KA
,
Haywood
 
AFM
,
Connors
 
S
,
French
 
VM
,
Drenckhahn
 
JD
, et al.  
Arrhythmogenic right ventricular cardiomyopathy type 5 is a fully penetrant, lethal arrhythmic disorder caused by a missense mutation in the TMEM43 gene
.
Am J Hum Genet
 
2008
;
82
:
809
821
. https://doi.org/10.1016/j.ajhg.2008.01.010

916

Costa
 
S
,
Medeiros-Domingo
 
A
,
Gasperetti
 
A
,
Akdis
 
D
,
Berger
 
W
,
James
 
CA
, et al.  
Impact of genetic variant reassessment on the diagnosis of arrhythmogenic right ventricular cardiomyopathy based on the 2010 Task Force Criteria
.
Circ Genom Precis Med
 
2021
;
14
:
e003047
. https://doi.org/10.1161/CIRCGEN.120.003047

917

Elliott
 
PM
,
Anastasakis
 
A
,
Asimaki
 
A
,
Basso
 
C
,
Bauce
 
B
,
Brooke
 
MA
, et al.  
Definition and treatment of arrhythmogenic cardiomyopathy: an updated expert panel report
.
Eur J Heart Fail
 
2019
;
21
:
955
964
. https://doi.org/10.1002/ejhf.1534

918

Gasperetti
 
A
,
Targetti
 
M
,
Olivotto
 
I
.
Anti-arrhythmic drugs in arrhythmogenic right ventricular cardiomyopathy: the importance of optimal beta-blocker dose titration
.
Int J Cardiol
 
2021
;
338
:
150
151
. https://doi.org/10.1016/j.ijcard.2021.06.009

919

Corrado
 
D
,
Wichter
 
T
,
Link
 
MS
,
Hauer
 
R
,
Marchlinski
 
F
,
Anastasakis
 
A
, et al.  
Treatment of arrhythmogenic right ventricular cardiomyopathy/dysplasia: an international task force consensus statement
.
Eur Heart J
 
2015
;
36
:
3227
3237
. https://doi.org/10.1093/eurheartj/ehv162

920

Cappelletto
 
C
,
Gregorio
 
C
,
Barbati
 
G
,
Romani
 
S
,
De Luca
 
A
,
Merlo
 
M
, et al.  
Antiarrhythmic therapy and risk of cumulative ventricular arrhythmias in arrhythmogenic right ventricle cardiomyopathy
.
Int J Cardiol
 
2021
;
334
:
58
64
. https://doi.org/10.1016/j.ijcard.2021.04.069

921

Marcus
 
GM
,
Glidden
 
DV
,
Polonsky
 
B
,
Zareba
 
W
,
Smith
 
LM
,
Cannom
 
DS
, et al.  
Efficacy of antiarrhythmic drugs in arrhythmogenic right ventricular cardiomyopathy: a report from the North American ARVC Registry
.
J Am Coll Cardiol
 
2009
;
54
:
609
615
. https://doi.org/10.1016/j.jacc.2009.04.052

922

Wichter
 
T
,
Borggrefe
 
M
,
Haverkamp
 
W
,
Chen
 
X
,
Breithardt
 
G
.
Efficacy of antiarrhythmic drugs in patients with arrhythmogenic right ventricular disease. Results in patients with inducible and noninducible ventricular tachycardia
.
Circulation
 
1992
;
86
:
29
37
. https://doi.org/10.1161/01.CIR.86.1.29

923

Ermakov
 
S
,
Gerstenfeld
 
EP
,
Svetlichnaya
 
Y
,
Scheinman
 
MM
.
Use of flecainide in combination antiarrhythmic therapy in patients with arrhythmogenic right ventricular cardiomyopathy
.
Heart Rhythm
 
2017
;
14
:
564
569
. https://doi.org/10.1016/j.hrthm.2016.12.010

924

Rolland
 
T
,
Badenco
 
N
,
Maupain
 
C
,
Duthoit
 
G
,
Waintraub
 
X
,
Laredo
 
M
, et al.  
Safety and efficacy of flecainide associated with beta-blockers in arrhythmogenic right ventricular cardiomyopathy
.
Europace
 
2022
;
24
:
278
284
. https://doi.org/10.1093/europace/euab182

925

Daimee
 
UA
,
Assis
 
FR
,
Murray
 
B
,
Tichnell
 
C
,
James
 
CA
,
Calkins
 
H
, et al.  
Clinical outcomes of catheter ablation of ventricular tachycardia in patients with arrhythmogenic right ventricular cardiomyopathy: insights from the Johns Hopkins ARVC Program
.
Heart Rhythm
 
2021
;
18
:
1369
1376
. https://doi.org/10.1016/j.hrthm.2021.04.028

926

Mathew
 
S
,
Saguner
 
AM
,
Schenker
 
N
,
Kaiser
 
L
,
Zhang
 
P
,
Yashuiro
 
Y
, et al.  
Catheter ablation of ventricular tachycardia in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia: a sequential approach
.
J Am Heart Assoc
 
2019
;
8
:
e010365
. https://doi.org/10.1161/JAHA.118.010365

927

Gandjbakhch
 
E
,
Laredo
 
M
,
Berruezo
 
A
,
Gourraud
 
JB
,
Sellal
 
JM
,
Martins
 
R
, et al.  
Outcomes after catheter ablation of ventricular tachycardia without implantable cardioverter-defibrillator in selected patients with arrhythmogenic right ventricular cardiomyopathy
.
Europace
 
2021
;
23
:
1428
1436
. https://doi.org/10.1093/europace/euab172

928

Assis
 
FR
,
Krishnan
 
A
,
Zhou
 
X
,
James
 
CA
,
Murray
 
B
,
Tichnell
 
C
, et al.  
Cardiac sympathectomy for refractory ventricular tachycardia in arrhythmogenic right ventricular cardiomyopathy
.
Heart Rhythm
 
2019
;
16
:
1003
1010
. https://doi.org/10.1016/j.hrthm.2019.01.019

929

Shen
 
L-S
,
Liu
 
L-M
,
Zheng
 
L-H
,
Hu
 
F
,
Hu
 
Z-C
,
Liu
 
S-Y
, et al.  
Ablation strategies for arrhythmogenic right ventricular cardiomyopathy: a systematic review and meta-analysis
.
J Geriatr Cardiol
 
2020
;
17
:
694
703
. https://doi.org/10.11909/j.issn.1671-5411.2020.11.001

930

Romero
 
J
,
Patel
 
K
,
Briceno
 
D
,
Alviz
 
I
,
Gabr
 
M
,
Diaz
 
JC
, et al.  
Endo-epicardial ablation vs endocardial ablation for the management of ventricular tachycardia in arrhythmogenic right ventricular cardiomyopathy: a systematic review and meta-analysis
.
J Cardiovasc Electrophysiol
 
2020
;
31
:
2022
2031
. https://doi.org/10.1111/jce.14593

931

Christiansen
 
MK
,
Haugaa
 
KH
,
Svensson
 
A
,
Gilljam
 
T
,
Madsen
 
T
,
Hansen
 
J
, et al.  
Incidence, predictors, and success of ventricular tachycardia catheter ablation in arrhythmogenic right ventricular cardiomyopathy (from the Nordic ARVC Registry)
.
Am J Cardiol
 
2020
;
125
:
803
811
. https://doi.org/10.1016/j.amjcard.2019.11.026

932

Briceno
 
DF
,
Liang
 
JJ
,
Shirai
 
Y
,
Markman
 
TM
,
Chahal
 
A
,
Tschabrunn
 
C
, et al.  
Characterization of structural changes in arrhythmogenic right ventricular cardiomyopathy with recurrent ventricular tachycardia after ablation: insights from repeat electroanatomic voltage mapping
.
Circ Arrhythm Electrophysiol
 
2020
;
13
:
e007611
. https://doi.org/10.1161/CIRCEP.119.007611

933

Laredo
 
M
,
Da Silva
 
L
,
Extramiana
 
F
,
Lellouche
 
N
,
Varlet
 
E
,
Amet
 
D
, et al.  
Catheter ablation of electrical storm in patients with arrhythmogenic right ventricular cardiomyopathy
.
Heart Rhythm
 
2020
;
17
:
41
48
. https://doi.org/10.1016/j.hrthm.2019.06.022

934

Berruezo
 
A
,
Acosta
 
J
,
Fernandez-Armenta
 
J
,
Pedrote
 
A
,
Barrera
 
A
,
Arana-Rueda
 
E
, et al.  
Safety, long-term outcomes and predictors of recurrence after first-line combined endoepicardial ventricular tachycardia substrate ablation in arrhythmogenic cardiomyopathy. Impact of arrhythmic substrate distribution pattern. A prospective multicentre study
.
Europace
 
2017
;
19
:
607
616
. https://doi.org/10.1093/europace/euw212

935

Finocchiaro
 
G
,
Papadakis
 
M
,