Article

Arrhythmogenic Cardiomyopathy: Electrical and Structural Phenotypes

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

This overview gives an update on the molecular mechanisms, clinical manifestations, diagnosis and therapy of arrhythmogenic cardiomyopathy (ACM). ACM is mostly hereditary and associated with mutations in genes encoding proteins of the intercalated disc. Three subtypes have been proposed: the classical right-dominant subtype generally referred to as ARVC/D, biventricular forms with early biventricular involvement and left-dominant subtypes with predominant LV involvement. Typical symptoms include palpitations, arrhythmic (pre)syncope and sudden cardiac arrest due to ventricular arrhythmias, which typically occur in athletes. At later stages, heart failure may occur. Diagnosis is established with the 2010 Task Force Criteria (TFC). Modern imaging tools are crucial for ACM diagnosis, including both echocardiography and cardiac magnetic resonance imaging for detecting functional and structural alternations. Of note, structural findings often become visible after electrical alterations, such as premature ventricular beats, ventricular fibrillation (VF) and ventricular tachycardia (VT). 12-lead ECG is important to assess for depolarisation and repolarisation abnormalities, including T-wave inversions as the most common ECG abnormality. Family history and the detection of causative mutations, mostly affecting the desmosome, have been incorporated in the TFC, and stress the importance of cascade family screening. Differential diagnoses include idiopathic right ventricular outflow tract (RVOT) VT, sarcoidosis, congenital heart disease, myocarditis, dilated cardiomyopathy, athlete´s heart, Brugada syndrome and RV infarction. Therapeutic strategies include restriction from endurance and competitive sports, b-blockers, antiarrhythmic drugs, heart failure medication, implantable cardioverter-defibrillators and endocardial/epicardial catheter ablation.

Disclosure:The authors have no conflicts of interest to declare.

Received:

Accepted:

Acknowledgements:This work and the Zurich ARVC Program are supported by grants from the Georg and Bertha Schwyzer-Winiker Foundation, Zurich, the Baugarten Foundation, Zurich, filling the gap grant, University of Zurich and the Swiss National Science Foundation, Switzerland.

Correspondence Details:Dr Ardan M Saguner, Department of Cardiology, University Heart Center Zurich Rämistrasse 100, CH-8091 Zurich, Switzerland. E: ardan.saguner@usz.ch

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Arrhythmogenic cardiomyopathy (ACM) is usually referred to as arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D).1A first historical description was made in 1736, whereas its first modern description dates back to 1982.2 Initially, ACM was thought to be an embryological malformation.3 Yet in recent years it became evident that the pathophysiology of an ongoing genetically determined myocardial atrophy did not fit the theory of a congenital myocardial aplasia. Genetic and pathological studies have been crucial to understand ACM. From autoptic studies we know that atrophy of the ventricular myocardium due to progressive myocyte loss and infiltration by fibrofatty tissue are key findings.4 These studies led to the assignment of ARVC/D as a primary cardiomyopathy by the World Health Organization in 1995.5

In its most typical form, the right ventricle (RV) is primarily affected, which is then referred to as ARVC/D.6 As the disease progresses, the left ventricle (LV) may also be involved.7 Molecular studies have identified causative mutations in genes encoding proteins of the intercalated disc, desmosomes in particular.8 These mutations impair the electrical and mechanical stability of the ventricular myocardium9,10 with subsequent inflammation, apoptosis, necrosis and fibrofatty infiltration, which usually begins after puberty.11,12 In a minority of patients non-desmosomal mutations in calcium regulating genes, growth factors and other structural genes have been associated with ACM. Nonetheless, genetic mutations cannot entirely account for phenotypic expression and disease progression, and genetic mutations cannot be identified in up to 50 % of the ACM population studied. Therefore, epigenetic and environmental factors such as exercise seem to play a pivotal role as disease modifiers.1,13,14 Increased workload for the myocardium during physical activity enhances this adverse remodelling, and the thinner RV is particularly prone.15 Due to the multiple facets of the disease, the term ARVC/D is somewhat misleading. Since biventricular involvement and LV involvement may be present,1,16 a broader term such as arrhythmogenic cardiomyopathy has been recently proposed by the Heart Rhythm Society/European Heart Rhythm Association (HRS/EHRA).1,16,17 Together with novel genetic evidence that further expanded the pathophysiology of this heterogeneous disease beyond desmosomal mutations, the term ACM is now commonly being used to describe a hereditary form of non-hypertrophic cardiomyopathy that primarily manifests with ventricular arrhythmias due to fibrofatty infiltration of the ventricular myocardium.1,18,19

Epidemiology

Phenotypic expression is more common in males (2–3:1).14 ACM usually manifests during adolescence, but can also emerge in the elderly.20 With a general prevalence of 1:2,000–1:5,000 ARVC/D is a rare disease, which is defined as a prevalence of ≤1:2,000 according to the European definition.21 However, in some endemic areas the prevalence may be higher.22 ACM is a leading cause of sudden cardiac death (SCD) due to ventricular tachyarrhythmias, particularly in young athletes ≤35 years of age.23,24 In one Italian study, ACM accounted for up to 22 % of SCD in young adults.6,25,26 Generally, ACM first manifests with ventricular arrhythmias. In classical ARVC/D, ventricular arrhythmias arise from the RV.27

Genetics and Disease Modulation

ARVC/D is usually inherited as an autosomal dominant trait. Recessive forms with additional phenotypic characteristics exist.28 Recessive mutations of desmosomal genes can cause severe forms of ARVC/D, such as Naxos disease and Carvajal syndrome, some with skin and hair involvement, since both alleles are involved.29,30 Moreover, it has been shown that in up to 18 % of patients with ARVC/D, compound or digenic heterozygosity is present, indicating that in some cases more than one pathogenic allele may be involved.31 Currently 13 different genetic loci have been reported to be associated with ARVC/D. The most common genes encode for desmosomal proteins.32,33 Recent studies have shown that non-desmosomal genes can also lead to an ARVC/D phenotype (see Table 1). However, in cardiomyopathies – similar to other genetic diseases – it is very important to use rigorous criteria when calling a genetic variant or mutation pathogenic, since many variants may only constitute innocent bystanders.

A recent transatlantic collaboration with ACM registries from the Netherlands and the US has suggested rigorous criteria on how to classify genetic variants/mutations in ACM, which are now widely accepted in this field.32 Another recent large-scale trial on long QT and Brugada syndromes from the US has revealed that even among laboratories experienced in genetic testing for cardiac arrhythmia disorders, there was low concordance in designating variants as pathogenic. In an unselected population, the putatively pathogenic genetic variants were not associated with phenotypic abnormalities, which raises questions about the implications of notifying patients of incidental genetic findings.1,34,35 Thus, it is important to consider whether the genetic finding is present in a symptomatic patient or family member with a rather high pre-test probability, or in an asymptomatic patient as an incidental finding, who as a matter of course has a low pre-test probability.35

The American College of Medical Genetics and Genomics has published recommendations on how to report incidental findings in clinical exome and genome sequencing.36 Genetically affected relatives demonstrate variable, and often milder phenotypes, which has been attributed to incomplete disease penetrance. Therefore, the prevalence of familial disease is probably underestimated.32 Nonetheless, an important reason why index patients usually present with more severe phenotypes than their family members is so-called ascertainment bias. The clinician should bear in mind that relatives due to ascertainment alone will likely have milder phenotypes since they were not the ones to initially present. As opposed to this, index patients (probands) by default are always the most affected, which is an important reason why some genetics literature would exclude probands in analyses to offset ascertainment bias. This sort of bias is inherent to genetic diseases and has to be considered when performing genetic testing of index patients and family cascade screening.37

Table 1: Genetic Mutations Associated With Arrhythmogenic Right Ventricular Cardiomyopathy-Dysplasia

Article image

Desmosomes provide cell-to-cell adhesion and consist of three major groups of proteins:38  a) the transmembrane proteins (cadherins) desmocollin-2 (DSC2) and desmoglein-2 (DSG2), b) desmoplakin (DSP) and c) the linker armadillo proteins plakoglobin (JUP) and plakophillin-2 (PKP2), which are mediators between the cadherins and DSP.9,39 In about 80 % of cases with confirmed pathogenic mutations, mutations in PKP2, DSP, and DSG2 are identified,8 with the most common mutations involving PKP2. Several mutations have been described in these desmosomal genes. Besides unambiguous pathogenic splice site and truncating mutations, it is crucial to also identify pathogenic missense mutations in patients with ACM, as they have been reported to have the same impact on disease outcome as other mutations.32,40 Yet, the variability of genotype–phenotype correlations and the heterogeneity of genetic mutations pose a great challenge for ACM diagnosis and risk stratification. Some mutations have been associated with specific clinical outcomes. For instance, truncating mutations in DSP or digenic/ compound heterozygosity of desmosomal genes are associated with more aggressive phenotypes and can be considered as risk factors of SCD and heart failure.32,41,42 Further studies with larger patient cohorts and rigorous genetic criteria are needed in order to improve our understanding of genotype–phenotype correlations in ACM.

In a minority of cases, mutations can be detected in non-desmosomal genes. Mutations in the ryanodine receptor, which releases calcium from the sarcoplasmic reticulum during muscle contraction,43 and in the key calcium regulating protein phospholamban (PLN) may also lead to ACM phenotypes, and although these patients may present at an older age, their long-term prognosis might be worse.32,44 Mutations in transforming growth factor β 3 (TGFβ3) have also been reported, resulting in its overexpression inducing myocardial fibrosis by stimulation of mesenchymal proliferation.

TGFβ3 modulates desmosomal expression as well, so mutations can alter desmosomal distribution and influence cell–cell stability.45 A mutation in the transmembrane protein43 (TMEM43) leads to a severe and highly lethal form of ARVC/D. This mutation was identified as a founder mutation in Newfoundland. Although this mutation is not located on the heterosomes, the phenotype is gender-specific with males having lower life expectancy than females, and carrying a higher risk for SCD.46 Although the pathophysiological role of TMEM43 remains uncertain, studies have proposed that it may interact within the adipogenic pathway and also lead to nuclear structural changes.47 The TMEM43 p.S358L (Newfoundland) mutation has been shown to be fully penetrant.48 In this subgroup, it is important to screen large pedigrees since this mutation can be highly lethal and often presents with SCD as a first symptom.49 Newer data shows that further proteins are associated with ACM. The intracellular filament protein desmin, α-T catenin (a cytoplasmic protein), lamin A/C (a nuclear protein) and titin (a large sarcomeric protein) have been associated with ACM and phenocopies.1,34,50

Various genetic mutations have been associated with ACM, but these cannot account for the entire spectrum of disease expression. Therefore, epigenetic and environmental factors may act as disease modulators. First evidence for this hypothesis arose from monozygotic twin studies, where differences were reported in symptom onset, disease severity and arrhythmic risk.51 A male predominance of disease expression has been generally described in ACM, with male gender constituting an independent risk factor for adverse outcome.52,53 Recent data of compound and digenic heterozygosity indicates that modifier genes may account for residual variation and disease severity.31,54 Most importantly, strenuous physical activity, particularly endurance sports, plays an important role for early disease manifestation, disease severity and progression.13,55,56

Understanding the relationship between genotype and phenotype is challenging. Although ACM is mostly inherited as an autosomal dominant trait, this is likely an oversimplification since disease expressivity and penetrance are generally low. Clinical presentation and disease course can substantially differ within the same affected family due to the complex genetic and epigenetic background. Yet, it should be noted that variable phenotypic expression is seen in almost all genetic diseases, and is not only encountered in ACM. Using genetic testing as a diagnostic tool can be challenging. This particularly holds true for next-generation sequencing (NGS) methods, by which an abundancy of genetic variants of unknown significance can be detected, which is particularly challenging when it comes to the interpretation of borderline or overlapping phenotypes.34,57 Therefore, as previously mentioned, rigorous criteria to label a mutation as pathogenic should be used. Data from exome sequencing projects and in silico predictive programs can be helpful in this regard, especially for missense mutations and variants in order to avoid 'genetic' overdiagnosis.1,32 Nevertheless, genetic testing may be very helpful to verify ACM in the index patient and in identifying affected relatives and subclinical/ concealed phases. It is hoped that NGS will facilitate the identification of new disease modifiers or causative mutations, particularly in patients in whom a desmosomal mutation is present, but disease penetrance is not complete in family members harbouring this particular desmosomal mutation, and the real genetic culprit is yet to be found.58,59

Pathogenesis

The desmosomal complex is crucial for cellular adhesion, tissue strength and stability. It is located at the cardiac intercalated disc. A defective desmosome can result in a loss of cardiomyocytes accompanied by fibro-fatty tissue replacement.9,60,61 Desmosomal proteins interact with other junctional molecules such as other cadherins, catenins, ion channels, e.g. the sodium channel Nav1.5, and gap junction molecules.62 A functional link between desmosomes, gap junctions and Nav1.5 has been described, and recently named connexome.63 Several mechanisms are being discussed in the pathogenesis of ACM. Reduced cell-to-cell adhesion by desmosomal dysfunction due to genetic mutations is thought to be a key mechanism.39,64 Mutations in desmosomes change the 3D structure, their length and the total amount of desmosomes.65,66 This can trigger intercalated disc remodelling, which does not only alter mechanical stability, but also electrical coupling between cells, and intra/intercellular signal transduction affecting apoptosis and lipid metabolism.39,61,67,68

A decrease of JUP at the intercalated disc with its nuclear translocation and suppression of the canonical Wnt signalling pathway has been reported, even in individuals with other desmosomal mutations.65 Early histology and electron microscopy studies revealed that fibrofatty replacement of the RV goes along with inflammation and apoptosis.4,69,70 Induced pluripotent stem cell models from patients harbouring a pathogenic PKP2 mutation have highlighted that enhanced mitochondrial fatty acid uptake may lead to changes in fatty acid oxidation and promote fatty infiltration.61 Further experimental and clinical data suggest that non-desmosomal structures such as Nav1.5, PLN, desmin, titin and TMEM43 also interact with desmosomes and that mutations in these genes may change intracellular signalling pathways (see Figure 1).71,72PLN knockout has been associated with calcium channel modulation leading to VT.73 Furthermore, PLN phosphorylation has been reported to be upregulated in patients with ARVC/D72 in analogy to the pathophysiology of heart failure, which can be the endstage clinical presentation of ACM.74 Regarding titin mutations it has been shown that certain mutations may lead to a reduced structural stability and increased susceptibility for undergoing proteolysis.75

TMEM43 has also been associated with the intercalated disc and the adipogenic pathway.47,48 It has been shown that the TMEM43 p.S358L (Newfoundland) mutation may affect the localisation of proteins involved in cellular conduction and reduce conduction velocity in cardiac tissue.76 Yet, the role of non-desmosomal genetic mutations in the disease pathogenesis has not been fully elucidated, and there is a need for further experimental and clinical studies. Patient specific induced pluripotent stem cell models may play an important role in the discovery of novel disease mechanisms.61 Furthermore, volume overload and mechanical stress have also been suggested to enhance desmosomal dysfunction, which may explain why vigorous exercise has a negative impact on ACM phenotype, and why endurance athletes are particularly prone to adverse remodelling.77 In mice with PKP2 mutations, exercise leads to RV dilation and dysfunction.78,79 Clinical studies underscore the negative impact of sports, particularly high-intensity endurance training on ACM phenotype. 13,80 Myocarditis has also been associated with ACM, especially in genotype elusive patients, since cardiotropic viruses and bacteria have been found in cardiac tissue of these patients.11,81 Yet, the role of cardiotropic agents in the pathogenesis of ACM needs further investigation.

Phenotypic Expression

Classification of ACM into three different subtypes has been suggested. Right-dominant ARVC/D is considered as the classical form. Nonclassical forms were recently described.38,82,83 LV involvement is reported with a prevalence of up to 70 % of cases,84 which may be attributed to improved diagnostic methods.85 Yet, the proposed classification below is simplistic. It is important to keep in mind that due to genetic heterogeneity, epigenetic and environmental modifying factors and ascertainment bias there is a phenotypic continuum with the right- and left-dominant subtypes at opposite ends. Some mutations have been described to confer all described phenotypes.46 With regard to the important issue of ascertainment bias, individuals seeking medical attention often present with symptoms and advanced phenotypes. This can cause a selection bias in registries, and studies investigating ACM and genotype–phenotype correlations derived from these studies may not apply to a general ACM population. Such a bias is difficult to avoid in rare diseases such as ACM but has to be considered when interpreting study results.37

In right-dominant ARVC/D, a dilated RV with regional wall motion abnormalities with no or minimal LV involvement is observed (see Figure 2A). Myocardial remodelling starts in the subepicardial layers and may become transmural later on.17 Myocardial wall thinning can be seen on macroscopic examination.22,86 The subtricuspid region and RVOT are particularly prone to this remodelling process, leading to aneurysm formation.17 The concept of RV apical involvement and the term 'triangle of dysplasia' have recently been questioned.87 If an affected region is accessible for histological evaluation, inflammation, fibrosis and/or fatty infiltration can be visible.18,88

Biventricular arrhythmogenic cardiomyopathy is characterised by early involvement of both ventricles (see Figure 2B).89,90 Disease progression is characterised by systolic impairment and biventricular dilation with clinical features of global congestive heart failure. In contrast to dilated cardiomyopathy (DCM) with biventricular involvement, ventricular arrhythmias of both, right bundle branch block (RBBB) configuration – originating in the LV – and left bundle branch block (LBBB) configuration are present at an early stage.

Left-dominant arrhythmogenic cardiomyopathy (ALVC) has been proposed as a distinct form of ACM. It is characterised by the early occurrence of LV involvement (arrhythmias precede gross structural alterations), when global RV function is preserved. Electrocardiographic (ECG) and structural findings are left-sided analogues to those observed in ARVC/D (see Table 2).89

Clinical Presentation

ACM should be suspected if the following symptoms occur, particularly in young athletes:

  • Palpitations
  • Arrhythmic (pre)syncope
  • Aborted SCD
  • Chest pain ± rise in cardiac biomarkers
  • Presumed DCM with early onset and frequent ventricular arrhythmias
  • Precordial T-wave inversions beyond V1 after puberty (see Table 2, Figure 3).

Although palpitations and (pre)syncope are the most frequent symptoms,91 they also occur in many other arrhythmic syndromes such as cardiac sarcoidosis, channelopathies and other cardiomyopathies.92 Since there is a considerable overlap between ARVC/D and 'arrhythmic forms' of idiopathic DCM, the broader term ACM has been introduced.18 It is important to distinguish syndromes leading to ventricular arrhythmias and SCD from benign forms and those forms primarily leading to heart failure.26,42,93 A high clinical suspicion should be raised if symptoms correlate with premature ventricular beats or VT, particularly LBBB morphology with a superior axis. However, left ventricular forms or biventricular disease can also present with VT with RBBB morphology. Monomorphic VT is associated with more advanced disease stages, although gross structural abnormalities are not always required for maintaining re-entry circuits.93,94 Up to onequarter of patients present with atrial arrhythmias, most frequently atrial fibrillation.95,96 This is associated with inappropriate implantable cardioverter defibrillator (ICD) shocks and an increased risk of both death and heart failure.97 It is not exceptional for ACM to manifest with SCD (annual incidence up to 9 %),98 both during strenuous physical activity99 and in the sedentary state.15 In ARVC/D caused by TMEM43 mutations, enhanced sympathetic activity has been shown as a trigger for lethal arrhythmias, particularly in males.47

Figure 1: Phospolamban Protein Expression and Localisation

Article image

T-wave inversions in V1–3 are benign until puberty, and their prevalence among athletes and controls seems to be similar thereafter.100 If T-wave inversions in V1–3 are detected after puberty, transthoracic echocardiography (TTE) can be performed to rule out structural heart disease. Dyspnoea and signs of right-sided heart failure are rare. Congestive heart failure may occur with progressive LV involvement. The treating physician should keep in mind that ACM cannot be excluded by the absence of structural abnormalities, as arrhythmias often occur in the early – so called 'concealed phase' – preceding structural abnormalities. In a study evaluating 37 ARVC/D families, <50 % of family members had overt disease and 17 family members did not display the phenotype despite harbouring the pathogenic mutation.101 Of note, during the concealed phase and in subtypes primarily presenting with heart failure, palpitations and (pre)syncope do not have a great diagnostic value. Therefore, it is important to combine several tools for diagnosis and risk stratification, including genetics whenever possible and reasonable. In certain subgroups a positive mutation status, particularly in males, may play a role in diagnosis and risk stratification,35,58,102 for example in families harbouring the TMEM43 p.S358L (Newfoundland) mutation – a fully penetrant, sex-influenced, highly lethal form of ARVC/D.48

Table 2: Clinical Characteristics of Arrhythmogenic Ventricular Cardiomyopathy

Article image

Diagnosis

Revised 2010 Task Force Criteria

In 2010, the original 1994 TFC by McKenna et al.103 were revised to increase diagnostic sensitivity, particularly in affected asymptomatic family members.102 Pathogenic mutations were included, and cut-off values for imaging and histology were provided. The impact of these changes has been recently evaluated. Some investigators reported an increased diagnostic yield,104 whereas others did not.105,106 The clinician should bear in mind that the 2010 TFC only apply to ARVC/D, but not left-dominant ACM. For most forms of ACM there is only weak evidence supporting pre-symptomatic diagnosis by genetic testing.1,32 However, in the case of the TMEM43 p.S358L mutation, presymptomatic diagnosis can be made by a positive genotype only.48,49 For most subforms of ACM we have to rely on the 2010 TFC to establish the diagnosis. The current gold standard for diagnosis of ARVC/D are the 2010 TFC, at least until these criteria can be be further improved.102 The revised TFC consist of six diagnostic categories (see Table 3):

  • Global and/or regional myocardial dysfunction and structural abnormalities
  • Histological characterisation
  • Repolarisation abnormalities on 12-lead surface ECG
  • Depolarisation abnormalities on 12-lead surface ECG
  • Arrhythmias
  • Family history and genetics.

A definite diagnosis can be made with two major criteria, one major and two minor criteria, or four minor criteria from different categories; 'borderline' diagnosis with one major and one minor criterion, or three minor criteria and a 'possible' diagnosis if one major criterion or two minor criteria are present. Comprehensive non-invasive evaluation is mandatory, which includes a thorough clinical history, pedigree analysis, 12-lead surface ECG, TTE with detailed assessment of the RV, CMR, stress testing and Holter ECG. Event recorders and invasive diagnostics may be necessary if suspicion remains high and symptoms are rare. The revised TFC define quantitative criteria and abnormalities mainly based on comparison between adult index patients with ARVC/D and healthy controls. This approach has some limitations that should be taken into account. First, reference values of patients with ARVC/D and healthy subjects were derived from selected populations from tertiary care centres specialised in applying this particular diagnostic modality. Therefore, these reference values are prone to a substantial selection and ascertainment bias, and thus may not apply to the general ACM population, family members and individuals younger than 12 years. Moreover, they do not apply to patients with left-dominant forms.102

12-lead Surface ECG and Signal-averaged ECG

12-lead surface ECG will be abnormal in about 50 % of patients. This will include T-wave inversions in the right precordial leads, sometimes also involving V4–6, slurred S-wave upstroke in V1–3 ≥55 ms (see Figure 3), and with more advanced stages ε waves.107 The interpretation of ε waves significantly varies among observers. Furthermore, ε waves occur at more advanced stages, when the vast majority of patients already fulfils other TFC for definite diagnosis. Thus, ε waves in the absence of other diagnostic criteria should be interpreted with caution.108 T-wave inversions can be found in healthy individuals, patients with anterior ischaemia or RV hypertrophy.108,109 A recent study highlighted the importance of serial ECG evaluations, since dynamic ECG changes may occur.110 Delay of ventricular depolarisation due to scar (zig-zag pathways) may be visible as QRS fragmentation,108 ε waves111 or late potentials recorded by signal-averaged ECG (SAECG).112 SAECG may not be sensitive enough to detect early forms of ACM.112 Over recent years, the role of SAECG for ARVC/D diagnosis has diminished due to major advances in imaging and genetics.

Stress Testing

Ventricular arrhythmias in patients with ACM are often triggered by sympathetic activation. Thus, treadmill testing can reveal VT/VF or increase premature ventricular contractions with different morphologies. In a recent study, exercise testing revealed typical ECG abnormalities in a considerable number of patients with latent ARVC/D.113 In another study, the evaluation of the arrhythmic potential during very high dose isoproterenol infusion was sensitive for diagnosis of early forms of ARVC/D.114

Transthoracic Echocardiography

TTE is readily available in most centres and rapidly informative. Thus, TTE is considered as the initial imaging tool for suspected ACM and for screening family members. It may show RV enlargement and regional contraction abnormalities, most commonly in the subtricuspid region and RVOT.115 RVOT dimensions are crucial for diagnosis according to the 2010 TFC.116 The LV can be affected in up to 70 % of patients with ARVC/D displaying hypokinesia and a reduced ejection fraction. Frequently, LV structural abnormalities are localised in the posterolateral region.87,117,118 Novel technologies such as strain imaging allow for better quantification of regional wall motion, thereby allowing earlier disease detection and LV involvement.84

Cardiac Magnetic Resonance Tomography

Cardiac magnetic resonance tomography (CMR) has emerged as the non-invasive gold standard for assessing the RV over the past 20 years.119 Assessment of right-sided volumes and ejection fraction is highly accurate. Late-gadolinium enhanced (LGE) CMR can reveal myocardial fibrosis.120 Yet, myocardial fibrosis and fat as potential diagnostic features were not integrated in the TFC, because of their limited specificity, high intra-and inter-observer variability and the need for highly specialised interpreters.121,122 This owes to the fact that the RV is very thin and epicardial fat cannot reliably be distinguished from intramyocardial fat. However, CMR plays an important role for ARVC/D diagnosis (see Figure 2A). Consensus documents for nonclassic forms are awaited. The advent of novel technologies such as CMR tagging may facilitate early diagnosis of ARVC.123–127

RV Angiography

RV angiography is considered very useful to diagnose ARVC/D128 and thus is equivalent to TTE and CMR in the 2010 TFC. It has a positive predictive value of ∼85 %, with a negative predictive value of 95 %.129 High quality images allow assessment of RV morphology and wall motion. Yet, clinicians want to apply non-invasive diagnostic strategies without ionising radiation, particularly in young patients. Serial followup angiographies for monitoring disease progression are not feasible. Of note, hypokinesia is not considered diagnostic in the 2010 TFC, since akinesia or dyskinesia are required.

Electrophysiological Study and Electroanatomical Voltage Mapping

The goal of an electrophysiological study (EPS) in patients with ACM is the induction of sustained ventricular arrhythmias for making the diagnosis, risk stratification and to guide ablation. Moreover, the susceptibility for arrhythmias ± sympathetic challenge with isoproterenol, ICD treatment algorithms and efficacy of antiarrhythmic drugs (AAD) can be assessed. Electroanatomical voltage mapping (EAM) is a technique using electrophysiological catheters to measure myocardial voltages. After obtaining several hundred to thousands of points, a voltage map can be reconstructed. Normal myocardium generally displays bipolar voltages >1.5 mV. 130,131 In diseased myocardium, lower voltages with a longer duration, splitting and fractionation of signals can be recorded. Myocardial voltage maps can be obtained both from the endocardium and epicardium. EAM is generally safe, and improves outcomes of VT ablation.132–135 The diagnostic and prognostic utility of EAM has not been implemented in the current TFC, but recent data indicate that it can be useful for diagnosis and risk stratification.136

Figure 2: Structural Phenotypes of Arrhythmogenic Cardiomyopathy

Article image

Endomyocardial Biopsy

Endomyocardial biopsy (EMB) has, for a long time, been considered the diagnostic gold standard for ACM diagnosis. Indeed, histological examination and immunostaining may allow confirmation of ACM, and exclude differential diagnoses, e.g. sarcoidosis, Chagas disease or other forms of myocarditis. Yet, biopsies are usually taken from the RV septum for safety reasons. Since the process of fibro-fatty infiltration spares the septum, EMB often yields false-negative results.137,138 EMB from diseased regions is problematic, as these regions are very thin, and sampling carries an increased risk of perforation. As ACM is patchy, several biopsies should be obtained. EAM-guided biopsies taken from low-voltage areas may improve diagnostic yield and better distinguish between myocarditis or sarcoidosis.130,139

Genetic Testing

When performing genetic testing, rigorous criteria to ascertain whether a mutation or variant is pathogenic should be used. An HRS/EHRA consensus statement for genetic testing in ACM was published recently.1 Genetic testing is performed to confirm ACM in individuals with a high (Class IIa) or intermediate (at least one major or two minor criteria; Class IIb) clinical suspicion and to identify genetically-affected relatives harbouring the pathogenic mutation (Class I). Genetic testing in cases fulfilling only one minor criterion is not recommended.8,42,58 A negative genetic test does not exclude ACM, since other unknown causal mutations and environmental factors may also cause the disease.60,140 With the exception of TMEM43 p.S358L mutation carriers48 and autosomal-recessive forms, genotype does not provide a diagnosis of ACM by itself.1,8,102 Yet, the identification of pathogenic mutations may be useful in the differential diagnosis of ACM and phenocopies, such as myocarditis, idiopathic RVOT tachycardia, DCM, muscular dystrophies or sarcoidosis.1,32,141 Genetic testing should not be seen as the only diagnostic tool, but may be very helpful especially in identifying affected relatives and subclinical/ concealed phases.58,59 Genetic cascade screening of relatives offers an alternative strategy to serial clinical evaluation. In this regard, the absence of a clear pathogenic mutation in a family member causing the disease in the index patient obviates the need for serial clinical evaluation in this family member. Current guidelines102 do not recommend genetic testing for risk stratification and therapeutic decision-making in ACM due to conflicting study results.8,142–144

Table 3: Revised (2010) Task Force Criteria for Diagnosis of ARVC/D

Article image

Recent studies showed an association between positive genotype and earlier disease onset in index patients harbouring a known genetic mutation as compared with index patients without a known genetic mutation. However, ascertainment bias has to be considered in such a comparison.32 Moreover, these results can be biased by the fact that ACM is almost never fully penetrant, >40 % of ACM patients are genotype elusive, and the pathogenic mechanism in these patients is not clear. Yet, the results of genotype–phenotype correlation studies can be clinically helpful for some genetic mutations. For instance, truncating mutations in DSP, digenic or compound heterozygous mutations and mutations in the non-desmosomal gene TMEM43 are associated with more severe phenotypes and therefore, may be considered as risk factors of SCD.8,32,41,42,102 For TMEM43 p.S358L carriers, a diagnosis of ARVC/D can be made only by the presence of this genetic mutation, and clinical screening of family members is strongly recommended.49

Differential Diagnosis

A common differential diagnosis is idiopathic right ventricular outflow tract VT (RVOT-VT), particularly in early stages of ARVC/D lacking gross structural abnormalities. 145 RVOT-VT is not associated with structural heart disease, and thus has a more benign course. In RVOT-VT, 12-lead surface ECG and SAECG are normal during sinus rhythm. A single VT morphology with LBBB pattern and an inferior axis is recorded in most of the cases, although slightly different morphologies (all with an inferior axis) can occur if VT origin is above the pulmonary cusps.146 ECG scoring systems to differentiate idiopathic RVOT-VT and ARVC/D have been suggested.147 In ARVC/D, QRS duration during VT was longer (≥120 ms in lead I).148 Notching of the QRS and precordial transition in lead V6 were exclusively seen in ARVC/D. Idiopathic RVOT-VT is difficult to induce by programmed ventricular stimulation.148,149 Idiopathic RVOTVT responds well to verapamil. Endocardial ablation is feasible and curative, whereas in ARVC/D, ablation is not curative and an epicardial approach may be necessary to eliminate the clinical VT. Genetic testing, a positive family history, EAM and EMB can help to differentiate ACM and localised forms of myocarditis.150 Cardiac sarcoid can mimic ARVC/D, and the current TFC do not reliably distinguish between these two. In a study of patients with suspected ARVC/D also being evaluated by EMB, a high incidence (15 %) of cardiac sarcoid was identified.151 Cardiac sarcoid should be considered if respiratory and systemic symptoms, high-grade atrioventricular conduction block and septal involvement are present, and familial disease is absent. Diagnosis is confirmed by EMB and thoracic CT scans.151

DCM is particularly difficult to distinguish from non-classic forms of ACM, and these two entities can significantly overlap and harbour similar mutations. Palpitations, (pre)syncope and ventricular arrhythmias are present at an early stage in ACM, often in the absence of gross structural abnormalities, which is the opposite in DCM.18 Atrioventricular conduction block is more common in DCM, but mutations in lamin A/C can lead to ACM with conduction defects, as well.152 Brugada syndrome can mimic ACM, as RV conduction delay has been demonstrated in both entities. The presence of structural abnormalities favours ACM. Recently, a genetic overlap between these two has been suggested. In vitro studies have shown a cellular interaction between desmosomes and ion channels. Mutations associated with Brugada syndrome were found in ACM patients and vice versa.153 Titin mutations have also been associated with a phenotypic overlap between ACM and DCM. The same holds true for PLN mutations.44 However, the genetic interpretation of the large titin molecule and its variants is particularly challenging.75 Treatment of these patients and their arrhythmogenic risk has not been established yet, which has to be addressed in future studies. Other common differential diagnoses include right ventricular infarction, congenital left-to-right shunts, Chagas disease and Uhl’s disease.22 Finally, adaptation of the RV to increased workload in endurance athletes can mimic ARVC/D, and there is a debatable grey zone of what is considered physiological adaptation.154

Disease Course

ACM is a progressive disease however individual disease course can vary. Cardiac mortality is currently estimated ∼0.9 % per year. Most patients die of progressive heart failure or ventricular tachyarrhythmias.155 Four disease phases have been proposed:65

Figure 3: Electrical Phenotypes of Arrhythmogenic Cardiomyopathy

Article image
  1. Concealed phase: patients are asymptomatic and structural abnormalities are absent. SCD due to VF can be the primary manifestation in this phase.
  2. Occurrence of symptomatic arrhythmias
  3. Early heart failure symptoms
  4. End-stage heart failure.

Diagnosis and risk stratification during the concealed phase can be very challenging. Genetic testing with rigorous criteria and pre-participation screening of young athletes might help to identify affected individuals and prevent SCD.1,26,156 In one study, 7 % of ACM patients received cardiac transplantation after a mean follow-up of 10 years, mostly due to severe LV involvement.7 Physical activity promotes earlier disease manifestation and more rapid disease progression. Identification of affected athletes by pre-participation screening seems to substantially reduce mortality in this cohort.143,156

Therapy

Recently, an international consensus statement on the treatment of ARVC/D has been published.144 Data on non-classic ACM subtypes is scarce.

Physical Activity Restriction

Competitive athletes with ACM have up-to five-fold increased risk of SCD compared with sedentary individuals with ACM. Other studies have confirmed that high-level physical activity, particularly endurance sports, promotes disease onset, progression and adverse outcome.13,55 Thus, it is recommended that patients with a definite diagnosis of ARVC/D do not participate in competitive and/or endurance sports (Class I). Furthermore, they should be restricted from participation in athletic activities, with the possible exception of recreational low intensity sports (Class IIa).56,157

Pharmacological Therapy

Pharmacological options consist of antiarrhythmic drugs (AAD), e.g. sotalol, amiodarone and mexiletine, b-blockers and heart failure drugs. AADs are recommended as an adjunct to ICD therapy in patients with frequent discharges (class I).158 Moreover, they should be considered to improve symptoms due to frequent premature ventricular beats or non-sustained VT (class IIa), and may be considered as an adjunct to catheter ablation in selected patients with haemodynamically stable VT without an ICD (class IIb). Of note, cardioselective β-blockers should be considered in all patients with the ARVC/D phenotype irrespective of arrhythmias (class IIa), but their prophylactic use in healthy gene carriers is not recommended.158

Implantable Cardioverter Defibrillator

According to the most recent guidelines by the American College of Cardiology (ACC)/American Heart Association (AHA), the European Society of Cardiology (ESC) and the most recent international task force consensus statement on the treatment of ARVC/D, an ICD is recommended in patients with aborted SCD, haemodynamically unstable VT or VF (class I), and according to the consensus statement in those with severe systolic dysfunction of the RV, LV or both, irrespective of arrhythmias (class I).144,159–161 An ICD is generally indicated in ARVC/D patients who have experienced an episode of haemodynamically stable, sustained VT. If major risk factors such as unexplained syncope, moderate ventricular dysfunction, or non-sustained VTs are present, ICD implantation should be considered. Nonetheless, there is no clear consensus regarding primary prevention of SCD in ACM patients without documented sustained ventricular arrhythmia (VA).

The AHA recommends an ICD in patients with a familial cardiomyopathy associated with SCD (class IIb, level of evidence C).161 The ESC states that in patients without documented VA or syncope, an ICD may be considered after detailed clinical assessment including family history, severity of RV and LV function and other factors such as psychological health and socioeconomic status.160 According to the international consensus statement, ICD implantation may be considered in patients with minor risk factors such as gender, age and ECG abnormalities and these patients should be evaluated on an individual basis. A single-chamber device is generally preferred in order to minimise the incidence of lead-related complications in this relatively young cohort. Since many patients benefit from antitachycardia pacing, there is currently no role for subcutaneous ICDs. Importantly, prophylactic ICD implantation is not recommended in asymptomatic patients without risk factors and most healthy gene carriers (class III).144 However, since the TMEM43 p.S358L mutation is known to be fully penetrant and highly lethal, an ICD for primary prevention is indicated in all patients and family members harbouring this mutation (males immediately post-puberty and females ≥30 years) in order to improve survival in this subpopulation.49

Catheter Ablation

Catheter ablation of VT is recommended in patients with incessant VT or frequent appropriate ICD interventions for VT despite AAD (on amiodarone: class I, off amiodarone: class IIa).144 If ≥1 endocardial ablation procedure fails, an epicardial approach is recommended (class I).135 ARVC/D begins in the subepicardial layers. Thus, in experienced hands, a combined endocardial/epicardial approach can be followed as an initial ablation strategy (class IIa). Since catheter ablation is not curative, it is not recommended as an alternative to ICD for prevention of SCD.144

Conclusion

Discoveries within the last decade have substantially improved our understanding of ACM from a purely RV dysplasia to an inherited polygenic disease with a broad phenotypic spectrum. Although ACM predominantly affects the RV, atypical forms also affecting the LV are encountered. Emerging technologies in imaging, genetics and device therapy have facilitated diagnosis and prevention of SCD. Future challenges comprise early identification of asymptomatic patients and family members, improved risk stratification, and causal therapies to cure this challenging disease.

Clinical Perspective

  • Arrhythmogenic cardiomyopathy (ACM) is a hereditary cardiomyopathy characterised by ventricular arrhythmias and structural/functional abnormalities of the ventricles.
  • The most common arrhythmogenic right ventricular cardiomyopathy is generally referred to as ARVC/D.
  • Causative mutations are detected in genes encoding the intercalated disc.
  • Non-desmosomal mutations (e.g. TMEM43) have also been associated with ARVC/D and can cause malignant phenotypes.
  • Exercise can promote disease onset, progression and adverse outcome.
  • ACM is a common cause of sudden cardiac death in young athletes.
  • The current gold standard for diagnosis are the 2010 TFC.
  • Therapeutic strategies include restriction from endurance and competitive sports, β-blockers, antiarrhythmic drugs, heart failure medication, implantable cardioverterdefibrillators and endocardial/epicardial catheter ablation.

References

  1. Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies: this document was developed as a partnership between the Heart Rhythm Society (HRS) and the European Heart Rhythm Association (EHRA). Europace 2011;13:1077–9.
    Crossref | PubMed
  2. Marcus FI, Fontaine GH, Guiraudon G, et al. Right ventricular dysplasia: a report of 24 adult cases. Circulation 1982;65: 384–98.
    Crossref | PubMed
  3. Angelini A, Basso C, Nava A, Thiene G. Endomyocardial biopsy in arrhythmogenic right ventricular cardiomyopathy. Am Heart J 1996;132:203–6.
    Crossref | PubMed
  4. Basso C, Thiene G, Corrado D, et al. Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, or myocarditis? Circulation 1996;94:983–91.
    Crossref | PubMed
  5. Richardson P, McKenna W, Bristow M, et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of cardiomyopathies. Circulation 1996;93:841–2.
    Crossref | PubMed
  6. Basso C, Calabrese F, Corrado D, Thiene G. Postmortem diagnosis in sudden cardiac death victims: macroscopic, microscopic and molecular findings. Cardiovasc Res 2001;50:290–300.
    Crossref | PubMed
  7. Pinamonti B, Dragos AM, Pyxaras SA, et al. Prognostic predictors in arrhythmogenic right ventricular cardiomyopathy: results from a 10-year registry. Eur Heart J 2011;32:1105–13. 
    Crossref | PubMed
  8. Fressart V, Duthoit G, Donal E, et al. Desmosomal gene analysis in arrhythmogenic right ventricular dysplasia/ cardiomyopathy: spectrum of mutations and clinical impact in practice. Europace 2010;12:861–8. 
    Crossref | PubMed
  9. Delmar M, McKenna WJ. The cardiac desmosome and arrhythmogenic cardiomyopathies: from gene to disease. Circ Res 2010;107:700–14.
    Crossref | PubMed
  10. Sato PY, Coombs W, Lin X, et al. Interactions between ankyrin-G, Plakophilin-2, and Connexin43 at the cardiac intercalated disc. Circ Res 2011;109:193–201.
    Crossref | PubMed
  11. Calabrese F, Basso C, Carturan E, et al. Arrhythmogenic right ventricular cardiomyopathy/dysplasia: is there a role for viruses? Cardiovasc Pathol 2006;15:11–7. 
    Crossref | PubMed
  12. Thiene G, Corrado D, Nava A, et al. Right ventricular cardiomyopathy: is there evidence of an inflammatory aetiology? Eur Heart J 1991;12:22–5.
    Crossref | PubMed
  13. James CA, Bhonsale A, Tichnell C, et al. Exercise increases age-related penetrance and arrhythmic risk in arrhythmogenic right ventricular dysplasia/cardiomyopathyassociated desmosomal mutation carriers. J Am Coll Cardiol 2013;62:1290–7.
    Crossref | PubMed
  14. Meyer S, van der Meer P, van Tintelen JP, van den Berg MP. Sex differences in cardiomyopathies. Eur J Heart Fail 2014;16:238–47.
    Crossref | PubMed
  15. Chimenti C, Pieroni M, Maseri A, Frustaci A. Histologic findings in patients with clinical and instrumental diagnosis of sporadic arrhythmogenic right ventricular dysplasia. J Am Coll Cardiol 2004;43:2305–13. 
    Crossref | PubMed
  16. Gallo P, d’Amati G, Pelliccia F. Pathologic evidence of extensive left ventricular involvement in arrhythmogenic right ventricular cardiomyopathy. Hum Pathol 1992;23:948–52.
    Crossref | PubMed
  17. Corrado D, Basso C, Thiene G, et al. Spectrum of clinicopathologic manifestations of arrhythmogenic right ventricular cardiomyopathy/dysplasia: a multicenter study. J Am Coll Cardiol 1997;30:1512–20.
    Crossref | PubMed
  18. Sen-Chowdhry S, Morgan RD, Chambers JC, McKenna WJ. Arrhythmogenic cardiomyopathy: etiology, diagnosis, and treatment. Annu Rev Med 2010;61 :233–53.
    Crossref | PubMed
  19. Sen-Chowdhry S, Syrris P, Pantazis A, et al. Mutational heterogeneity, modifier genes, and environmental influences contribute to phenotypic diversity of arrhythmogenic cardiomyopathy. Circ Cardiovasc Genet 2010;3:323–30.
    Crossref | PubMed
  20. Koulouris S, Pastromas S, Sakellariou D, et al. Arrhythmogenic right ventricular cardiomyopathy in an octogenarian presenting with ventricular tachycardia. Pacing Clin Electrophysiol 2009;32:e43–7.
    Crossref | PubMed
  21. Richter T, Nestler-Parr S, Babela R, et al. Rare disease terminology and definitions – a systematic global review: report of the ISPOR rare disease special interest group. Value Health 2015;18:906–14. DOI: 10.1016/j.jval.2015.05.008; PMID: 26409619
    Crossref | PubMed
  22. Herren T, Gerber PA, Duru F. Arrhythmogenic right ventricular cardiomyopathy/dysplasia: a not so rare «disease of the desmosome» with multiple clinical presentations. Clin Res Cardiol 2009;98:141–58. 
    Crossref | PubMed
  23. Basso C, Corrado D, Thiene G. Cardiovascular causes of sudden death in young individuals including athletes. Cardiol Rev 1999;7:127–35. 
    Crossref | PubMed
  24. Tabib A, Loire R, Miras A, et al. Unsuspected cardiac lesions associated with sudden unexpected perioperative death. Eur J Anaesthesiol 2000;17:230–5.
    Crossref | PubMed
  25. Nava A, Canciani B, Daliento L, et al. Juvenile sudden death and effort ventricular tachycardias in a family with right ventricular cardiomyopathy. Int J Cardiol 1988;21 :111–26.
    Crossref | PubMed
  26. Corrado D, Drezner J, Basso C, et al. Strategies for the prevention of sudden cardiac death during sports. Eur J Cardiovasc Prev Rehabil 2011;18:197–208.
    Crossref | PubMed
  27. Marcus FI, Fontaine G. Arrhythmogenic right ventricular dysplasia/cardiomyopathy: a review. Pacing Clin Electrophysiol 1995;18:1298–314.
    Crossref | PubMed
  28. Corrado D, Basso C, Pilichou K, Thiene G. Molecular biology and clinical management of arrhythmogenic right ventricular cardiomyopathy/dysplasia. Heart 2011;97:530–9.
    Crossref | PubMed
  29. Saffitz JE, Asimaki A, Huang H. Arrhythmogenic right ventricular cardiomyopathy: new insights into mechanisms of disease. Cardiovasc Pathol 2010;19:166–70. 
    Crossref | PubMed
  30. Lorenzon A, Pilichou K, Rigato I, et al. Homozygous desmocollin-2 mutations and arrhythmogenic cardiomyopathy. Am J Cardiol 2015;116:1245–51. 
    Crossref | PubMed
  31. Xu T, Yang Z, Vatta M, et al. Compound and digenic heterozygosity contributes to arrhythmogenic right ventricular cardiomyopathy. J Am Coll Cardiol 2010;55:587–97. 
    Crossref | PubMed
  32. Bhonsale A, Groeneweg JA, James CA, et al. Impact of genotype on clinical course in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated mutation carriers. Eur Heart J 2015;36:847–55. 
    Crossref | PubMed
  33. Christensen AH, Benn M, Bundgaard H, et al. Wide spectrum of desmosomal mutations in Danish patients with arrhythmogenic right ventricular cardiomyopathy. J Med Genet 2010;47:736–44. 
    Crossref | PubMed
  34. Andreasen C, Nielsen JB, Refsgaard L, et al. New populationbased exome data are questioning the pathogenicity of previously cardiomyopathy-associated genetic variants. Eur J Hum Genet 2013;21 :918–28. 
    Crossref | PubMed
  35. Van Driest SL, Wells QS, Stallings S, et al. Association of arrhythmia-related genetic variants with phenotypes documented in electronic medical records. JAMA 2016;315: 47–57.
    Crossref | PubMed
  36. Green RC, Berg JS, Grody WW, et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet Med 2013;15:565–74.
    Crossref | PubMed
  37. Park S, Lee S, Lee Y, et al. Adjusting heterogeneous ascertainment bias for genetic association analysis with extended families. BMC Med Genet 2015;16:62.
    Crossref | PubMed
  38. Huber O. Structure and function of desmosomal proteins and their role in development and disease. Cell Mol Life Sci 2003;60:1872–90.
    Crossref | PubMed
  39. Basso C, Czarnowska E, Della Barbera M, et al. Ultrastructural evidence of intercalated disc remodelling in arrhythmogenic right ventricular cardiomyopathy: an electron microscopy investigation on endomyocardial biopsies. Eur Heart J 2006;27:1847–54.
    Crossref | PubMed
  40. Bauce B, Rampazzo A, Basso C, et al. Clinical phenotype and diagnosis of arrhythmogenic right ventricular cardiomyopathy in pediatric patients carrying desmosomal gene mutations. Heart Rhythm 2011;8:1686–95.
    Crossref | PubMed
  41. López-Ayala JM, Gómez-Milanés I, Sánchez Muñoz JJ, et al. Desmoplakin truncations and arrhythmogenic left ventricular cardiomyopathy: characterizing a phenotype. Europace 2014;16:1838–46.
    Crossref | PubMed
  42. Rigato I, Bauce B, Rampazzo A, et al. Compound and digenic heterozygosity predicts life-time arrhythmic outcome and sudden cardiac death in desmosomal generelated arrhythmogenic right ventricular cardiomyopathy. Circ Cardiovasc Genet 2013;6:533–42.
    Crossref | PubMed
  43. Tiso N, Stephan DA, Nava A, et al. Identification of mutations in the cardiac ryanodine receptor gene in families affected with arrhythmogenic right ventricular cardiomyopathy type 2 (ARVD2). Hum Mol Genet 2001;10:189–94.
    Crossref | PubMed
  44. van der Zwaag PA, van Rijsingen IA, Asimaki A, 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– 207.
    Crossref | PubMed
  45. Beffagna G, Occhi G, Nava A, et al. Regulatory mutations in transforming growth factor-beta3 gene cause arrhythmogenic right ventricular cardiomyopathy type 1. Cardiovasc Res 2005;65:366–73.
    Crossref | PubMed
  46. Hodgkinson K, Connors S, Merner N, et al. The natural history of a genetic subtype of arrhythmogenic right ventricular cardiomyopathy caused by a p.S358L mutation in TMEM43. Clin Genet 2012;83:321–31. 
    Crossref | PubMed
  47. Milting H, Klauke B, Christensen AH, et al. The TMEM43 Newfoundland mutation p.S358L causing ARVC-5 was imported from Europe and increases the stiffness of the cell nucleus. Eur Heart J 2015;36:872–81.
    Crossref | PubMed
  48. Merner ND, Hodgkinson KA, Haywood AF, 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;82:809–21.
    Crossref | PubMed
  49. Hodgkinson KA, Howes AJ, Boland P, 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.
    Crossref | PubMed
  50. Chauveau C, Rowell J, Ferreiro A. A rising titan: TTN review and mutation update. Hum Mutat 2014;35:1046–59.
    Crossref | PubMed
  51. Wlodarska EK, Konka M, Zaleska T, et al. Arrhythmogenic right ventricular cardiomyopathy in two pairs of monozygotic twins. Int J Cardiol 2005;105:126–33.
    Crossref | PubMed
  52. Bauce B, Frigo G, Marcus FI, et al. Comparison of clinical features of arrhythmogenic right ventricular cardiomyopathy in men versus women. Am J Cardiol 2008;102:1252–7. 
    Crossref | PubMed
  53. Link MS, Laidlaw D, Polonsky B, et al. Ventricular arrhythmias in the North American multidisciplinary study of ARVC: predictors, characteristics, and treatment. J Am Coll Cardiol 2014;64:119–25.
    Crossref | PubMed
  54. Rigato I, Bauce B, Rampazzo A, et al. Compound and digenic heterozygosity predicts lifetime arrhythmic outcome and sudden cardiac death in desmosomal generelated arrhythmogenic right ventricular cardiomyopathy. Circ Cardiovasc Genet 2013;6:533–42.
    Crossref | PubMed
  55. Sawant AC, Bhonsale A, te Riele AS, et al. Exercise has a disproportionate role in the pathogenesis of arrhythmogenic right ventricular dysplasia/cardiomyopathy in patients without desmosomal mutations. J Am Heart Assoc 2014;3:e001471.
    Crossref | PubMed
  56. Ruwald AC, Marcus F, Estes NA, et al. Association of competitive and recreational sport participation with cardiac events in patients with arrhythmogenic right ventricular cardiomyopathy: results from the North American multidisciplinary study of arrhythmogenic right ventricular cardiomyopathy. Eur Heart J 2015;36:1735–43.
    Crossref | PubMed
  57. Medeiros-Domingo A, Saguner AM, Magyar I, et al. Arrhythmogenic right ventricular cardiomyopathy: implications of next-generation sequencing in appropriate diagnosis. Europace 2016:euw098.
    Crossref | PubMed
  58. Kapplinger JD, Landstrom AP, Salisbury BA, et al. Distinguishing arrhythmogenic right ventricular cardiomyopathy/dysplasia-associated mutations from background genetic noise. J Am Coll Cardiol 2011;57:2317–27.
    Crossref | PubMed
  59. Marcus FI, Edson S, Towbin JA. Genetics of arrhythmogenic right ventricular cardiomyopathy: a practical guide for physicians. J Am Coll Cardiol 2013;61 :1945–8. 
    Crossref | PubMed
  60. Awad MM, Calkins H, Judge DP. Mechanisms of disease: molecular genetics of arrhythmogenic right ventricular dysplasia/cardiomyopathy. Nat Clin Pract Cardiovasc Med 2008;5:258–67.
    Crossref | PubMed
  61. Kim C, Wong J, Wen J, et al. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature 2013;494:105–10.
    Crossref | PubMed
  62. Delmar M. Desmosome-ion channel interactions and their possible role in arrhythmogenic cardiomyopathy. Pediatr Cardiol 2012;33:975–9. 
    Crossref | PubMed
  63. Agullo-Pascual E, Reid DA, Keegan S, et al. Super-resolution fluorescence microscopy of the cardiac connexome reveals plakophilin-2 inside the connexin43 plaque. Cardiovasc Res 2013;100:231–40.
    Crossref | PubMed
  64. Rizzo S, Lodder EM, Verkerk AO, et al. Intercalated disc abnormalities, reduced Na(+) current density, and conduction slowing in desmoglein-2 mutant mice prior to cardiomyopathic changes. Cardiovasc Res 2012;95:409–18.
    Crossref | PubMed
  65. Asimaki A, Tandri H, Huang H, et al. A new diagnostic test for arrhythmogenic right ventricular cardiomyopathy. N Engl J Med 2009;360:1075–84.
    Crossref | PubMed
  66. Lombardi R, da Graca Cabreira-Hansen M, Bell A, et al. Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy. Circ Res 2011;109:1342–53.
    Crossref | PubMed
  67. Garcia-Gras E, Lombardi R, Giocondo MJ, et al. Suppression of canonical Wnt/beta-catenin signaling by nuclear plakoglobin recapitulates phenotype of arrhythmogenic right ventricular cardiomyopathy. J Clin Invest 2006;116:2012–21.
    Crossref | PubMed
  68. Noorman M, Hakim S, Kessler E, et al. Remodeling of the cardiac sodium channel, connexin43, and plakoglobin at the intercalated disk in patients with arrhythmogenic cardiomyopathy. Heart Rhythm 2013;10:412–9.
    Crossref | PubMed
  69. Hariharan V, Asimaki A, Michaelson JE, et al. Arrhythmogenic right ventricular cardiomyopathy mutations alter shear response without changes in cell-cell adhesion. Cardiovasc Res 2014;104:280–9. 
    Crossref | PubMed
  70. Oxford EM, Everitt M, Coombs W, et al. Molecular composition of the intercalated disc in a spontaneous canine animal model of arrhythmogenic right ventricular dysplasia/ cardiomyopathy. Heart Rhythm 2007;4:1196–205.
    Crossref | PubMed
  71. Agullo-Pascual E, Lin X, Leo-Macias A, et al. Super-resolution imaging reveals that loss of the C-terminus of connexin43 limits microtubule plus-end capture and NaV1.5 localization at the intercalated disc. Cardiovasc Res 2014;104:371–81.
    Crossref | PubMed
  72. Akdis D, Medeiros-Domingo A, Gaertner-Rommel A, et al. Myocardial expression profiles of candidate molecules in arrhythmogenic right ventricular cardiomyopathy/ dysplasia compared with dilated cardiomyopathy and healthy controls. Heart Rhythm 2015;13:731–41.
    Crossref | PubMed
  73. Bai Y, Jones PP, Guo J, et al. Phospholamban knockout breaks arrhythmogenic Ca²+ waves and suppresses catecholaminergic polymorphic ventricular tachycardia in mice. Circ Res 2013;113:517–26. 
    Crossref | PubMed
  74. Haghighi K, Bidwell P, Kranias EG. Phospholamban interactome in cardiac contractility and survival: A new vision of an old friend. J Mol Cell Cardiol 2014;77:160–7. 
    Crossref | PubMed
  75. Taylor M, Graw S, Sinagra G, et al. Genetic variation in titin in arrhythmogenic right ventricular cardiomyopathy-overlap syndromes. Circulation 2011;124:876–85. 
    Crossref | PubMed
  76. Siragam V, Cui X, Masse S, et al. TMEM43 mutation p.S358L alters intercalated disc protein expression and reduces conduction velocity in arrhythmogenic right ventricular cardiomyopathy. PLoS One 2014;9:e109128.
    Crossref | PubMed
  77. Fabritz L, Hoogendijk MG, Scicluna BP, et al. Load-reducing therapy prevents development of arrhythmogenic right ventricular cardiomyopathy in plakoglobin-deficient mice. J Am Coll Cardiol 2011;57:740–50. 
    Crossref | PubMed
  78. Kirchhof P, Fabritz L, Zwiener M, et al. Age- and training dependent development of arrhythmogenic right ventricular cardiomyopathy in heterozygous plakoglobin-deficient mice. Circulation 2006;114:1799–806. 
    Crossref | PubMed
  79. Cruz FM, Sanz-Rosa D, Roche-Molina M, et al. Exercise triggers ARVC phenotype in mice expressing a disease causing mutated version of human plakophilin-2. J Am Coll Cardiol 2015;65:1438–50.
    Crossref | PubMed
  80. Saberniak J, Hasselberg NE, Borgquist R, et al. Vigorous physical activity impairs myocardial function in patients with arrhythmogenic right ventricular cardiomyopathy and in mutation positive family members. Eur J Heart Fail 2014;16:1337–44.
    Crossref | PubMed
  81. H Fischer A, van der Loo B, M Shär G, et al. Serological evidence for the association of Bartonella henselae infection with arrhythmogenic right ventricular cardiomyopathy. Clin Cardiol 2008;31:469–71.
    Crossref | PubMed
  82. De Pasquale CG, Heddle WF. Left sided arrhythmogenic ventricular dysplasia in siblings. Heart 2001;86:128–30. PMCID: PMC1729841
    Crossref | PubMed
  83. Michalodimitrakis M, Papadomanolakis A, Stiakakis J, Kanaki K. Left side right ventricular cardiomyopathy. Med Sci Law 2002;42:313–7.
    PubMed
  84. Mast TP, Teske AJ, vd Heijden JF, et al. Left Ventricular Involvement in Arrhythmogenic Right Ventricular Dysplasia/ Cardiomyopathy Assessed by Echocardiography Predicts Adverse Clinical Outcome. J Am Soc Echocardiogr 2015;28:1103– 13.e1109.
    Crossref | PubMed
  85. Abecasis J, Masci PG, Aquaro GD, et al. Arrhythmogenic biventricular dysplasia? Rev Port Cardiol 2009;28:1459–63.
    PubMed
  86. Burke AP, Farb A, Tashko G, Virmani R. Arrhythmogenic right ventricular cardiomyopathy and fatty replacement of the right ventricular myocardium: are they different diseases? Circulation 1998;97:1571–80.
    Crossref | PubMed
  87. Te Riele AS, James CA, Philips B, et al. Mutation-positive arrhythmogenic right ventricular dysplasia/cardiomyopathy: the triangle of dysplasia displaced. J Cardiovasc Electrophysiol 2013;24:1311–20.
    Crossref | PubMed
  88. Basso C, Corrado D, Bauce B, Thiene G. Arrhythmogenic Right Ventricular Cardiomyopathy. Circ Arrhythm Electrophysiol 2012;5(6):1233–46.
    Crossref | PubMed
  89. Sen-Chowdhry S, Syrris P, Prasad SK, et al. Left-dominant arrhythmogenic cardiomyopathy: an under-recognized clinical entity. J Am Coll Cardiol 2008;52:2175–87.
    Crossref | PubMed
  90. Kjaergaard J, Hastrup Svendsen J, Sogaard P, et al. Advanced quantitative echocardiography in arrhythmogenic right ventricular cardiomyopathy. J Am Soc Echocardiogr 2007;20: 27–35. 
    Crossref | PubMed
  91. Saguner AM, Medeiros-Domingo A, Schwyzer MA, et al. Usefulness of inducible ventricular tachycardia to predict long-term adverse outcomes in arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol 2013;111 :250–7.
    Crossref | PubMed
  92. Dechering DG, Kochhauser S, Wasmer K, et al. Electrophysiological characteristics of ventricular tachyarrhythmias in cardiac sarcoidosis versus arrhythmogenic right ventricular cardiomyopathy. Heart Rhythm 2013;10:158–64.
    Crossref | PubMed
  93. Zorzi A, Rigato I, Pilichou K, et al. Phenotypic expression is a prerequisite for malignant arrhythmic events and sudden cardiac death in arrhythmogenic right ventricular cardiomyopathy. Europace 2015;18:1086–94.
    Crossref | PubMed
  94. Basso C, Corrado D, Marcus FI, et al. Arrhythmogenic right ventricular cardiomyopathy. Lancet 2009;373:1289–300. 
    Crossref | PubMed
  95. Chu AF, Zado E, Marchlinski FE. Atrial arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy/ dysplasia and ventricular tachycardia. Am J Cardiol 2010;106:720–2. 
    Crossref | PubMed
  96. Camm CF, James CA, Tichnell C, et al. Prevalence of atrial arrhythmias in arrhythmogenic right ventricular dysplasia/ cardiomyopathy. Heart Rhythm 2013;10:1661–8. 
    Crossref | PubMed
  97. Saguner AM, Ganahl S, Kraus A, et al. Clinical role of atrial arrhythmias in patients with arrhythmogenic right ventricular dysplasia. Circ J 2014;78:2854-61. 
    Crossref | PubMed
  98. Zipes DP, Camm AJ, Borggrefe M, et al. [Guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death. Executive summary]. Rev Esp Cardiol 2006;59:1328.  
    PubMed
  99. Tabib A, Loire R, Chalabreysse L, et al. Circumstances of death and gross and microscopic observations in a series of 200 cases of sudden death associated with arrhythmogenic right ventricular cardiomyopathy and/or dysplasia. Circulation 2003;108:3000–5.
    Crossref | PubMed
  100. Uberoi A, Stein R, Perez MV, et al. Interpretation of the electrocardiogram of young athletes. Circulation 2011;124: 746–57.
    Crossref | PubMed
  101. Nava A, Bauce B, Basso C, et al. Clinical profile and long-term follow-up of 37 families with arrhythmogenic right ventricular cardiomyopathy. J Am Coll Cardiol 2000;36:2226–33.
    Crossref | PubMed
  102. Marcus FI, McKenna WJ, Sherrill D, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the Task Force Criteria. Eur Heart J 2010;31 :806–14. 
    Crossref | PubMed
  103. McKenna WJ, Thiene G, Nava A, et al. Diagnosis of arrhythmogenic right ventricular dysplasia/cardiomyopathy. Task Force of the Working Group Myocardial and Pericardial Disease of the European Society of Cardiology and of the Scientific Council on Cardiomyopathies of the International Society and Federation of Cardiology. Br Heart J 1994;71 :215–8.PMCID: PMC483655
    Crossref | PubMed
  104. Cox MG, van der Zwaag PA, van der Werf C, et al. Arrhythmogenic right ventricular dysplasia/cardiomyopathy: pathogenic desmosome mutations in index-patients predict outcome of family screening: Dutch arrhythmogenic right ventricular dysplasia/cardiomyopathy genotype-phenotype follow-up study. Circulation 2011;123:2690–700.
    Crossref | PubMed
  105. Vermes E, Strohm O, Otmani A, et al. Impact of the revision of arrhythmogenic right ventricular cardiomyopathy/ dysplasia task force criteria on its prevalence by CMR criteria. JACC Cardiovasc Imaging 2011;4:282–7.
    Crossref | PubMed
  106. Szymanski P, Klisiewicz A, Hoffman P. ARVC/D task force imaging criteria: it is difficult to get along with the guidelines. JACC Cardiovasc Imaging 2011;4:686.
    Crossref | PubMed
  107. Steriotis AK, Bauce B, Daliento L, et al. Electrocardiographic pattern in arrhythmogenic right ventricular cardiomyopathy. Am J Cardiol 2009;103:1302–8.
    Crossref | PubMed
  108. Saguner AM, Ganahl S, Baldinger SH, et al. Usefulness of electrocardiographic parameters for risk prediction in arrhythmogenic right ventricular dysplasia. Am J Cardiol 2014;113:1728–34. 
    Crossref | PubMed
  109. Platonov PG, Calkins H, Hauer RN, et al. High interobserver variability in the assessment of epsilon waves: Implications for diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia. Heart Rhythm 2016;13:208–16.
    Crossref | PubMed
  110. Quarta G, Ward D, Tome Esteban MT, et al. Dynamic electrocardiographic changes in patients with arrhythmogenic right ventricular cardiomyopathy. Heart 2010;96:516–22. 
    Crossref | PubMed
  111. Fontaine G, Frank R, Tonet JL, et al. Arrhythmogenic right ventricular dysplasia: a clinical model for the study of chronic ventricular tachycardia. Jpn Circ J 1984;48:515–38.
    Crossref | PubMed
  112. Kinoshita O, Fontaine G, Rosas F, et al. Time- and frequency domain analyses of the signal-averaged ECG in patients with arrhythmogenic right ventricular dysplasia. Circulation 1995;91 :715–21.
    Crossref | PubMed
  113. Perrin MJ, Angaran P, Laksman Z, et al. Exercise testing in asymptomatic gene carriers exposes a latent electrical substrate of arrhythmogenic right ventricular cardiomyopathy. J Am Coll Cardiol 2013;62:1772–9.
    Crossref | PubMed
  114. Denis A, Sacher F, Derval N, et al. Diagnostic value of isoproterenol testing in arrhythmogenic right ventricular cardiomyopathy. Circ Arrhythm Electrophysiol 2014;7:590–7. 
    Crossref | PubMed
  115. Baran A, Nanda NC, Falkoff M, et al. Two-dimensional echocardiographic detection of arrhythmogenic right ventricular dysplasia. Am Heart J 1982;103:1066–7.
    Crossref | PubMed
  116. Yoerger DM, Marcus F, Sherrill D, 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;45:860–5.
    Crossref | PubMed
  117. Pinamonti B, Pagnan L, Bussani R, et al. Right ventricular dysplasia with biventricular involvement. Circulation 1998;98:1943–5. 
    Crossref | PubMed
  118. Lindstrom L, Nylander E, Larsson H, Wranne B. Left ventricular involvement in arrhythmogenic right ventricular cardiomyopathy – a scintigraphic and echocardiographic study. Clin Physiol Funct Imaging 2005;25:171–7.
    Crossref | PubMed
  119. Pennell D, Casolo G. Right ventricular arrhythmia: emergence of magnetic resonance imaging as an investigative tool. Eur Heart J 1997;18:1843–5.
    Crossref | PubMed
  120. Dalal D, Tandri H, Judge DP, et al. Morphologic variants of familial arrhythmogenic right ventricular dysplasia/ cardiomyopathy a genetics-magnetic resonance imaging correlation study. J Am Coll Cardiol 2009;53:1289–99.
    Crossref | PubMed
  121. Tandri H, Calkins H, Marcus FI. Controversial role of magnetic resonance imaging in the diagnosis of arrhythmogenic right ventricular dysplasia. Am J Cardiol 2003;92:649.
    Crossref | PubMed
  122. Bluemke DA, Krupinski EA, Ovitt T, et al. MR Imaging of arrhythmogenic right ventricular cardiomyopathy: morphologic findings and interobserver reliability. Cardiology 2003;99:153–62.  
    Crossref | PubMed
  123. Prakasa KR, Wang J, Tandri H, et al. Utility of tissue Doppler and strain echocardiography in arrhythmogenic right ventricular dysplasia/cardiomyopathy. Am J Cardiol 2007;100:507–12. 
    Crossref | PubMed
  124. Teske AJ, Cox MG, De Boeck BW, et al. Echocardiographic tissue deformation imaging quantifies abnormal regional right ventricular function in arrhythmogenic right ventricular dysplasia/cardiomyopathy. J Am Soc Echocardiogr 2009;22:920–7.
    Crossref | PubMed
  125. Jain A, Shehata ML, Stuber M, et al. Prevalence of left ventricular regional dysfunction in arrhythmogenic right ventricular dysplasia: a tagged MRI study. Circulation Cardiovascular imaging 2010;3:290–7. 
    Crossref | PubMed
  126. Teske AJ, Cox MG, Te Riele AS, et al. Early detection of regional functional abnormalities in asymptomatic ARVD/C gene carriers. J Am Soc Echocardiogr 2012;25:997–1006. 
    Crossref | PubMed
  127. Vitarelli A, Cortes Morichetti M, Capotosto L, et al. Utility of strain echocardiography at rest and after stress testing in arrhythmogenic right ventricular dysplasia. Am J Cardiol 2013;111 :1344–50.
    Crossref | PubMed
  128. Indik JH, Dallas WJ, Gear K, et al. Right ventricular volume analysis by angiography in right ventricular cardiomyopathy. Int J Cardiovasc Imaging 2011;28:995–1001.
    Crossref | PubMed
  129. Francés RJ. Arrhythmogenic right ventricular dysplasia/ cardiomyopathy. A review and update. Int J Cardiol 2006;110:279–87.
    Crossref | PubMed
  130. Corrado D, Basso C, Leoni L, et al. Three-dimensional electroanatomic voltage mapping increases accuracy of diagnosing arrhythmogenic right ventricular cardiomyopathy/ dysplasia. Circulation 2005;111 :3042–50.
    Crossref | PubMed
  131. Ejima K, Shoda M, Manaka T, Hagiwara N. Targeted endomyocardial biopsy using electroanatomical voltage mapping in the early stage of arrhythmogenic right ventricular cardiomyopathy. Europace 2009;11 :388–9.
    Crossref | PubMed
  132. Marchlinski FE, Callans DJ, Gottlieb CD, Zado E. Linear ablation lesions for control of unmappable ventricular tachycardia in patients with ischemic and nonischemic cardiomyopathy. Circulation 2000;101 :1288–96.
    Crossref | PubMed
  133. Sosa E, Scanavacca M, d›Avila A, Pilleggi F. A new technique to perform epicardial mapping in the electrophysiology laboratory. J Cardiovasc Electrophysiol 1996;7:531–6.
    Crossref | PubMed
  134. Bai R, Di Biase L, Shivkumar K, et al. Ablation of ventricular arrhythmias in arrhythmogenic right ventricular dysplasia/ cardiomyopathy: arrhythmia-free survival after endo epicardial substrate based mapping and ablation. Circ Arrhythm Electrophysiol 2011;4:478–85.
    Crossref | PubMed
  135. Garcia FC, Bazan V, Zado ES, et al. Epicardial substrate and outcome with epicardial ablation of ventricular tachycardia in arrhythmogenic right ventricular cardiomyopathy/ dysplasia. Circulation 2009;120:366–75.
    Crossref | PubMed
  136. Migliore F, Zorzi A, Silvano M, et al. Prognostic value of endocardial voltage mapping in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia. Circ Arrhythm Electrophysiol 2013;6:167–76. 
    Crossref | PubMed
  137. Asimaki A, Saffitz JE. The role of endomyocardial biopsy in ARVC: looking beyond histology in search of new diagnostic markers. J Cardiovasc Electrophysiol 2011;22:111–7.
    Crossref | PubMed
  138. Basso C, Ronco F, Marcus F, et al. Quantitative assessment of endomyocardial biopsy in arrhythmogenic right ventricular cardiomyopathy/dysplasia: an in vitro validation of diagnostic criteria. Eur Heart J 2008;29:2760–71.
    Crossref | PubMed
  139. Avella A, d›Amati G, Pappalardo A, et al. Diagnostic value of endomyocardial biopsy guided by electroanatomic voltage mapping in arrhythmogenic right ventricular cardiomyopathy/dysplasia. J Cardiovasc Electrophysiol 2008;19:1127–34.
    Crossref | PubMed
  140. Bauce B, Rampazzo A, Basso C, et al. Clinical Phenotype and Diagnosis of Arrhythmogenic Right Ventricular Cardiomyopathy in Paediatric Patients Carrying Desmosomal Gene Mutations. Heart Rhythm 2011;8:1686–95.
    Crossref | PubMed
  141. Sen-Chowdhry S, Syrris P, McKenna WJ. Role of genetic analysis in the management of patients with arrhythmogenic right ventricular dysplasia/cardiomyopathy. J Am Coll Cardiol 2007;50:1813–21.
    Crossref | PubMed
  142. Basso C, Carturan E, Pilichou K, et al. Sudden cardiac death with normal heart: molecular autopsy. Cardiovasc Pathol 2010;19:321–5.
    Crossref | PubMed
  143. Basso C, Wichter T, Danieli GA, et al. Arrhythmogenic right ventricular cardiomyopathy: clinical registry and database, evaluation of therapies, pathology registry, DNA banking. Eur Heart J 2004;25:531–4. 
    Crossref | PubMed
  144. Corrado D, Wichter T, Link MS, et al. Treatment of arrhythmogenic right ventricular cardiomyopathy/dysplasia: an international task force consensus statement. Eur Heart J 2015;36:3227–37. 
    Crossref | PubMed
  145. O, Donnell D, Cox D, Bourke J, et al. Clinical and electrophysiological differences between patients with arrhythmogenic right ventricular dysplasia and right ventricular outflow tract tachycardia. Eur Heart J 2003;24: 801–10.
    Crossref | PubMed
  146. Liao Z, Zhan X, Wu S, et al. Idiopathic Ventricular Arrhythmias Originating From the Pulmonary Sinus Cusp: Prevalence, Electrocardiographic/Electrophysiological Characteristics, and Catheter Ablation. J Am Coll Cardiol 2015;66:2633–44.
    Crossref | PubMed
  147. Hoffmayer KS, Bhave PD, Marcus GM, et al. An electrocardiographic scoring system for distinguishing right ventricular outflow tract arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy from idiopathic ventricular tachycardia. Heart Rhythm 2013;10: 477–82.
    Crossref | PubMed
  148. Ainsworth CD, Skanes AC, Klein GJ, et al. Differentiating arrhythmogenic right ventricular cardiomyopathy from right ventricular outflow tract ventricular tachycardia using multilead QRS duration and axis. Heart Rhythm 2006;3:416–23.
    Crossref | PubMed
  149. Hoffmayer KS, Machado ON, Marcus GM, et al. Electrocardiographic comparison of ventricular arrhythmias in patients with arrhythmogenic right ventricular cardiomyopathy and right ventricular outflow tract tachycardia. J Am Coll Cardiol 2011;58:831–8.
    Crossref | PubMed
  150. Pieroni M, Dello Russo A, Marzo F, et al. High prevalence of myocarditis mimicking arrhythmogenic right ventricular cardiomyopathy differential diagnosis by electroanatomic mapping-guided endomyocardial biopsy. J Am Coll Cardiol 2009;53:681–9.
    Crossref | PubMed
  151. Vasaiwala SC, Finn C, Delpriore J, et al. Prospective study of cardiac sarcoid mimicking arrhythmogenic right ventricular dysplasia. J Cardiovasc Electrophysiol 2009;20:473–6.
    Crossref | PubMed
  152. Quarta G, Syrris P, Ashworth M, et al. Mutations in the Lamin A/C gene mimic arrhythmogenic right ventricular cardiomyopathy. Eur Heart J 2012;33:1128–36.
    Crossref | PubMed
  153. Cerrone M, Lin X, Zhang M, et al. Missense mutations in plakophilin-2 cause sodium current deficit and associate with a Brugada syndrome phenotype. Circulation 2014;129:1092–103.
    Crossref | PubMed
  154. Heidbüchel H, La Gerche A. The right heart in athletes. Evidence for exercise-induced arrhythmogenic right ventricular cardiomyopathy. Herzschrittmacherther Elektrophysiol 2012;23:82–6. 
    PubMed
  155. Hulot JS, Jouven X, Empana JP, et al. Natural history and risk stratification of arrhythmogenic right ventricular dysplasia/cardiomyopathy. Circulation 2004;110:1879–84. 
    Crossref | PubMed
  156. Corrado D, Basso C, Pavei A, et al. Trends in sudden cardiovascular death in young competitive athletes after implementation of a preparticipation screening program. JAMA 2006;296:1593–601. 
    Crossref | PubMed
  157. Sawant AC, Te Riele AS, Tichnell C, et al. Safety of American Heart Association-recommended minimum exercise for desmosomal mutation carriers. Heart Rhythm 2016;13: 199–207. 
    Crossref | PubMed
  158. Rigato I, Corrado D, Basso C, et al. Pharmacotherapy and other therapeutic modalities for managing Arrhythmogenic Right Ventricular Cardiomyopathy. Cardiovasc Drugs Ther 2015;29:171–7. 
    Crossref | PubMed
  159. Corrado D, Leoni L, Link MS, et al. Implantable cardioverterdefibrillator therapy for prevention of sudden death in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia. Circulation 2003;108:3084–91.
    Crossref | PubMed
  160. Priori SG, Blomström-Lundqvist C, Mazzanti A, et al. 2015 ESC Guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: The Task Force for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death of the European Society of Cardiology (ESC). Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC). Eur Heart J 2015;36:2793–867.
    Crossref | PubMed
  161. Epstein AE, DiMarco JP, Ellenbogen KA, et al. ACC/ AHA/HRS 2008 Guidelines for Device-Based Therapy of Cardiac Rhythm Abnormalities: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to Revise the ACC/AHA/NASPE 2002 Guideline Update for Implantation of Cardiac Pacemakers and Antiarrhythmia Devices): developed in collaboration with the American Association for Thoracic Surgery and Society of Thoracic Surgeons. Circulation 2008;117:e350–408.
    Crossref | PubMed