Review Article

Arrhythmogenic Cardiomyopathy: A Disease or Merely a Phenotype?

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.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Arrhythmogenic cardiomyopathy (AC) is a clinical entity that has evolved conceptually over the past 30 years. Advances in cardiac imaging and the introduction of genetics into everyday practice have revealed that AC comprises multiple phenotypes that are dependent on genetic or acquired factors. In this study, the authors summarise the approach to the identification of the AC phenotype and its underlying causes. They believe that AC represents a paradigm for personalised medicine in cardiology and that better stratification of the disease will enhance the development of mechanism-based treatments.

Disclosure:PME is on the European Cardiology Review editorial board; this did not influence peer review. AP has no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Perry Elliott, UCL Institute for Cardiovascular Science, Paul O’Gorman Building, University College London, 72 Huntley St, London WC1E 6DD, UK. E: perry.elliott@ucl.ac.uk

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Until the 20th century, conventional medicine relied almost exclusively on observable characteristics to classify and treat human disease.1 Even in the modern era, physicians define diseases using phenotypic similarities and employ relatively simple algorithms to interpret diagnostic tools and plan treatment. The advent of low-cost genetic sequencing and its introduction into clinical care provides new insights into pathogenesis and improves the recognition of inherited diseases and complex associated entities. In this article, we review a relatively new concept, arrhythmogenic cardiomyopathy (AC), and explain why it is an exemplar for a more nuanced approach to diagnosis that promises more effective treatments.

The Paradigm of Arrhythmogenic Right Ventricular Cardiomyopathy

In 1982, Marcus et al. described a group of patients with right ventricular dysplasia who presented with ventricular tachycardias of left bundle branch block configuration, T-wave inversion in the right precordial leads and structural abnormalities of the right ventricle (RV).2 In cases of sudden death, histological examination showed fibrofatty replacement of the right ventricular myocardium combined with signs of myocardial degeneration and necrosis, with and without inflammation, which led to the adoption of the term ‘arrhythmogenic right ventricular cardiomyopathy’ (ARVC), rather than ‘dysplasia’ to describe this new potentially lethal myocardial disorder.3

The familial nature of the disease was recognised very early and inspired the identification of the first genetic mutation in ARVC, which was a two-nucleotide pair deletion in the junction plakoglobin (JUP) gene in patients with Naxos disease, a recessive syndromic form of ARVC associated with palmoplantar keratoderma and woolly hair.4,5 Around the same time, a recessive mutation in the desmoplakin (DSP) gene was identified as the cause of an autosomal recessive cardiocutaneous syndrome in Ecuadorian families known as Carvajal syndrome.6–8 Both JUP and DSP are key components of the myocyte intercalated disc, and candidate gene evaluation in families with more common dominant forms of ARVC soon revealed mutations in DSP and other desmosomal genes, including plakophilin-2 (PKP-2), desmocollin-2 (DSC-2) and desmoglein-2 (DSG-2), forming the concept of ARVC as a disease of the desmosome.9

The Emergence of Nosological Confusion

Even in early reports of ARVC, fibrous/fibrofatty lesions were found in the left ventricular myocardium, as well as the right.2,3 Electrocardiograms from patients with ARVC often demonstrated features consistent with left ventricular involvement and cardiac imaging occasionally demonstrated coexistent wall motion abnormalities, dilation and systolic dysfunction.2,5 With the widespread use of cardiac MRI to assess patients with suspected ARVC, it became apparent that subtle left ventricle (LV) disease in the form of myocardial fibrosis is almost the norm, particularly in patients with mutations in DSP and DSG2.10 In response to this, many investigators started to use terms such as ‘arrhythmogenic left ventricular cardiomyopathy’, and even more confusingly, ‘left-dominant arrhythmogenic cardiomyopathy’, to describe this broader spectrum of disease. However, this effort to preserve the concept of ARVC as originally described in the 1980s and 1990s has become increasingly tenuous with the demonstration of overlapping phenotypes, such as dilated cardiomyopathy and non-desmosomal gene mutations, such as phospholamban (PLN), transmembrane protein 43 (TMEM43), filamin C (FLNC), desmin, cadherin 2 (CDH2), lamin A/C (LMNA) and titin (TTN) in individuals fulfilling current diagnostic criteria for ARVC, as well as other phenotypes (Figure 1).11–17

The Type of Ventricular Involvement in Genetic Subgroups with Arrhythmogenic Cardiomyopathy

Article image

Clinical Presentations and Differential Diagnoses

Article image

The Arrhythmogenic Cardiomyopathy Phenotype

Phenotypes describe a set of identifiable clinical characteristics that help to distinguish affected individuals from unaffected. The term AC represents an evolution of diagnostic terms describing disease of the LV and RV, but it was only recently that the term was formally used to describe this disorder.18

Although diagnostic criteria for the expanded AC spectrum do not yet exist, it is possible to summarise the key components of the AC phenotype:

  • Structural abnormalities of the myocardium, including regional or global systolic dysfunction or myocardial scar in either ventricle, unexplained by ischaemic heart disease, hypertension or an acute inflammatory trigger, such as viral infection or exposure to a toxic agent.
  • Frequent ventricular ectopy, sustained or non-sustained ventricular tachycardia (VT) or unexplained cardiac arrest.
  • A family history of sudden cardiac death or cardiomyopathy and a history of myocarditis presentation are also key features (Figure 2).

When defined in this way, the term AC serves as the first step towards disease classification and elucidation of aetiology. Contemporary studies show that AC encompasses a broad spectrum of genetic and non-genetic diseases, each with its own characteristic features and clinical course. A detailed description of all contenders for the label of AC is beyond the scope of this article, but two groups deserve emphasis due to their potential to cause premature death (often in young people) and their influence on therapy.

Arrhythmogenic Dilated Cardiomyopathy

It is important to appreciate that, by definition, the AC phenotype overlaps with that of dilated cardiomyopathy (DCM). DCM is currently defined only by LV size and function. However, a subgroup of patients characterised by syncope, rapid non-sustained ventricular tachycardia, increased ventricular ectopy and a family history of sudden cardiac death has been described.19 The term ‘arrhythmogenic DCM’ has been used to describe these cases, but in practice it aligns with the definition of AC.20

Arrhythmogenic DCM includes many genetic forms that are characterised mainly by dilatation and dysfunction of the LV. Examples include disease caused by mutations in desmosomal genes, such as DSP and LMNA encoding the nuclear envelope protein lamin AC.21,22 The latter may also present with conduction abnormalities (atrioventricular node and/or left bundle branch disease) and clinical or subclinical skeletal myopathy. An increased risk of sudden cardiac death has also been identified in cases with mutations in desmosomal genes and LMNA, as compared to other genetic and non-genetic causes of DCM.23,24

Myocardial Inflammation

Ever since the first reports of AC, inflammatory cell infiltrates have been described in patients diagnosed at post-mortem, with the highest incidence in patients with more diffuse disease.3,25–29 Inflammatory infiltrates are also described in the transgenic models of AC.30,31 The subepicardial distribution of the typical AC lesions is very similar to that reported in myocarditis, and on occasion, patients with AC seek medical attention with symptoms of chest pain and palpitations accompanied by electrocardiographic changes and cardiac biomarkers consistent with myocarditis.32,33 These ‘hot phases’ can be the initial presentation of AC, sometimes preceding the development of classical ECG, imaging and histological features.34–37 18F-fluorodeoxyglucose PET can be positive in a subset of symptomatic AC patients, particularly in cases with left-dominant AC.32 Cardiac MRI in AC often demonstrates patterns of subepicardial scarring with a predilection for the inferior and lateral walls of the LV, similar to that seen following myocarditis.36–38 There are two studies showing raised cytokine levels in the blood of patients compared to controls.38,39

Algorithmic Representation of the Evaluation

Article image

Cardiac sarcoidosis can show marked resemblance to genetic forms of AC, and the differential diagnosis is sometimes only made at a histological level.32 Furthermore, a link at the molecular level is also present, as both giant cell myocarditis and sarcoidosis exhibit a similar pattern of reduced plakoglobin staining in the absence of any mutations in the desmosomal genes.38 Finally, there is recent evidence of cardiac autoimmunity in a study of AC reporting anti-desmoglein antibodies associated with the extent of the disease.40 Importantly, this was a consistent finding among desmosomal and non-desmosomal forms of the disease.40

Approach to Diagnosis

Identifying phenotypes and defining the discrepancies between them might seem as an overly academic and mundane task; however, these concepts and their application are fundamental to treatment decisions and guide clinical and basic research.

The phenotypic spectrum encompassed by ‘classic’ ARVC, biventricular AC and left-dominant AC challenges the development of a single set of diagnostic criteria. However, a stepwise approach to first identify the broad AC phenotype, as defined above, and then to characterise the genetic and non-genetic causes is possible in everyday practice, although the involvement of different specialities, such as clinical genetics, microbiology, immunology and haematology, is also essential in specific cases (Figure 3).

In this way, it is possible to move beyond the crude phenotypes currently used to define disease (e.g. heart failure with reduced or preserved ejection fraction) towards personalised algorithms for investigation and management.

Opportunities for Improved Management and New Therapies

The elucidation of specific subtypes of AC offers opportunities for tailored therapies. Examples where this is already possible include arrhythmia risk stratification, myocarditis treatment and the role of exercise.

Arrhythmia Risk Stratification

Risk stratification in AC is critical, as ICDs provide a life-saving therapy for patients prone to haemodynamically unstable ventricular arrhythmias.41 It is uncontentious that patients with previous unstable ventricular arrhythmias are at high risk of recurrence and an ICD is always recommended for secondary prevention.41 Although there have been numerous studies on arrhythmic risk stratification in affected individuals, and particularly family members from cascade screening with no previous arrhythmic events, the timing of ICD implantation remains a challenge.42–45 A new prediction model for ventricular arrhythmias (https://arvcrisk.com) has been recently developed and published by a multi-centre cohort. However, this is based only on individuals with a definite diagnosis of ARVC based on the 2010 Task Force criteria, and its utility remains obscure for the left-dominant forms.42

In a recent report, the Heart Rhythm Society provided consensus recommendations on the management of different forms of AC using a genotype-guided approach.18 Evidence of increased arrhythmic risk in cases with only mildly impaired LV systolic function exist for specific genes, such as PLN, LMNA and FLNC, and ICD implantation should be considered, irrespective of symptoms, in such cases.18 In the case of LMNA, additional parameters, such as male sex and non-sustained VT, can guide decision-making.

Myocarditis Treatment

As mentioned previously, myocardial inflammation is an important component of the AC spectrum.32,46 Identification of specific forms of inflammation, such as sarcoidosis, can be challenging, as a definite diagnosis can be reached only by endomyocardial biopsy, but is critical as it can dictate further management with corticosteroid therapy and long-term immunosuppression.47–49 The effects of anti-inflammatory therapy in gene-positive individuals presenting with a ‘hot phase’ remains to be investigated.

The increasing recognition of myocardial inflammation as a mechanism of disease progression in genetic forms of AC raises several possibilities in regards to therapeutic approaches.37 Although conventional treatment options for myocarditis, such intravenous immunoglobulin, corticosteroids, cyclosporine and azathioprine, can be considered, they remain largely non-selective in terms of the mechanisms and the sites of inflammation that they target.50,51 The elucidation of the cellular mechanisms in the various genetic forms of AC will be crucial in the identification of the potential causes of inflammatory induction that can be targeted therapeutically.

The Role of Exercise

It is common to elicit a history of high-intensity exercise among individuals affected by classic ARVC. Furthermore, patients with ARVC who were athletes have been found to have increased risk of sudden death compared with non-athletes.52 Desmosomal mutation carriers who participate in endurance and frequent exercise have a higher incidence of ventricular tachyarrhythmias and heart failure compared to those who are less athletic.53 A history of vigorous exercise seems to have an even greater effect in patients without an identifiable mutation, and patients seem to have greater benefit from exercise restriction.54,55 Further research is warranted to better understand the role of exercise in other subtypes of AC and non-desmosomal mutation carriers to prescribe exercise restriction more effectively.

Future Developments in Mechanism-based
Therapies

Recent studies have revealed a key role of the glycogen synthase kinase-3 beta (GSK3B) in the pathogenesis of AC.31 In the hearts of patients with AC, it has been shown that GSK3B translocates from the cytoplasm, where it is present in controls, to the intercalated disc.31 This observation was consistent among all cases with desmosomal gene mutations, including DSP, but less consistent in FLNC and PLN mutation carriers.56,57 The inhibition of GSK3B led to rescue of the phenotype when administered early in transgenic mice with mutant DSG2 or JUP.31

In disease caused by LMNA mutations, a selective p38 mitogen-activated protein kinase inhibitor, ARRY-797, has shown reversal of cardiac dysfunction in a transgenic murine model of LMNA. A Phase III clinical trial is now underway in patients with DCM caused by LMNA mutations (NCT03439514).

Examples of other molecular-targeted treatments include gene therapy enabling trans-splicing in a LMNA-related muscular dystrophy mouse model with partial rescue of the mutant phenotype.58 Antisense-mediated exon skipping can remove mutant exons with frameshift or nonsense variants in cardiomyopathy genes, and has been used to eliminate TTN truncating variants in patient-derived induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM).59 Gene correction using transcription activator-like effector nucleases has successfully corrected PLN variants in iPSC-CM by establishing the biallelic expression of the normal PLN transcripts.60

Conclusion

In the era of ‘-omic’ technologies and precision medicine, there are increasing opportunities to redefine diseases and test new therapies. AC serves as a prime example of a heterogeneous family of disorders in which there are multiple genetic and non-genetic causes amenable to existing and developing therapies that promise to slow, reverse and perhaps prevent disease.

References

  1. Protonotarios A, Elliott PM. Arrhythmogenic cardiomyopathies (ACs): diagnosis, risk stratification and management. Heart 2019;105:1117–28.
    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. Basso C, Thiene G, Corrado D, et al. Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, or myocarditis? Circulation 1996;94:983–91.
    Crossref | PubMed
  4. McKoy G, Protonotarios N, Crosby A, et al. Identification of a deletion in plakoglobin in arrhythmogenic right ventricular cardiomyopathy with palmoplantar keratoderma and woolly hair (Naxos disease). Lancet 2000;355:2119–24.
    Crossref | PubMed
  5. Protonotarios N, Tsatsopoulou A, Patsourakos P, et al. Cardiac abnormalities in familial palmoplantar keratosis. Br Heart J 1986;56:321–6.
    Crossref | PubMed
  6. Norgett EE, Hatsell SJ, Carvajal-Huerta L, et al. Recessive mutation in desmoplakin disrupts desmoplakin-intermediate filament interactions and causes dilated cardiomyopathy, woolly hair and keratoderma. Hum Mol Genet 2000;9:2761–6.
    Crossref | PubMed
  7. Carvajal-Huerta L. Epidermolytic palmoplantar keratoderma with woolly hair and dilated cardiomyopathy. J Am Acad Dermatol 1998;39:418–21.
    Crossref | PubMed
  8. Kaplan SR, Gard JJ, Carvajal-Huerta L, et al. Structural and molecular pathology of the heart in Carvajal syndrome. Cardiovasc Pathol 2004;13:26–32.
    Crossref | PubMed
  9. Karmouch J, Protonotarios A, Syrris P. Genetic basis of arrhythmogenic cardiomyopathy. Curr Opin Cardiol 2018;33:276–81.
    Crossref | PubMed
  10. Sen-Chowdhry S, Syrris P, Ward D, et al. Clinical and genetic characterization of families with arrhythmogenic right ventricular dysplasia/cardiomyopathy provides novel insights into patterns of disease expression. Circulation 2007;115:1710–20.
    Crossref | PubMed
  11. 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
  12. 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
  13. van Tintelen JP, Van Gelder IC, Asimaki A, 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;6:1574–83.
    Crossref | PubMed
  14. Ortiz-Genga MF, Cuenca S, Dal Ferro M, et al. Truncating FLNC mutations are associated with high-risk dilated and arrhythmogenic cardiomyopathies. J Am Coll Cardiol 2016;68:2440–51.
    Crossref | PubMed
  15. 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
  16. Mayosi BM, Fish M, Shaboodien G, et al. Identification of cadherin 2 (CDH2) mutations in arrhythmogenic right ventricular cardiomyopathy. Circ Cardiovasc Genet 2017;10: e001605.
    Crossref | PubMed
  17. 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
  18. Towbin JA, McKenna WJ, Abrams DJ, et al. 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy. Heart Rhythm 2019;16:e301–72.
    Crossref | PubMed
  19. Spezzacatene A, Sinagra G, Merlo M, et al. Arrhythmogenic phenotype in dilated cardiomyopathy: natural history and predictors of life-threatening arrhythmias. J Am Heart Assoc 2015;4:e002149.
    Crossref | PubMed
  20. Pinto YM, Elliott PM, Arbustini E, 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;37:1850–8.
    Crossref | PubMed
  21. Elliott P, O’Mahony C, Syrris P, et al. Prevalence of desmosomal protein gene mutations in patients with dilated cardiomyopathy. Circ Cardiovasc Genet 2010;3:314–22.
    Crossref | PubMed
  22. Captur G, Arbustini E, Bonne G, et al. Lamin and the heart. Heart 2018;104:468–79.
    Crossref | PubMed
  23. Merlo M, Cannata A, Gobbo M, et al. Evolving concepts in dilated cardiomyopathy. Eur J Heart Fail 2018;20:228–39.
    Crossref | PubMed
  24. Gigli M, Merlo M, Graw SL, et al. Genetic risk of arrhythmic phenotypes in patients with dilated cardiomyopathy. J Am Coll Cardiol 2019;74:1480–90.
    Crossref | PubMed
  25. Fontaine G, Fontaliran F, Lascault G, et al. Congenital and acquired right ventricular dysplasia. Arch Mal Coeur Vaiss 1990;83:915–20 [in French].
    PubMed
  26. Lobo FV, Heggtveit HA, Butany J, et al. Right ventricular dysplasia: morphological findings in 13 cases. Can J Cardiol 1992;8:261–8.
    PubMed
  27. 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
  28. Fornes P, Ratel S, Lecomte D. Pathology of arrhythmogenic right ventricular cardiomyopathy/dysplasia – an autopsy study of 20 forensic cases. J Forensic Sci 1998;43:777–83.
    PubMed
  29. Campuzano O, Alcalde M, Iglesias A, et al. Arrhythmogenic right ventricular cardiomyopathy: severe structural alterations are associated with inflammation. J Clin Pathol 2012;65:1077–83.
    Crossref | PubMed
  30. Pilichou K, Remme CA, Basso C, et al. Myocyte necrosis underlies progressive myocardial dystrophy in mouse dsg2-related arrhythmogenic right ventricular cardiomyopathy. J Exp Med 2009;206:1787–802.
    Crossref | PubMed
  31. Chelko SP, Asimaki A, Andersen P, et al. Central role for GSK3beta in the pathogenesis of arrhythmogenic cardiomyopathy. JCI Insight 2016;1:85923.
    Crossref | PubMed
  32. Protonotarios A, Wicks E, Ashworth M, 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.
    Crossref | PubMed
  33. Charron P, Elliott PM, Gimeno JR, 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;39:1784–93.
    Crossref | PubMed
  34. Bauce B, Basso C, Rampazzo A, et al. Clinical profile of four families with arrhythmogenic right ventricular cardiomyopathy caused by dominant desmoplakin mutations. Eur Heart J 2005;26:1666–75.
    Crossref | PubMed
  35. Patrianakos AP, Protonotarios N, Nyktari E, et al. Arrhythmogenic right ventricular cardiomyopathy/dysplasia and troponin release. Myocarditis or the ‘hot phase’ of the disease? Int J Cardiol 2012;157:e26–8.
    Crossref | PubMed
  36. Mavrogeni S, Protonotarios N, Tsatsopoulou A, et al. Naxos disease evolution mimicking acute myocarditis: the role of cardiovascular magnetic resonance imaging. Int J Cardiol 2013;166:e14–5.
    Crossref | PubMed
  37. Lopez-Ayala JM, Pastor-Quirante F, Gonzalez-Carrillo J, et al. Genetics of myocarditis in arrhythmogenic right ventricular dysplasia. Heart Rhythm 2015;12:766–73.
    Crossref | PubMed
  38. Asimaki A, Tandri H, Duffy ER, et al. Altered desmosomal proteins in granulomatous myocarditis and potential pathogenic links to arrhythmogenic right ventricular cardiomyopathy. Circ Arrhyth Electrophysiol 2011;4:743–52.
    Crossref | PubMed
  39. Campian ME, Verberne HJ, Hardziyenka M, et al. Assessment of inflammation in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia. Eur J Nucl Med Mol Imaging 2010;37:2079–85.
    Crossref | PubMed
  40. Chatterjee D, Fatah M, Akdis D, et al. An autoantibody identifies arrhythmogenic right ventricular cardiomyopathy and participates in its pathogenesis. Eur Heart J 2018;39:3932–44.
    Crossref | PubMed
  41. 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
  42. Cadrin-Tourigny J, Bosman LP, Nozza A, et al. A new prediction model for ventricular arrhythmias in arrhythmogenic right ventricular cardiomyopathy. Eur Heart J 2019;40:1850–8.
    Crossref | PubMed
  43. Lie OH, Rootwelt-Norberg C, Dejgaard LA, et al. Prediction of life-threatening ventricular arrhythmia in patients with arrhythmogenic cardiomyopathy: a primary prevention cohort study. JACC Cardiovasc Imaging 2018;11:1377–86.
    Crossref | PubMed
  44. Vischer AS, Castelletti S, Syrris P, et al. Risk score for the exclusion of arrhythmic events in arrhythmogenic right ventricular cardiomyopathy at first presentation. Int J Cardiol 2019;290:100–5.
    Crossref | PubMed
  45. Maupain C, Badenco N, Pousset F, et al. Risk stratification in arrhythmogenic right ventricular cardiomyopathy/dysplasia without an implantable cardioverter-defibrillator. JACC Clin Electrophysiol 2018;4:757–68.
    Crossref | PubMed
  46. Philips B, Madhavan S, James CA, et al. Arrhythmogenic right ventricular dysplasia/cardiomyopathy and cardiac sarcoidosis: distinguishing features when the diagnosis is unclear. Circ Arrhythm Electrophysiol 2014;7:230–6.
    Crossref | PubMed
  47. Yazaki Y, Isobe M, Hiroe M, et al. Prognostic determinants of long-term survival in Japanese patients with cardiac sarcoidosis treated with prednisone. Am J Cardiol 2001;88:1006–10.
    Crossref | PubMed
  48. Sadek MM, Yung D, Birnie DH, et al. Corticosteroid therapy for cardiac sarcoidosis: a systematic review. Can J Cardiol 2013;29:1034–41.
    Crossref | PubMed
  49. Ballul T, Borie R, Crestani B, et al. Treatment of cardiac sarcoidosis: a comparative study of steroids and steroids plus immunosuppressive drugs. Int J Cardiol 2019;276:208–11.
    Crossref | PubMed
  50. Tschope C, Cooper LT, Torre-Amione G, Van Linthout S. Management of myocarditis-related cardiomyopathy in adults. Circ Res 2019;124:1568–83.
    Crossref | PubMed
  51. Stephenson E, Savvatis K, Mohiddin SA, Marelli-Berg FM. T-cell immunity in myocardial inflammation: pathogenic role and therapeutic manipulation. Br J Pharmacol 2017;174:3914–25.
    Crossref | PubMed
  52. Corrado D, Basso C, Rizzoli G, et al. Does sports activity enhance the risk of sudden death in adolescents and young adults? J Am Coll Cardiol 2003;42:1959–63.
    Crossref | PubMed
  53. James CA, Bhonsale A, Tichnell C, 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;62:1290–7.
    Crossref | PubMed
  54. 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
  55. Wang W, Orgeron G, Tichnell C, et al. Impact of exercise restriction on arrhythmic risk among patients with arrhythmogenic right ventricular cardiomyopathy. J Am Heart Assoc 2018;7:e008843.
    Crossref | PubMed
  56. Begay RL, Graw SL, Sinagra G, et al. Filamin C truncation mutations are associated with arrhythmogenic dilated cardiomyopathy and changes in the cell-cell adhesion structures. JACC Clin Electrophysiol 2018;4:504–14.
    Crossref | PubMed
  57. Te Rijdt WP, Asimaki A, Jongbloed JDH, et al. Distinct molecular signature of phospholamban p.Arg14del arrhythmogenic cardiomyopathy. Cardiovasc Pathol 2018;40:2–6.
    Crossref | PubMed
  58. Azibani F, Brull A, Arandel L, et al. Gene therapy via trans-splicing for LMNA-related congenital muscular dystrophy. Mol Ther Nucleic Acids 2018;10:376–86.
    Crossref | PubMed
  59. Gramlich M, Pane LS, Zhou Q, et al. Antisense-mediated exon skipping: a therapeutic strategy for titin-based dilated cardiomyopathy. EMBO Mol Med 2015;7:562–76.
    Crossref | PubMed
  60. Karakikes I, Stillitano F, Nonnenmacher M, et al. Correction of human phospholamban R14del mutation associated with cardiomyopathy using targeted nucleases and combination therapy. Nat Commun 2015;6:6955.
    Crossref | PubMed
  61. Elliott PM, Anastasakis A, Asimaki A, et al. Definition and treatment of arrhythmogenic cardiomyopathy: an updated expert panel report. Eur J Heart Fail 2019;21:955–64.
    Crossref | PubMed