Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Cardiol. Jan 26, 2022; 14(1): 29-39
Published online Jan 26, 2022. doi: 10.4330/wjc.v14.i1.29
Role of genetic testing in cardiomyopathies: Α primer for cardiologists
Georgia Vogiatzi, Emmanouil Vavuranakis, The Third Department of Cardiology, Sotiria Hospital, Athens 11527, Greece
George Lazaros, Emilia Lazarou, Dimitris Tousoulis, The First Department of Cardiology, Hippokration Hospital, Athens 11526, Greece
Evangelos Oikonomou, The First Department of Cardiology, Hippokration Hospital, Medical School of National and Kapodistrian University of Athens, Athens 11527, Greece
ORCID number: Georgia Vogiatzi (0000-0002-6290-8714); George Lazaros (0000-0003-4822-2393); Evangelos Oikonomou (0000-0001-8079-0599); Emilia Lazarou (0000-0003-4979-6452); Emmanouil Vavuranakis (0000-0002-5907-4392); Dimitris Tousoulis (0000-0001-7492-4984).
Author contributions: All authors contributed to the literature review, data collection and interpretation, and manuscript drafting and revision for important intellectual content; and all authors approved the final version.
Conflict-of-interest statement: The authors declare that there are no conflicts of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Georgia Vogiatzi, PhD, Consultant Physician-Scientist, The Third Department of Cardiology, Sotiria Hospital, Mesogeion 152, Athens 11527, Greece. gvogiatz@yahoo.gr
Received: May 31, 2021
Peer-review started: May 31, 2021
First decision: June 16, 2021
Revised: July 18, 2021
Accepted: January 11, 2022
Article in press: January 11, 2022
Published online: January 26, 2022
Processing time: 232 Days and 20.2 Hours

Abstract

Recent advances in cardiovascular genetics have transformed genetic testing into a valuable part of management of families with inherited cardiomyopathies. As novel mutations have been identified, understanding when to consider genetic testing has emerged as an important consideration in the management of these cases. Specific genetic testing has a paramount importance in the risk stratification of family members, in the prognosis of probands at higher risk of a serious phenotype expression, and finally in the identification of new mutations, all of which are discussed in this review. The indications for each type of cardiomyopathy are described, along with the limitations of genetic testing. Finally, the importance of public sharing of variants in large data sets is emphasized. The ultimate aim of this review is to present key messages about the genetic testing process in order to minimize potential harms and provide suggestions to specialized clinicians who act as a part of a multidisciplinary team in order to offer the best care to families with inherited cardiomyopathies.

Key Words: Cardiomyopathy, Genetic counselling, Genetic testing, Variant, Hereditary

Core Tip: In a considerable percentage of patients with cardiomyopathies, there is a genetic component. If the presence or severity of the cardiomyopathy cannot be explained by acquired causes, the genetic component should be investigated in order to reveal potential inherited forms of cardiomyopathies. The genetic testing process is also helpful to minimize potential harms and provide suggestions to specialized clinicians who act as a part of a multidisciplinary team, with the objective of offering the best care to families with inherited cardiomyopathies.



INTRODUCTION

Cardiomyopathies represent a group of disorders of the myocardium associated with cardiac dysfunction, aggravated by arrhythmias, heart failure, and sudden cardiac death (SCD). They can be classified according to their morphological and functional phenotypes, specifically as hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), restrictive cardiomyopathy (RCM), and arrhythmogenic right ventricular (ARVC)/arrhythmogenic cardiomyopathy (ACM), and as unclassified forms[1]. In a considerable percentage of patients with cardiomyopathies, there is a genetic component, even for cases in which interaction with environmental factors cannot be excluded[2-5]. If the presence or severity of the cardiomyopathy cannot be explained by acquired causes, the genetic component should be investigated in order to reveal potential inherited forms of cardiomyopathies.

The continual rapid progress of molecular techniques over the past 2 decades and the related increased choices of different genetic testing options have led to a dramatic increase in our understanding of the genetic architecture of these diseases[6,7]. For better understanding of the terms used in the genetic testing process, relevant definitions are provided in Table 1. The aim of this review is to discuss the benefits of genetic testing in cardiomyopathies and the current indications in diagnosis and prognostication of probands and risk stratification for family members, in order to promote provision of optimal care for patients and their families.

Table 1 Terminology of commonly used genetics vocabulary.
Term
Definition
AlleleOne of several alternative versions of a particular gene
HeterozygoteAn individual who has different alleles at a particular gene locus on homologous chromosomes (carrier of a single copy of the mutation)
MutationAny alteration in the inherited nucleic acid sequence of the genotype of an organism; a mutation considered in the context of a genetic disease usually refers to an alteration that causes a Mendelian disease
PenetranceProportion of individuals carrying a mutation who also express a cardiomyopathy phenotype
Genome sequencingSequencing of entire genome (coding and non-coding regions)
Exome sequencing Sequencing of the coding regions (exons)
Proband or index caseIndex case in the family, usually the one with the most severe phonotype
VariantA change in the DNA sequence which may or may not be disease-causing
Pathogenicity Process of determining whether a variant is causative or not
GENETIC TESTING TECHNIQUES

The type and the available technologies for gene-based sequencing have been in constant evolution; turnaround times are shorter, while their cost has been dramatically decreasing. Whole blood, dried blood spots, or tissue specimens regarding post-mortem examination could serve as acceptable specimens for genetic testing. Conventionally, genetic testing by Sanger sequencing, for individual genes has been per-formed[8]. Due to the genetically heterogeneous nature of cardiomyopathies and the development and availability of next-generation sequencing (NGS) technique in the clinical setting, a multi-gene panel instead of individual gene-testing is now desirable practice for these diseases. Therefore, Sanger sequencing and targeted analysis is only preferable for cascade testing, when a pathogenic or likely variant has been identified in the proband. The composition of gene panels varies and some laboratories propose larger gene panels. However, increasing the number of genes in the panel also possibly increases the likelihood of identifying variants of uncertain significance. It is therefore evident that the ordering cardiologist should be aware of the benefits and limitations of specific test types in order to select the most appropriate technique[9]. Currently, testing a small specific panel of genes is usually recom-mended for each well-defined phenotype. Together, these techniques provide excellent precision and accuracy to detect single nucleotide substitutions that produce missense, nonsense, and splice site mutations and small insertion/deletions. A subset of cases with large insertion or deletion variants or other structural DNA changes, whereas the above analyses turn out negative, might benefit from copy number assays using microarray or multiplex ligation-dependent probe amplification[8,9].

CURRENT RECOMMENDATIONS FOR EACH CARDIOMYOPATHY

The prevalence, inheritance pattern, genes and indications for genetic testing involved in specific cardiomyopathies are summarized in Table 2.

Table 2 Prevalence, inheritance pattern, genes and indications for genetic testing involved in specific cardiomyopathies.
Inherited CMP
Prevalence
Pattern of inheritance
Key genes
Diagnostic yield of genetic testing
Recommendation for genetic testing
HCM1 in 500ADMYH7, MYBPC3, TNNT2, TNNI3, TPM1, ACTC1, MYL2, MYL3, GLA, PRKAG2, LAMP2 30%-60%For any patient with clinical diagnosis of HCM; Familial screening with a mutation after identified in the index case
DCM1 in 2500AD, X-linkedDES, DMD, DSP, FLNC, LMNA, MYH7, PLN, RBM20, TNNI3, TNNT2, TTN, TPM120%-30%For patients with DCM and conduction disease and/or family history of SCD; Familial screening with a mutation after identified in the index case
ARVC1 in 2000-5000AD, AR DSC2, DSG2, DSP, JUP, PLN, TMEM4350%Familial screening with a mutation after identified in the index case
RCMRareAD, AR X-linked or mitochondrialTroponin; MYBPC3, MYL3UnknownFamilial screening with a mutation after identified in the index case
HCM

ΗCM, a disease of the sarcomere, is the most common inherited cardiomyopathy, with an autosomal-dominant type of transmission, leading to left ventricular (LV) hyper-trophy and diastolic dysfunction. It is most often caused by variants in genes encoding cardiac sarcomere proteins[10]. The phenotype ranges from asymptomatic forms to SCD as the first and only manifestation, as seen especially in young athletes, all depending on the penetrance of the disease-causing mutations. Genetic testing should include not only the most common sarcomere genes [β-myosin heavy chain (MYH7); myosin-binding protein C3 (MYBPC3); troponin T2; troponin I3; tropo-myosin; actin alpha cardiac muscle 1; myosin light chain 2; myosin light protein] but also genes causing rare syndromic diseases with a HCM-phenotype [α-galactosidase (Fabry disease); protein kinase AMP-activated non-catalytic subunit gamma 2 (PRKAG2) (PRKAG2-glycogen storage disease); lysosomal-associated membrane protein 2 (Danon disease)][11,12]. In some of these diseases, such as Fabry or Danon disease, the positive genetic test result may change the clinical care of the proband, such as to involve enzyme replacement therapy or a more aggressive clinical management, respectively[12]. The diagnostic yield for the proband with a definite clinical diagnosis of HCM is approximately 30%-60%, and even higher in individuals who have severe LV hypertrophy, a known family history of HCM, or who were diagnosed at a younger age. MYBPC3 and MYH7 account for approximately 80% of all cases with positive genetic test.

DCM

About 30% of DCM cases appear to be familial in origin, isolated, or as a part of a syndrome[13-15]. The mode of inheritance is mostly autosomal dominant (AD). However, sporadic forms of DCM could be caused by non-genetic factors (i.e., drugs, alcohol, viruses). Similar to HCM, the clinical severity of DCM is heterogeneous, so genetic testing is important for the surveillance of the asymptomatic genotype-positive carriers. Clinical screening combined with a three-generation pedigree of the proband are warranted in order to establish the need for genetic testing in DCM[5].

Over 100 genes have been implicated in DCM, all encoding sarcomeric and cytoskeleton proteins[16]. After the addition of titin (TTN) variants in genetic testing, the diagnoses of familial DCM increased by about 10%[17]. However, interpretation of the TTN mutations is challenging, due to the large size of the gene and the high frequency of benign variants in healthy populations. Mutations in the lamin A/C (LMNA) gene are also detected in approximately 4%-6% of familial DCM cases, causing a distinct phenotype characterized by systolic impairment together with progressive conduction disturbances and malignant arrhythmias[15]. DCM may also appear as a complication of neuromuscular diseases (e.g., Duchenne and Becker muscular dystrophy). As the mode of inheritance is X-linked recessive, genetic testing should be provided in mothers of probands with Duchenne or Becker, because carrier females may develop later on[18]. At this point, we should also emphasize that due to phenotype overlapping among cardiomyopathies, HCM and ACM genes are included in DCM panels.

ACM, including right ventricular

ARVC is currently considered as a subtype of the broader group of ACM, with fibro-fatty replacement of the ventricular myocytes[19]. Clinical manifestations may vary with age and stage of disease, from asymptomatic but at-risk for SCD to end-stage heart failure with symptomatic arrhythmias. It is considered familial, with AD inheritance; although, there are recessive forms (e.g., Naxos disease, Carvajal syndrome)[20]. Initially, it was identified as a disease caused by mutations in genes encoding desmosomal proteins. A pathogenic variant has been detected in up to 50% of cases referred for genetic testing who meet the 2010 Task Force criteria[3]. Mutations in the plakophilin-2 gene account for 20%-30% of cases, while variants in four other desmosomal genes (desmocollin 2; desmoglein 2; desmoplakin; junction plakoglobin) have also been detected. Changes in DNA sequence of the abovementioned genes should be cautiously approached, since different benign variants may present frequently in unaffected control populations[20].

Further genetic subtypes have been described recently; so, under the broad term of arrhythmogenic cardiomyopathies, additional genes should be included in the genetic testing, such as transmembrane protein 43, LMNA, filamin C, desmin, RNA-binding protein 20, phospholamban (PLN) and TTN genes. According to the revised Task Force criteria for the diagnosis of ARVC/ACM, for a definite diagnosis, two major criteria or one major and two minor criteria, or four minor criteria from different categories are needed. The presence of a disease-causing mutation is considered as a major criterion. However, in order to avoid confusing results, if the patient shows only one minor criterion from the revised Task Force Criteria, then genetic testing should not be recommended[3]. It is noteworthy that desmosome gene mutations have also been identified in patients diagnosed with DCM.

RCM

RCM is a rare form of cardiomyopathy, with a heterogeneous phenotype. Multiple causes have been identified, including infiltrative storage, non-infiltrative and endomyocardial diseases, many of which are associated with specific genes[21]. The most frequent variants that have been found in RCM cases are in genes known to cause HCM. Therefore, genetic testing for RCM should include HCM genes[22].

IMPORTANCE OF FAMILY HISTORY IN THE PROCESS OF GENETIC TESTING

Irrespective of the type of cardiomyopathy, thorough comprehension of the proband’s family history with an at least three-generation family pedigree and counselling should precede genetic testing[23]. Therefore, the family history is of paramount importance and should not be overlooked in clinical practice. Specifically, it reveals the phenotype of each member, the pattern of inheritance (AD, autosomal recessive, X-linked and maternal mitochondrial conditions) and enables the clinicians to provide preliminary recommendations for clinical surveillance[6,24]. Importantly, it also helps genetic counsellors to discuss the process of genetic testing with the proband and the family members, including providing realistic expectations of the findings and to develop a relationship of trust. Effective communication and psychological support are key points. Pre-genetic counselling constitutes an important clinical part of the medical management of such cases[4,25].

VARIANT INTERPRETATION AND RECLASSIFICATION

The most challenging aspect of the genetic testing process is the interpretation of results. According to the Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology, each variant should be classified according to specific criteria in one of the following categories: Benign, likely benign, variant of uncertain significance (VUS), likely pathogenic, or pathogenic (Table 3)[26]. Causality is based on various criteria, such as population frequency and type of identified variant (i.e., missense, nonsense, de novo, truncating splice site); information of segregation in a pedigree and in silico tools are also used. It is important to bear in mind that in order to avoid the vast amount of uncertainty, rather than choosing a binary ‘yes’ or ‘no’ outcome, it is preferable to gather and weigh-up sufficient lines of evidence. A key step in this direction is the public sharing of important variant information among laboratories, such as through scientific repositories (e.g., ClinVar) and the creation of large reference datasets[27]. Another important issue is the periodic reclassification of variants; although, guidelines on how this can be performed feasibly are lacking[28]. Certainly, the latest technologic advancements of NGS, together with bioinformatics software, allow accurate and rapid high-output sequencing of the human genome[29]. And, in the real world, the ability to efficiently sequence coding and non-coding regions has led to the production of multigene panels used in clinical practice currently.

Table 3 Definitions of the variant classifications.
Variant
Definition
Pathogenic Variant is disease-causing with > 99% confidence; Cascade genetic testing should be offered to family members
Likely pathogenicVariant is disease-causing with > 90%-95% confidence; Cascade genetic testing should be offered to family members
VUSVariant is considered uncertain with an unknown effect on clinical phenotype, as there is insufficient or conflicting evidence for pathogenicity; Cascade genetic testing cannot be offered to family members
Likely benignVariant is probably not disease-causing; Cascade genetic testing should not be offered to family members
BenignVariant is not disease-causing; Cascade genetic testing should be offered to family members

At this point, we should also note that the complexity of cardiomyopathies and the difficulties in defining a specific phenotype according to genetic causes present new challenges for cardiologists who manage these patients singlehandedly. Therefore, a multidisciplinary team is required for genetic counselling and testing, as well as psychological support. This can be achieved through trained healthcare providers, in order to help individuals deal with the psychological, social, professional, ethical and legal implications of a genetic disease[30].

SPECIFIC BENEFITS OF GENETIC TESTING
Identification of undiagnosed family members

The principal advantage of the process of genetic testing is the detection of family members of the proband who have inherited the causal variant of the gene and are at-risk of developing the clinical phenotype of each cardiomyopathy. The importance of this procedure is huge, considering that life-threatening arrhythmias or SCD can be the first manifestation of a cardiomyopathy. Thereafter, only genotype-positive indivi-duals need periodic monitoring instead of all first-degree relatives[5]. The importance of familial screening is better established in HCM and ARVC, where the yield of genetic testing is higher, and gives the opportunity for better planning to assess and address the need for implantable cardioverter-defibrillator (ICD) devices in genotype-positive carriers.

Regarding SCD in young people, almost 30% of cases are caused by a non-diagnosed type of cardiomyopathy[31]. Genetic testing in post-mortem samples could enhance the likelihood of finding a disease-causing variant and performing subse-quent genetic screening of family members. Indeed, in a clinical screening in 198 cases of unexplained SCD in people below 35 years of age, five families suffering from inherited cardiomyopathies were detected; whereas, 27% of the cases had a definite or likely disease-causing mutation[32]. Therefore, according to European Society of Cardiology (ESC) guidelines, targeted post-mortem genetic analysis of potentially disease-causing genes should be considered for victims in whom a specific inheritable or cardiomyopathy is suspected[33].

Prognostic role

Several studies have proven that genotype status itself is associated with worse outcomes in HCM[34,35]. Sarcomere protein mutations in HCM have been found to be associated with increased rates of cardiovascular and SCD-related mortality[34]. In 628 HCM patients with a 12-year follow-up, positive genetic test result was identified as an independent prognostic factor for mortality in SCD and heart failure as well as all-cause mortality for the carrier, after adjustment for established risk factors[36]. In DCM, the approximately 5%-10% of probands with mutations in the LMNA gene are the best characterized, as LMNA variants manifest early-onset atrial arrhythmias and early development of conduction disturbances, coupled with a higher risk of SCD, often with only LV systolic impairment[37]. According to ESC guidelines, an ICD implantation should be considered in patients with DCM along with a confirmed disease-causing LMNA mutation and clinical risk factors, such as a non-sustained ventricular tachycardia, LV ejection fraction below 45% at first evaluation, male sex, and non-missense mutations[33].

Regarding ARVC, the prognostic role of genetic testing is not fully realized. Al-though probands with ARVC and known causal variants have been reported to develop ventricular arrhythmias approximately 4 years earlier than patients without a known mutation, no difference in mortality was detected in that study[38]. In another study of 105 probands with known ARVC-causal variants, the identification of a variant was found to be less important for predicting risk of life-threating arrhythmia or SCD than other factors, like sex and repolarization abnormalities in electrocardiogram or LV dysfunction[39]. Mutations in the PLN gene are also related to a poorer prognosis[40].

NGS analysis has allowed widespread identification of novel disease-causing genetic mutations[41]. Open availability of the ClinVar and ExAC databases in recent years has helped laboratories and clinicians to determine the possible pathogenicity of new detected variants. As a result, previously characterized genetic variants have been reclassified[42]. Genetic testing may identify predisposition for cardiomyopathies with unclear pathophysiological mechanisms; for example, risk of peripartum cardiomyopathy or anthracycline-induced cardiomyopathy may be identifiable by genetic testing. In patients with peripartum cardiomyopathy, in particular, and carrying mutations in the TTN gene, LV ejection fraction remains significantly lower compared to patients without this mutation[43]. Similarly, there is emerging evidence that a genetic predisposition may also be involved in anthracycline-induced cardiomyopathy[44,45]. Unfortunately, the current evidence is not sufficient to alter management decisions about the use of anthracycline but could lead to a personalized medicine approach in patients carrying specific mutations[46].

Prenatal genetic counselling

Prenatal genetic counselling is helpful in couples at risk of transmitting a genetic condition to their offspring. Through this process, a certified genetic counsellor explains the risk of transmission of disease, the impact of the disease on an affected child, as well as the benefits and limitations of all the available reproductive options. These options include in vitro fertilization with preimplantation genetic diagnosis, prenatal genetic screening, and postnatal genetic testing. The benefits and potential harms can be discussed for each of these options, such that the individual or couple can make a fully informed decision[12].

BARRIERS IN GENETIC TESTING

Despite the novelties in genetic techniques, there are still limitations in performing genetic testing for every diagnosed cardiomyopathy. Genetic testing seems to be of little to no value in low-yield cases with unclear inheritance pattern[36-38,47]; for this reason, the guidelines discourage genetic testing in isolated cases of idiopathic DCM where no evidence of inheritance exists[4,5]. In a study of 102 patients with idiopathic DCM, a disease-causing mutation was identified in only 10, whereas the clinical management was changed for only 1 patient, who received an ICD[47]. Unfortunately, even in disease where the yield of genetic testing is high, such as in HCM, when more possible genes were added, few additional causal variants were identified[48]. Another limitation in performing genetic testing when there is no clear clinical indication is the finding of a VUS. VUS may mislead clinicians, as it can neither confirm a genetic diagnosis nor exclude the need for familial surveillance[12]. The best practice in order to avoid an uncertain result is the performance of genetic testing after a thorough evaluation of the clinical case and a multidisciplinary management in pre-genetic counselling. Hence, any result from a genetic test can be further investigated at the research level[6,12].

CONCLUSION

At present, the mainstay of genetic testing in inherited cardiomyopathies is the identification of family members who have inherited the same causal mutation with the proband, which is important in terms of clinical decision-making. On the other hand, when the mutation is not detected in family members, anxiety and unnecessary clinical screening are avoided. This procedure is more helpful in cardiomyopathies where the yield of a positive genetic result is higher, such as HCM and ARVC. However, recent studies have provided more evidence on the genetic predisposition in specific cardiomyopathies and suggested a prognostic component when pathogenic variants are found.

Despite the progress in genetic techniques, there are currently well-documented limitations of cardiac genetic testing. Therefore, the role of family history should not be downgraded when the utility of genetic testing is questioned. A specialized multidisciplinary clinic incorporating cardiologists and genetic counsellors along with a move to a more precision-based approach seems to be the ideal model of manage-ment. Last but not least, focus should be given to teamwork of worldwide research groups in order to develop large databases that may elucidate the complexity of underlying genetics in inherited cardiomyopathies.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cardiac and cardiovascular systems

Country/Territory of origin: Greece

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Feng R S-Editor: Wang JJ L-Editor: A P-Editor: Wang JJ

References
1.  Arbustini E, Narula N, Dec GW, Reddy KS, Greenberg B, Kushwaha S, Marwick T, Pinney S, Bellazzi R, Favalli V, Kramer C, Roberts R, Zoghbi WA, Bonow R, Tavazzi L, Fuster V, Narula J. The MOGE(S) Classification for a Phenotype-Genotype Nomenclature of Cardiomyopathy: Endorsed by the World Heart Federation. Glob Heart. 2013;8:355-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
2.  Gersh BJ, Maron BJ, Bonow RO, Dearani JA, Fifer MA, Link MS, Naidu SS, Nishimura RA, Ommen SR, Rakowski H, Seidman CE, Towbin JA, Udelson JE, Yancy CW; American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. 2011 ACCF/AHA Guideline for the Diagnosis and Treatment of Hypertrophic Cardiomyopathy: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Developed in collaboration with the American Association for Thoracic Surgery, American Society of Echocardiography, American Society of Nuclear Cardiology, Heart Failure Society of America, Heart Rhythm Society, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. J Am Coll Cardiol. 2011;58:e212-e260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 815]  [Cited by in F6Publishing: 823]  [Article Influence: 63.3]  [Reference Citation Analysis (0)]
3.  Marcus FI, McKenna WJ, Sherrill D, Basso C, Bauce B, Bluemke DA, Calkins H, Corrado D, Cox MG, Daubert JP, Fontaine G, Gear K, Hauer R, Nava A, Picard MH, Protonotarios N, Saffitz JE, Sanborn DM, Steinberg JS, Tandri H, Thiene G, Towbin JA, Tsatsopoulou A, Wichter T, Zareba W. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the task force criteria. Circulation. 2010;121:1533-1541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1527]  [Cited by in F6Publishing: 1354]  [Article Influence: 96.7]  [Reference Citation Analysis (0)]
4.  Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJ, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WH, Tsai EJ, Wilkoff BL; American College of Cardiology Foundation;  American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;62:e147-e239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4116]  [Cited by in F6Publishing: 4509]  [Article Influence: 409.9]  [Reference Citation Analysis (0)]
5.  Ackerman MJ, Priori SG, Willems S, Berul C, Brugada R, Calkins H, Camm AJ, Ellinor PT, Gollob M, Hamilton R, Hershberger RE, Judge DP, Le Marec H, McKenna WJ, Schulze-Bahr E, Semsarian C, Towbin JA, Watkins H, Wilde A, Wolpert C, Zipes DP. 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). Heart Rhythm. 2011;8:1308-1339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 761]  [Cited by in F6Publishing: 729]  [Article Influence: 60.8]  [Reference Citation Analysis (0)]
6.  Ingles J, Semsarian C. Conveying a probabilistic genetic test result to families with an inherited heart disease. Heart Rhythm. 2014;11:1073-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
7.  Tennessen JA, Bigham AW, O'Connor TD, Fu W, Kenny EE, Gravel S, McGee S, Do R, Liu X, Jun G, Kang HM, Jordan D, Leal SM, Gabriel S, Rieder MJ, Abecasis G, Altshuler D, Nickerson DA, Boerwinkle E, Sunyaev S, Bustamante CD, Bamshad MJ, Akey JM; Broad GO;  Seattle GO;  NHLBI Exome Sequencing Project. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science. 2012;337:64-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1394]  [Cited by in F6Publishing: 1217]  [Article Influence: 101.4]  [Reference Citation Analysis (0)]
8.  Lee HH, Ching CK. Practical Aspects in Genetic Testing for Cardiomyopathies and Channelopathies. Clin Biochem Rev. 2019;40:187-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
9.  Tester DJ, Ackerman MJ. Genetic testing for potentially lethal, highly treatable inherited cardiomyopathies/channelopathies in clinical practice. Circulation. 2011;123:1021-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 136]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
10.  Lind JM, Chiu C, Semsarian C. Genetic basis of hypertrophic cardiomyopathy. Expert Rev Cardiovasc Ther. 2006;4:927-934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
11.  Thevenon J, Laurent G, Ader F, Laforêt P, Klug D, Duva Pentiah A, Gouya L, Maurage CA, Kacet S, Eicher JC, Albuisson J, Desnos M, Bieth E, Duboc D, Martin L, Réant P, Picard F, Bonithon-Kopp C, Gautier E, Binquet C, Thauvin-Robinet C, Faivre L, Bouvagnet P, Charron P, Richard P. High prevalence of arrhythmic and myocardial complications in patients with cardiac glycogenosis due to PRKAG2 mutations. Europace. 2017;19:651-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
12.  Ommen SR, Mital S, Burke MA, Day SM, Deswal A, Elliott P, Evanovich LL, Hung J, Joglar JA, Kantor P, Kimmelstiel C, Kittleson M, Link MS, Maron MS, Martinez MW, Miyake CY, Schaff HV, Semsarian C, Sorajja P. 2020 AHA/ACC Guideline for the Diagnosis and Treatment of Patients With Hypertrophic Cardiomyopathy: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2020;142:e558-e631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 81]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
13.  Akinrinade O, Ollila L, Vattulainen S, Tallila J, Gentile M, Salmenperä P, Koillinen H, Kaartinen M, Nieminen MS, Myllykangas S, Alastalo TP, Koskenvuo JW, Heliö T. Genetics and genotype-phenotype correlations in Finnish patients with dilated cardiomyopathy. Eur Heart J. 2015;36:2327-2337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 117]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
14.  Millat G, Bouvagnet P, Chevalier P, Sebbag L, Dulac A, Dauphin C, Jouk PS, Delrue MA, Thambo JB, Le Metayer P, Seronde MF, Faivre L, Eicher JC, Rousson R. Clinical and mutational spectrum in a cohort of 105 unrelated patients with dilated cardiomyopathy. Eur J Med Genet. 2011;54:e570-e575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
15.  Pérez-Serra A, Toro R, Campuzano O, Sarquella-Brugada G, Berne P, Iglesias A, Mangas A, Brugada J, Brugada R. A novel mutation in lamin a/c causing familial dilated cardiomyopathy associated with sudden cardiac death. J Card Fail. 2015;21:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Hershberger RE, Hedges DJ, Morales A. Dilated cardiomyopathy: the complexity of a diverse genetic architecture. Nat Rev Cardiol. 2013;10:531-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in F6Publishing: 647]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
17.  Herman DS, Lam L, Taylor MR, Wang L, Teekakirikul P, Christodoulou D, Conner L, DePalma SR, McDonough B, Sparks E, Teodorescu DL, Cirino AL, Banner NR, Pennell DJ, Graw S, Merlo M, Di Lenarda A, Sinagra G, Bos JM, Ackerman MJ, Mitchell RN, Murry CE, Lakdawala NK, Ho CY, Barton PJ, Cook SA, Mestroni L, Seidman JG, Seidman CE. Truncations of titin causing dilated cardiomyopathy. N Engl J Med. 2012;366:619-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1072]  [Cited by in F6Publishing: 960]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
18.  Mccaffrey T, Guglieri M, Murphy AP, Bushby K, Johnson A, Bourke JP. Cardiac involvement in female carriers of duchenne or becker muscular dystrophy. Muscle Nerve. 2017;55:810-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Towbin JA, McKenna WJ, Abrams DJ, Ackerman MJ, Calkins H, Darrieux FCC, Daubert JP, de Chillou C, DePasquale EC, Desai MY, Estes NAM 3rd, Hua W, Indik JH, Ingles J, James CA, John RM, Judge DP, Keegan R, Krahn AD, Link MS, Marcus FI, McLeod CJ, Mestroni L, Priori SG, Saffitz JE, Sanatani S, Shimizu W, van Tintelen JP, Wilde AAM, Zareba W. 2019 HRS expert consensus statement on evaluation, risk stratification, and management of arrhythmogenic cardiomyopathy. Heart Rhythm. 2019;16:e301-e372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 419]  [Article Influence: 83.8]  [Reference Citation Analysis (0)]
20.  van der Zwaag PA, Jongbloed JD, van den Berg MP, van der Smagt JJ, Jongbloed R, Bikker H, Hofstra RM, van Tintelen JP. A genetic variants database for arrhythmogenic right ventricular dysplasia/cardiomyopathy. Hum Mutat. 2009;30:1278-1283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 96]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
21.  Muchtar E, Blauwet LA, Gertz MA. Restrictive Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res. 2017;121:819-837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 155]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
22.  Gallego-Delgado M, Delgado JF, Brossa-Loidi V, Palomo J, Marzoa-Rivas R, Perez-Villa F, Salazar-Mendiguchía J, Ruiz-Cano MJ, Gonzalez-Lopez E, Padron-Barthe L, Bornstein B, Alonso-Pulpon L, Garcia-Pavia P. Idiopathic Restrictive Cardiomyopathy Is Primarily a Genetic Disease. J Am Coll Cardiol. 2016;67:3021-3023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
23.  Morales A, Cowan J, Dagua J, Hershberger RE. Family history: an essential tool for cardiovascular genetic medicine. Congest Heart Fail. 2008;14:37-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
24.  Bennett RL, Steinhaus KA, Uhrich SB, O'Sullivan CK, Resta RG, Lochner-Doyle D, Markel DS, Vincent V, Hamanishi J. Recommendations for standardized human pedigree nomenclature. Pedigree Standardization Task Force of the National Society of Genetic Counselors. Am J Hum Genet. 1995;56:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Hershberger RE, Lindenfeld J, Mestroni L, Seidman CE, Taylor MR, Towbin JA; Heart Failure Society of America. Genetic evaluation of cardiomyopathy--a Heart Failure Society of America practice guideline. J Card Fail. 2009;15:83-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 304]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
26.  Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding K, Rehm HL; ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19696]  [Cited by in F6Publishing: 18606]  [Article Influence: 2067.3]  [Reference Citation Analysis (0)]
27.  Golbus JR, Puckelwartz MJ, Fahrenbach JP, Dellefave-Castillo LM, Wolfgeher D, McNally EM. Population-based variation in cardiomyopathy genes. Circ Cardiovasc Genet. 2012;5:391-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 115]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
28.  Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, Gu B, Hart J, Hoffman D, Hoover J, Jang W, Katz K, Ovetsky M, Riley G, Sethi A, Tully R, Villamarin-Salomon R, Rubinstein W, Maglott DR. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 2016;44:D862-D868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1642]  [Cited by in F6Publishing: 1755]  [Article Influence: 195.0]  [Reference Citation Analysis (0)]
29.  Wilson KD, Shen P, Fung E, Karakikes I, Zhang A, InanlooRahatloo K, Odegaard J, Sallam K, Davis RW, Lui GK, Ashley EA, Scharfe C, Wu JC. A Rapid, High-Quality, Cost-Effective, Comprehensive and Expandable Targeted Next-Generation Sequencing Assay for Inherited Heart Diseases. Circ Res. 2015;117:603-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
30.  Caleshu C, Kasparian NA, Edwards KS, Yeates L, Semsarian C, Perez M, Ashley E, Turner CJ, Knowles JW, Ingles J. Interdisciplinary psychosocial care for families with inherited cardiovascular diseases. Trends Cardiovasc Med. 2016;26:647-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
31.  Robertus JL, Sheppard MN. 78 The Cry Sudden Cardiac Death Database.18 years of Referral for Expert Opinion. Heart. 2015;101:A42-A43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
32.  Bagnall RD, Weintraub RG, Ingles J, Duflou J, Yeates L, Lam L, Davis AM, Thompson T, Connell V, Wallace J, Naylor C, Crawford J, Love DR, Hallam L, White J, Lawrence C, Lynch M, Morgan N, James P, du Sart D, Puranik R, Langlois N, Vohra J, Winship I, Atherton J, McGaughran J, Skinner JR, Semsarian C. A Prospective Study of Sudden Cardiac Death among Children and Young Adults. N Engl J Med. 2016;374:2441-2452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 499]  [Cited by in F6Publishing: 527]  [Article Influence: 65.9]  [Reference Citation Analysis (0)]
33.  Priori SG, Blomström-Lundqvist C, Mazzanti A, Blom N, Borggrefe M, Camm J, Elliott PM, Fitzsimons D, Hatala R, Hindricks G, Kirchhof P, Kjeldsen K, Kuck KH, Hernandez-Madrid A, Nikolaou N, Norekvål TM, Spaulding C, Van Veldhuisen DJ; ESC Scientific Document Group. 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;36:2793-2867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2447]  [Cited by in F6Publishing: 2530]  [Article Influence: 281.1]  [Reference Citation Analysis (0)]
34.  Lopes LR, Syrris P, Guttmann OP, O'Mahony C, Tang HC, Dalageorgou C, Jenkins S, Hubank M, Monserrat L, McKenna WJ, Plagnol V, Elliott PM. Novel genotype-phenotype associations demonstrated by high-throughput sequencing in patients with hypertrophic cardiomyopathy. Heart. 2015;101:294-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 113]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
35.  Li Q, Gruner C, Chan RH, Care M, Siminovitch K, Williams L, Woo A, Rakowski H. Genotype-positive status in patients with hypertrophic cardiomyopathy is associated with higher rates of heart failure events. Circ Cardiovasc Genet. 2014;7:416-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
36.  van Velzen HG, Vriesendorp PA, Oldenburg RA, van Slegtenhorst MA, van der Velden J, Schinkel AFL, Michels M. Value of Genetic Testing for the Prediction of Long-Term Outcome in Patients With Hypertrophic Cardiomyopathy. Am J Cardiol. 2016;118:881-887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
37.  Brodt C, Siegfried JD, Hofmeyer M, Martel J, Rampersaud E, Li D, Morales A, Hershberger RE. Temporal relationship of conduction system disease and ventricular dysfunction in LMNA cardiomyopathy. J Card Fail. 2013;19:233-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
38.  Groeneweg JA, Bhonsale A, James CA, te Riele AS, Dooijes D, Tichnell C, Murray B, Wiesfeld AC, Sawant AC, Kassamali B, Atsma DE, Volders PG, de Groot NM, de Boer K, Zimmerman SL, Kamel IR, van der Heijden JF, Russell SD, Jan Cramer M, Tedford RJ, Doevendans PA, van Veen TA, Tandri H, Wilde AA, Judge DP, van Tintelen JP, Hauer RN, Calkins H. Clinical Presentation, Long-Term Follow-Up, and Outcomes of 1001 Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy Patients and Family Members. Circ Cardiovasc Genet. 2015;8:437-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 303]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
39.  Protonotarios A, Anastasakis A, Panagiotakos DB, Antoniades L, Syrris P, Vouliotis A, Stefanadis C, Tsatsopoulou A, McKenna WJ, Protonotarios N. Arrhythmic risk assessment in genotyped families with arrhythmogenic right ventricular cardiomyopathy. Europace. 2016;18:610-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
40.  Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, Falk V, González-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GMC, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P; ESC Scientific Document Group. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129-2200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8928]  [Cited by in F6Publishing: 8852]  [Article Influence: 1106.5]  [Reference Citation Analysis (3)]
41.  Cuenca S, Ruiz-Cano MJ, Gimeno-Blanes JR, Jurado A, Salas C, Gomez-Diaz I, Padron-Barthe L, Grillo JJ, Vilches C, Segovia J, Pascual-Figal D, Lara-Pezzi E, Monserrat L, Alonso-Pulpon L, Garcia-Pavia P; Inherited Cardiac Diseases Program of the Spanish Cardiovascular Research Network (Red Investigación Cardiovascular). Genetic basis of familial dilated cardiomyopathy patients undergoing heart transplantation. J Heart Lung Transplant. 2016;35:625-635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
42.  Walsh R, Thomson KL, Ware JS, Funke BH, Woodley J, McGuire KJ, Mazzarotto F, Blair E, Seller A, Taylor JC, Minikel EV;  Exome Aggregation Consortium; MacArthur DG, Farrall M, Cook SA, Watkins H. Reassessment of Mendelian gene pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference samples. Genet Med. 2017;19:192-203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 470]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
43.  Ware JS, Li J, Mazaika E, Yasso CM, DeSouza T, Cappola TP, Tsai EJ, Hilfiker-Kleiner D, Kamiya CA, Mazzarotto F, Cook SA, Halder I, Prasad SK, Pisarcik J, Hanley-Yanez K, Alharethi R, Damp J, Hsich E, Elkayam U, Sheppard R, Kealey A, Alexis J, Ramani G, Safirstein J, Boehmer J, Pauly DF, Wittstein IS, Thohan V, Zucker MJ, Liu P, Gorcsan J 3rd, McNamara DM, Seidman CE, Seidman JG, Arany Z; IMAC-2 and IPAC Investigators. Shared Genetic Predisposition in Peripartum and Dilated Cardiomyopathies. N Engl J Med. 2016;374:233-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 368]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
44.  Aminkeng F, Ross CJ, Rassekh SR, Hwang S, Rieder MJ, Bhavsar AP, Smith A, Sanatani S, Gelmon KA, Bernstein D, Hayden MR, Amstutz U, Carleton BC; CPNDS Clinical Practice Recommendations Group. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol. 2016;82:683-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 156]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
45.  Aminkeng F, Bhavsar AP, Visscher H, Rassekh SR, Li Y, Lee JW, Brunham LR, Caron HN, van Dalen EC, Kremer LC, van der Pal HJ, Amstutz U, Rieder MJ, Bernstein D, Carleton BC, Hayden MR, Ross CJ; Canadian Pharmacogenomics Network for Drug Safety Consortium. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet. 2015;47:1079-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 182]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
46.  Reichwagen A, Ziepert M, Kreuz M, Gödtel-Armbrust U, Rixecker T, Poeschel V, Reza Toliat M, Nürnberg P, Tzvetkov M, Deng S, Trümper L, Hasenfuss G, Pfreundschuh M, Wojnowski L. Association of NADPH oxidase polymorphisms with anthracycline-induced cardiotoxicity in the RICOVER-60 trial of patients with aggressive CD20(+) B-cell lymphoma. Pharmacogenomics. 2015;16:361-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 42]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
47.  Broch K, Andreassen AK, Hopp E, Leren TP, Scott H, Müller F, Aakhus S, Gullestad L. Results of comprehensive diagnostic work-up in 'idiopathic' dilated cardiomyopathy. Open Heart. 2015;2:e000271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
48.  Alfares AA, Kelly MA, McDermott G, Funke BH, Lebo MS, Baxter SB, Shen J, McLaughlin HM, Clark EH, Babb LJ, Cox SW, DePalma SR, Ho CY, Seidman JG, Seidman CE, Rehm HL. Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: expanded panels offer limited additional sensitivity. Genet Med. 2015;17:880-888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 296]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]