Next Article in Journal
Survival Outcomes in Squamous Cell Carcinoma of the External Auditory Canal: A Systematic Review and Meta-Analysis
Next Article in Special Issue
Real-World Use and Predictors of Response to Disopyramide in Patients with Obstructive Hypertrophic Cardiomyopathy
Previous Article in Journal
Opioids in the Treatment of Chronic Idiopathic Diarrhea in Humans—A Systematic Review and Treatment Guideline
Previous Article in Special Issue
Association of Types of Sleep Apnea and Nocturnal Hypoxemia with Atrial Fibrillation in Patients with Hypertrophic Cardiomyopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Genetic Testing and Counselling in Hypertrophic Cardiomyopathy: Frequently Asked Questions

1
Pediatric Cardiology Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
2
Doctoral School of Clinical Medicine, University of Szeged, 6720 Szeged, Hungary
3
Cardiomyopathy Unit, Careggi University Hospital, 50134 Florence, Italy
4
Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, 20100 Milan, Italy
5
Department of Medicine and Surgery, University Milano Bicocca, 20126 Milan, Italy
6
Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(7), 2489; https://doi.org/10.3390/jcm12072489
Submission received: 12 February 2023 / Revised: 13 March 2023 / Accepted: 24 March 2023 / Published: 24 March 2023

Abstract

:
Genetic counselling and genetic testing in hypertrophic cardiomyopathy (HCM) represent an integral part of the diagnostic algorithm to confirm the diagnosis, distinguish it from phenocopies, and suggest tailored therapeutic intervention strategies. Additionally, they enable cascade genetic testing in the family. With the implementation of Next Generation Sequencing technologies (NGS), the interpretation of genetic data has become more complex. In this regard, cardiologists play a central role, aiding geneticists to correctly evaluate the pathogenicity of the identified genetic alterations. In the ideal setting, geneticists and cardiologists must work side by side to diagnose HCM as well as convey the correct information to patients in response to their many questions and concerns. After a brief overview of the role of genetics in the diagnosis of HCM, we present and discuss the frequently asked questions by HCM patients throughout our 20-year genetic counselling experience. Appropriate communication between the team and the families is key to the goal of delivering the full potential of genetic testing to our patients.

1. Introduction

Hypertrophic Cardiomyopathy (HCM) is a genetic heart condition characterized by the presence of asymmetric hypertrophy of the left ventricle (LVH) that is not solely explained by abnormal loading conditions [1,2,3,4,5]. However, in the latest AHA/ACC HCM guidelines, the term is applied only to patients that have a sarcomeric gene variant or a negative gene test in the absence of another cardiac, systemic, or metabolic disease [5]. In the 1990s, an MYH7 (β myosin heavy chain) variant was identified in a large Canadian family with four generations of HCM-affected subjects [6,7]. Over the past 30 years, the complexity of the genetic basis of HCM has been increasingly elucidated. Today HCM is considered the most common genetic cardiac disease, transmitted in an autosomal dominant manner. HCM has a prevalence of roughly 1:500 in adults, thus affecting an estimated 1 million individuals in Europe [8]. Mutation penetrance varies widely and may be significantly delayed [9]. HCM is known to be associated with over ~1500 mutations, described in about 40 genes, especially in genes encoding for cardiac sarcomeric proteins. HCM has been designed as the “disease of the sarcomere” [10]. MYH7 and MYBPC3 (cardiac myosin-binding protein C) are the two most commonly mutated sarcomeric genes, and they account for about 40% of diagnoses. In addition, MYL2 (regulatory myosin light chain), MYL3 (essential myosin light chain), TPM1 (α tropomyosin), TNNT2 (cardiac troponin T), TNNI3 (cardiac troponin I), and ACTC1 (cardiac actin) represent the “core disease” genes which are considered essential for establishing a reliable and accurate molecular diagnosis. Rare mutations have been found in additional sarcomere genes or genes coding for proteins from the nearby Z-disc, such as MYH6 (α myosin heavy chain), CSRP3 (muscle LIM protein), TCAP (telethonin), and genes implicated in calcium homeostasis pathways, such as VCL (vinculin) and JPH2 (junctophilin 2) [11,12,13,14]. A small number of hypertrophic patients exhibit Fabry syndrome (GLA gene), Danon disease (LAMP2 gene), cardiac amyloidosis (TTR gene), Wolff—Parkinson—White syndrome (PRKAG2 gene), and mitochondrial cardiomyopathies [15,16,17,18]. These phenocopies are different disease entities with distinct heredity, pathogenesis, natural history, extra-cardiac characteristics, and therapy. Moreover, HCM phenotype may be associated with malformation syndromes, mainly RASophaties, a heterogeneous group of diseases caused by mutations in genes of RAS-MAPK cascade, including PTPN11, BRAF, RAF1, SOS1, HRAS, and KRAS [14]. For these diseases, the genetic test, in conjunction with a detailed diagnostic examination of the patient and their family, is a critical tool for the diagnosis and proper treatment [19].
Currently, with Next Generation Sequencing (NGS) technologies, it is possible to identify a responsible mutation in 30–60% of HCM patients. However, it is true that the yield of positive genetic testing is higher in the case of positive family history of HCM and is lower in sporadic cases [5,20]. The genetic cause remains unknown and may be polygenic in the rest of the cases, together with some environmental factors, such as obesity, arterial hypertension, and intense sports activity [14,20]. About 5–10% of patients have been reported to have complex genotypes carrying multiple (two or more rarely three) mutations in sarcomere genes; this condition is usually associated with a poorer prognosis [21]. Since 2005, NGS technologies have enabled a breakthrough rise in high-throughput sequencing capabilities by analyzing multigene panels, whole-exome (WES), and whole-genome sequencing (WGS). The latter is mostly aimed at discovering novel disease-causing genes and is mostly used for research purposes. In selected cases, for example, in children with HCM, the WES analysis in a trio (together with the parent’s DNA sequencing) should be performed [22]. The NGS strategy is to sequence millions of short-DNA fragments in massively parallel arrays, then realign and map the short reads back to the reference genome [23]. The accurate interpretation of data sequencing is the weakness of the NGS strategy. It may be very difficult to predict if a DNA variant identified in a patient is truly disease-causing (pathogenic or likely pathogenic), a benign polymorphism (negative test) or a rare variant with unclear clinical significance and not predicted to cause HCM (variant of uncertain significance, VUS) [24]. In this last case, the genetic test’s result is not clinically actionable and may be considered one limitation of the genetic study. Private and public cardiogenetic laboratories often use commercial panels for genetic tests. The panels are typically designed to contain genes having a definitive or moderate association with the disorders; the panels must be assessed on a regular basis and may potentially be expanded if new genes are identified [10,20,25].

2. Genetic Counselling

Genetic counselling represents an essential part of genetic testing and is recommended for all patients with HCM who choose to undergo genetic testing [5]. The main objective of counselling is informing patients and families about the genetic aspects of their disease, the advantages and the limitations of the test and the possibility of transmitting the disease to their relatives [26,27,28,29]. It is also critical to help ensure that patients and families have realistic expectations about the benefits of genetic testing. Counselling can be divided into pre-test and post-test counselling (Figure 1). According to the latest guidelines on HCM, pre-and post-test genetic counselling should be performed by a trained genetic counsellor or by other experts in the genetics of cardiovascular diseases (Class 1, level of evidence B) [5]. Due to technological advances and new recommendations, the number of genetic testing requests is increasing rapidly, which may expose cardiologists to more often facing similar questions about the implication of genetic testing results. Therefore, in this article, we collected and discussed the frequently asked questions by our HCM patients to help cardiologists manage genetic testing-related scenarios more easily and confidently.
Pre-test counselling. During pre-test counselling, the patient should be informed about the specific type of test that will be performed on the DNA sample (for example, which are the genes studied and by which technologies), the possible test results, the potential clinical benefit, and the test turn-around time. Pre-test counselling is of paramount importance to identify other clinically affected family members, patterns of disease transmission, consanguinity within the family, and history of sudden cardiac death in a relative. Moreover, it helps to assist decision-making by fully informing patients [5,23]. A three generations pedigree of the family, written informed consent and a blood sample are collected during this interview.
Post-test counselling. Test results should be communicated during post-test genetic counselling. Post-test genetic counselling should involve skilled healthcare professionals working in multidisciplinary teams to assist patients in understanding and handling the psychological, social, professional, ethical, and legal consequences of test results. During post-test genetic counselling, the patient becomes aware of the impact of test results on himself/herself and his/her relatives. Importantly, the patient is informed about the clinical implications and the possibility of performing cascade testing screening on other family members (predictive genetic testing in relatives). In young patients, the risk of transmission when planning future pregnancies should be discussed [5].

3. Patients’ Questions, Real-World Answers

Interpreting the clinical significance of genetic variants is a complex process in which the role of clinicians is key by providing evidence that supports the variants’ classification, both in the proband and in family members [23]. Cardiologists need to be able to answer questions from patients with genetic heart diseases, collaborating closely with the geneticist. In this article, we present and discuss the frequently asked questions by HCM patients over our 20-year genetic counselling experience.
Why is it useful to undergo a genetic test for me? Genetic testing in HCM adult patients remains critical for the identification of at-risk asymptomatic relatives. In addition, it provides a definitive molecular diagnosis and allows the identification of phenocopies (for example Anderson-Fabry disease, TTR amyloidosis, Danon disease, or PRKAG2 cardiac glycogenosis). Conversely, the impact on risk stratification and the prognosis is limited as genotype status alone cannot predict patient-specific outcomes. However, it is important to note that patients with pathogenic or likely pathogenic sarcomere mutations had a greater risk for adverse outcomes compared with patients without mutations [5,10,11,21].
Will the identified DNA mutation predict the course and outcome of my disease? Because HCM is a genetic disease with incomplete penetrance and variable expressivity, the possibility of associating mutations with a specific disease course is limited. Some genes or specific mutations have been linked to a high rate of sudden mortality and adverse remodeling (e.g., TNNT2 mutations; MYH7 p.Arg403Gln), a low rate of LVH (TNNT2), or delayed cardiac expression (MYBPC3). Additionally, patients with complex genotypes are more likely to have severe phenotypes or early disease manifestations [21]. Yet, no clear and consistent associations have been discovered for the majority of mutations to date [20,22,23,30,31,32].
Is it possible that I will never develop the disease, even if I have inherited the familial mutation? HCM is known to be a disease with incomplete penetrance and an intra-family variability of phenotype expression. Hence, the identification of a sarcomeric mutation (genotype positive) in an otherwise healthy individual (phenotype negative) does not necessarily mean that the subject will develop the disease but does indicate an increased risk of developing it. Although HCM may rarely be present in neonates and children, usually, the mean age of onset in HCM ranges from the third to the fifth decade of life. However, it has been reported that in a proportion of cases, it may remain latent or be identified even after age 60 [33].
If my genetic mutation gets identified, will I have targeted therapies? Although experimental studies hold hope for the future, no etiological/preventive therapies are available today. Nonetheless, the identification of a pathogenic mutation may help the cardiologist make tailored therapeutic choices and distinguish rare mimics such as Fabry disease, Danon disease, amyloidosis caused by TTR mutation, or atypical glycogenosis due to mutations in the PRKAG2 gene. Emerging, targeted therapies, including gene therapy for HCM patients, are now opening new perspectives [24,34,35,36,37,38].
In the case of an uncertain clinical diagnosis, can the identification of a genetic mutation definitively help to clarify it? In the case of inconclusive clinical findings, the identification of a pathogenic/likely pathogenic HCM rare variant can confirm the diagnosis [39].
My echocardiography is normal, but my genetic test is positive. Should I stop the physical activity? In general, for most patients with HCM, mild- to moderate-intensity recreational exercise is beneficial and is recommended to improve cardiorespiratory fitness, quality of life, and overall health. For phenotype-negative individuals, due to their low risk of sudden cardiac death (SCD), there is no restriction from competitive sports unless the family history indicates a high risk for SCD. However, serial echocardiography is recommended to assess for any phenotype development at periodic intervals depending on age [1,5,40].
Can a positive genetic test influence my insurance policies? Genetic data are strictly personal and protected by different regulations from country to country. In the USA, The Genetic Information Non-discrimination Act (GINA) protects individuals against discrimination based on their genetic information in health coverage and employment. Italy also has these types of laws (Authorization No. 8/2016—Garante Privacy). For this reason, it is not required to declare to have carried out the genetic test, either in the working environment or during the stipulation of life insurance [41].
How likely is it that my children will inherit the mutation? Sarcomeric HCM is inherited in an autosomal dominant manner, which means that both female and male children have a 50% probability of carrying the disease-causing variant, although variable penetrance can result in differences in the onset and severity of clinical manifestations [5].
I know genetic test needs a long time to be performed. However, once a mutation has been identified, is it going to take that much time to test my family? Once the mutation is identified in the first patient of the family, we can easily look for it in the other family members. We already know which “word” is misspelled and on which “page” it is located. Therefore, the predictive test takes only a few days [42].
Since I have HCM and a causative mutation has been found, is it possible to perform the genetic test on my five-year-old son/daughter, even if he/she does not show any sign of the disease? Performing a genetic test on an unaffected child is generally not recommended [43]. The fundamental justification behind this is the lack of knowledge on the severity, age of onset, and phenotypic manifestations associated with HCM, as well as the inability to stop its progression. For these reasons, the medical benefit of a pre-symptomatic diagnosis of HCM is questionable, and since the request is made by the parents, the child is often not mature enough to understand the consequences of the test and make any decision. In our opinion, it is of utmost importance to balance benefits and harms, case by case, and keep in mind that he/she should be free to choose by himself to know whether he/she has inherited the mutation. Therefore, in the case of an unaffected child, a genetic test should be carried out after 10–12 years of age. However, in certain situations, such as a possible competitive sports career or a family history of malignant early-onset disease, it may be performed earlier [22,44].
After the identification of the mutation in my husband, is it possible to perform a prenatal test on our child? The decision to pursue a prenatal diagnosis is a personal one. Through invasive prenatal diagnosis (by amniocentesis or chorionic villus sample), it is possible to obtain the fetus’s DNA and proceed with searching for the known mutation [5,23,26]. Nonetheless, in our opinion, it is not appropriate to carry out this type of test due to the above-mentioned reasons: the uncertain age of onset of symptoms, the severity of symptoms, the risk of complications, etc. Clinicians cannot precisely anticipate the clinical course of an individual with a mutation since extremely different manifestations of HCM can occur in family members carrying the same mutation. Both incomplete penetrance and variable expressivity are probably due to a combination of genetic, environmental, and lifestyle factors. In our experience, none of the couples persisted to request prenatal testing after the first steps of the multidisciplinary shared decision-making process.
If the mutation is not identified, does it mean that my cardiomyopathy does not have a genetic base? The available genetic test in the cardiogenetic laboratories involves the analysis of a panel of genes known to be associated with HCM. The probability of identifying a disease-causing mutation in these genes is about 40–60% (test sensitivity). It is believed that both familial and isolated HCM forms are genetically determined. Therefore, even if no causative variant gets identified, a genetic basis cannot be excluded [5]. In this scenario, the causative variant could lie in modulator or unknown genes or in DNA regions that we are not able to analyze yet. For these reasons, both the HCM patients with negative genetic tests and their relatives should undergo regular cardiological follow-ups [5].
What does “Inconclusive genetic test result” mean? It means that a rare variant was identified in the patient’s DNA, called a variant of uncertain significance (VUS). It is unclear whether this variant is associated with the health condition or not. Due to their rarity in the population, only a limited amount of information is available on VUS. Thus, when a VUS gets identified, the test result is ambiguous and inconclusive at a clinical level. For this reason, a VUS should not be used to establish a genetic diagnosis and guide cascade family screening. Future knowledge or scientific reports may ameliorate our understanding of the VUS and help to re-evaluate them. The identification of a VUS, even if it represents one of the three possible results of a genetic test (1, positive: pathogenic/likely pathogenic causative variant; 2, rare VUS; 3, no identified rare variant) can be considered a limitation of genetic testing [24,45,46].
In Table 1, we have collected some pieces of information for cardiologists who deem it appropriate to have the patient undergo genetic testing. We consider these recommendations necessary to avoid setting unrealistic expectations and to ensure that it is consistent with the information provided during genetic counselling.

4. Conclusions

For cardiologists to achieve confidence in ordering genetic testing for HCM and counselling, they must be able to answer the major questions of their patients. Continuing close collaboration with geneticists is the key to success.

Funding

This research received no external funding. E.D.P. was a recipient of the Erasmus+ grant for medical and doctorate students in Florence from September to December 2021 and January to December 2022.

Institutional Review Board Statement

The study was conducted following the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We are thankful to all patients.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Elliott, P.M.; Anastasakis, A.; Borger, M.A.; Borggrefe, M.; Cecchi, F.; Charron, P.; Hagege, A.A.; Lafont, A.; Limongelli, G.; Mahrholdt, H.; et al. 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy: The Task Force for the Diagnosis and Management of Hypertrophic Cardiomyopathy of the European Society of Cardiology (ESC). Eur. Heart J. 2014, 35, 2733–2779. [Google Scholar]
  2. Maron, B.J.; Desai, M.Y.; Nishimura, R.A.; Spirito, P.; Rakowski, H.; Towbin, J.A.; Dearani, J.A.; Rowin, E.J.; Maron, M.S.; Sherrid, M.V. Management of Hypertrophic Cardiomyopathy: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2022, 79, 390–414. [Google Scholar] [CrossRef] [PubMed]
  3. Maron, B.J.; Maron, M.S. Hypertrophic cardiomyopathy. Lancet 2013, 381, 242–255. [Google Scholar] [CrossRef]
  4. Braunwald, E.; Lambrew, C.T.; Rockoff, S.D.; Ross, J., Jr.; Morrow, A.G. Idiopathic Hypertrophic Subaortic Stenosis. I. A Description of the Disease Based Upon an Analysis of 64 Patients. Circulation 1964, 30 (Suppl. S4), 3–119. [Google Scholar] [CrossRef]
  5. Ommen, S.R.; Mital, S.; Burke, M.A.; Day, S.M.; Deswal, A.; Elliott, P.; Evanovich, L.L.; Hung, J.; Joglar, J.A.; Kantor, P.; et al. 2020 AHA/ACC Guideline for the Diagnosis and Treatment of Patients with Hypertrophic Cardiomyopathy: Executive Summary: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2020, 76, 3022–3055. [Google Scholar] [CrossRef]
  6. Jarcho, J.A.; McKenna, W.; Pare, J.A.; Solomon, S.D.; Holcombe, R.F.; Dickie, S.; Levi, T.; Donis-Keller, H.; Seidman, J.G.; Seidman, C.E. Mapping a gene for familial hypertrophic cardiomyopathy to chromosome 14q1. N. Engl. J. Med. 1989, 321, 1372–1378. [Google Scholar] [CrossRef] [Green Version]
  7. Seidman, C.E.; Seidman, J.G. Identifying sarcomere gene mutations in hypertrophic cardiomyopathy: A personal history. Circ. Res. 2011, 108, 743–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Maron, B.J.; Maron, M.S.; Semsarian, C. Genetics of hypertrophic cardiomyopathy after 20 years: Clinical perspectives. JACC 2012, 60, 705–715. [Google Scholar] [CrossRef] [Green Version]
  9. Semsarian, C.; Semsarian, C.R. Variable Penetrance in Hypertrophic Cardiomyopathy: In Search of the Holy Grail. JACC 2020, 76, 560–562. [Google Scholar] [CrossRef]
  10. Ho, C.Y.; Charron, P.; Richard, P.; Girolami, F.; Van Spaendonck-Zwarts, K.Y.; Pinto, Y. Genetic advances in sarcomeric cardiomyopathies: State of the art. Cardiovasc. Res. 2015, 105, 397–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Olivotto, I.; Girolami, F.; Ackerman, M.J.; Nistri, S.; Bos, J.M.; Zachara, E.; Ommen, S.R.; Theis, J.L.; Vaubel, R.A.; Re, F.; et al. Myofilament protein gene mutation screening and outcome of patients with hypertrophic cardiomyopathy. Mayo. Clin. Proc. 2008, 83, 630–638. [Google Scholar] [CrossRef] [PubMed]
  12. Ingles, J.; Goldstein, J.; Thaxton, C.; Caleshu, C.; Corty, E.W.; Crowley, S.B.; Dougherty, K.; Harrison, S.M.; McGlaughon, J.; Milko, L.V.; et al. Evaluating the Clinical Validity of Hypertrophic Cardiomyopathy Genes. Circ. Genom. Precis. Med. 2019, 12, e002460. [Google Scholar] [CrossRef] [Green Version]
  13. Walsh, R.; Thomson, K.L.; Ware, J.S.; Funke, B.H.; Woodley, J.; McGuire, K.J.; Mazzarotto, F.; Blair, E.; Seller, A.; Taylor, J.C.; et al. Reassessment of Mendelian gene pathogenicity using 7855 cardiomyopathy cases and 60,706 reference samples. Genet. Med. 2017, 19, 192–203. [Google Scholar] [CrossRef] [Green Version]
  14. Hershberger, R.E.; Givertz, M.M.; Ho, C.Y.; Judge, D.P.; Kantor, P.F.; McBride, K.L.; Morales, A.; Taylor, M.R.G.; Vatta, M.; Ware, S.M.; et al. Genetic evaluation of cardiomyopathy: A clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet. Med. 2018, 20, 899–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Maron, B.J.; Roberts, W.C.; Arad, M.; Haas, T.S.; Spirito, P.; Wright, G.B.; Almquist, A.K.; Baffa, J.M.; Saul, J.P.; Ho, C.Y.; et al. Clinical outcome and phenotypic expression in LAMP2 cardiomyopathy. JAMA 2009, 301, 1253–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Banankhah, P.; Fishbein, G.A.; Dota, A.; Ardehali, R. Cardiac manifestations of PRKAG2 mutation. BMC Med. Genet. 2018, 19, 1. [Google Scholar] [CrossRef] [Green Version]
  17. Arends, M.; Wijburg, F.A.; Wanner, C.; Vaz, F.M.; van Kuilenburg, A.B.P.; Hughes, D.A.; Biegstraaten, M.; Mehta, A.; Hollak, C.E.M.; Langeveld, M. Favourable effect of early versus late start of enzyme replacement therapy on plasma globotriaosylsphingosine levels in men with classical Fabry disease. Mol. Genet. Metab. 2017, 121, 157–161. [Google Scholar] [CrossRef]
  18. Gagliardi, C.; Perfetto, F.; Lorenzini, M.; Ferlini, A.; Salvi, F.; Milandri, A.; Quarta, C.C.; Taborchi, G.; Bartolini, S.; Frusconi, S.; et al. Phenotypic profile of Ile68Leu transthyretin amyloidosis: An underdiagnosed cause of heart failure. Eur. J. Heart Fail. 2018, 20, 1417–1425. [Google Scholar] [CrossRef] [Green Version]
  19. Girolami, F.; Vergaro, G.; Pieroni, M.; Passantino, S.; Giannotti, G.; Grippo, G.; Canale, M.L.; Favilli, S.; Cappelli, F.; Olivotto, I.; et al. Clinical pathway for cardiomyopathies: A genetic testing strategy proposed by ANMCO in Tuscany. G. Ital. Cardiol. 2020, 21, 926–934. [Google Scholar]
  20. Mazzarotto, F.; Girolami, F.; Boschi, B.; Barlocco, F.; Tomberli, A.; Baldini, K.; Coppini, R.; Tanini, I.; Bardi, S.; Contini, E.; et al. Defining the diagnostic effectiveness of genes for inclusion in panels: The experience of two decades of genetic testing for hypertrophic cardiomyopathy at a single center. Genet. Med. 2019, 21, 284–292. [Google Scholar] [CrossRef] [PubMed]
  21. Ho, C.Y.; Day, S.M.; Ashley, E.A.; Michels, M.; Pereira, A.C.; Jacoby, D.; Cirino, A.L.; Fox, J.C.; Lakdawala, N.K.; Ware, J.S.; et al. Genotype and Lifetime Burden of Disease in Hypertrophic Cardiomyopathy: Insights from the Sarcomeric Human Cardiomyopathy Registry (SHaRe). Circulation 2018, 138, 1387–1398. [Google Scholar] [CrossRef]
  22. Girolami, F.; Iascone, M.; Pezzoli, L.; Passantino, S.; Limongelli, G.; Monda, E.; Rubino, M.; Adorisio, R.; Lombardi, M.; Ragni, L.; et al. Clinical pathway on pediatric cardiomyopathies: A genetic testing strategy proposed by the Italian Society of Pediatric Cardiology. G. Ital. Cardiol. 2022, 23, 505–515. [Google Scholar]
  23. Girolami, F.; Frisso, G.; Benelli, M.; Crotti, L.; Iascone, M.; Mango, R.; Mazzaccara, C.; Pilichou, K.; Arbustini, E.; Tomberli, B.; et al. Contemporary genetic testing in inherited cardiac disease: Tools, ethical issues, and clinical applications. J. Cardiovasc. Med. 2018, 19, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Arbustini, E.; Behr, E.R.; Carrier, L.; van Duijn, C.; Evans, P.; Favalli, V.; van der Harst, P.; Haugaa, K.H.; Jondeau, G.; Kääb, S.; et al. Interpretation and actionability of genetic variants in cardiomyopathies: A position statement from the European Society of Cardiology Council on cardiovascular genomics. Eur. Heart J. 2022, 43, 1901–1916. [Google Scholar] [CrossRef] [PubMed]
  25. Alfares, A.A.; Kelly, M.A.; McDermott, G.; Funke, B.H.; Lebo, M.S.; Baxter, S.B.; Shen, J.; McLaughlin, H.M.; Clark, E.H.; Babb, L.J.; et al. Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: Expanded panels offer limited additional sensitivity. Genet. Med. 2015, 17, 880–888. [Google Scholar] [CrossRef] [Green Version]
  26. Charron, P.; Héron, D.; Gargiulo, M.; Richard, P.; Dubourg, O.; Desnos, M.; Bouhour, J.B.; Feingold, J.; Carrier, L.; Hainque, B.; et al. Genetic testing and genetic counselling in hypertrophic cardiomyopathy: The French experience. J. Med. Genet. 2002, 39, 741–746. [Google Scholar] [CrossRef] [Green Version]
  27. Cirino, A.L.; Seidman, C.E.; Ho, C.Y. Genetic Testing and Counselling for Hypertrophic Cardiomyopathy. Cardiol. Clin. 2019, 37, 35–43. [Google Scholar] [CrossRef]
  28. Skrzynia, C.; Demo, E.M.; Baxter, S.M. Genetic counselling and testing for hypertrophic cardiomyopathy: An adult perspective. J. Cardiovasc. Transl. Res. 2009, 2, 493–499. [Google Scholar] [CrossRef]
  29. Cirino, A.L.; Harris, S.L.; Murad, A.M.; Hansen, B.; Malinowski, J.; Natoli, J.L.; Kelly, M.A.; Christian, S. The uptake and utility of genetic testing and genetic counselling for hypertrophic cardiomyopathy—A systematic review and meta-analysis. J. Genet. Couns. 2022, 31, 1290–1305. [Google Scholar] [CrossRef]
  30. Chen, Y.; Xu, F.; Munkhsaikhan, U.; Boyle, C.; Borcky, T.; Zhao, W.; Purevjav, E.; Towbin, J.A.; Liao, F.; Williams, R.W.; et al. Identifying modifier genes for hypertrophic cardiomyopathy. J. Mol. Cell. Cardiol. 2020, 144, 119–126. [Google Scholar] [CrossRef]
  31. Torricelli, F.; Girolami, F.; Olivotto, I.; Passerini, I.; Frusconi, S.; Vargiu, D.; Richard, P.; Cecchi, F. Prevalence and clinical profile of troponin T mutations among patients with hypertrophic cardiomyopathy in Tuscany. Am. J. Cardiol. 2003, 92, 1358–1362. [Google Scholar] [CrossRef]
  32. Girolami, F.; Ho, C.Y.; Semsarian, C.; Baldi, M.; Will, M.L.; Baldini, K.; Torricelli, F.; Yeates, L.; Cecchi, F.; Ackerman, M.J.; et al. Clinical features and outcome of hypertrophic cardiomyopathy associated with triple sarcomere protein gene mutations. J. Am. Coll. Cardiol. 2010, 55, 1444–1453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Maurizi, N.; Michels, M.; Rowin, E.J.; Semsarian, C.; Girolami, F.; Tomberli, B.; Cecchi, F.; Maron, M.S.; Olivotto, I.; Maron, B.J. Clinical Course and Significance of Hypertrophic Cardiomyopathy Without Left Ventricular Hypertrophy. Circulation 2019, 139, 830–833. [Google Scholar] [CrossRef] [PubMed]
  34. Iavarone, M.; Monda, E.; Vritz, O.; Albert, D.C.; Rubino, M.; Verrillo, F.; Caiazza, M.; Lioncino, M.; Amodio, F.; Guarnaccia, N.; et al. Medical treatment of patients with hypertrophic cardiomyopathy: An overview of current and emerging therapy. Arch. Cardiovasc. Dis. 2022, 115, 529–537. [Google Scholar] [CrossRef] [PubMed]
  35. Helms, A.S.; Thompson, A.D.; Day, S.M. Translation of New and Emerging Therapies for Genetic Cardiomyopathies. JACC 2021, 7, 70–83. [Google Scholar] [CrossRef]
  36. Investor Portal. Tenaya Therapeutics Receives Orphan Drug Designation and Presents Pre-Clinical Data for Its Most Advanced Gene Therapy Product Candidate for Genetic Hypertrophic Cardiomyopathy. Available online: https://investors.tenayatherapeutics.com/news-releases/news-release-details/tenaya-therapeutics-receives-orphan-drug-designation-and (accessed on 11 February 2023).
  37. Globe Newswire. LEXEO Therapeutics Expands Cardiac Gene Therapy Pipeline with Acquisition of Stelios Therapeutics and its Gene Therapy Programs for Rare Cardiovascular Diseases. Available online: https://www.globenewswire.com/news-release/2021/07/21/2266478/0/en/LEXEO-Therapeutics-Expands-Cardiac-Gene-Therapy-Pipeline-with-Acquisition-of-Stelios-Therapeutics-and-its-Gene-Therapy-Programs-for-Rare-Cardiovascular-Diseases.html (accessed on 11 February 2023).
  38. Olivotto, I.; Oreziak, A.; Barriales-Villa, R.; Abraham, T.P.; Masri, A.; Garcia-Pavia, P.; Saberi, S.; Lakdawala, N.K.; Wheeler, M.T.; Owens, A.; et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 396, 759–769. [Google Scholar] [CrossRef]
  39. Baldi, M.; Girolami, F. A new type of “tracing” for cardiologists? G. Ital. Cardiol. 2009, 10, 266. [Google Scholar]
  40. Semsarian, C.; Gray, B.; Haugaa, K.H.; Lampert, R.; Sharma, S.; Kovacic, J.C. Athletic Activity for Patients with Hypertrophic Cardiomyopathy and Other Inherited Cardiovascular Diseases: JACC Focus Seminar ¾. JACC 2022, 80, 1268–1283. [Google Scholar] [CrossRef]
  41. GPDP. Autorizzazione Generale al Trattamento Dei Dati Genetici. Autorizzazione n. 8/2016, 15 Dicembre 2016. Available online: https://www.garanteprivacy.it/home/docweb/-/docweb-display/docweb/5803688 (accessed on 11 February 2023).
  42. Kassem, H.S.; Girolami, F.; Sanoudou, D. Molecular genetics made simple. Glob. Cardiol. Sci. Pract. 2012, 2012, 6. [Google Scholar] [CrossRef]
  43. Semsarian, C.; Ho, C.Y. Screening children at risk for hypertrophic cardiomyopathy: Balancing benefits and harms. Eur. Heart J. 2019, 40, 3682–3684. [Google Scholar] [CrossRef]
  44. Mital, S.; Ommen, S. To Screen or Not to Screen, That Is the Question. Circulation 2019, 140, 193–195. [Google Scholar] [CrossRef] [PubMed]
  45. Richards, S.; Aziz, D.; Bale, S.; Bick, J.; Das, W.W.; Gastier-Foster, M.; Grody, E.; Hegde, E.; Lyon, K.; Spector, H.L.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Wilde, A.A.M.; Semsarian, C.; Márquez, M.F.; Shamloo, A.S.; Ackerman, M.J.; Barajas-Martinez, H.S.; Behr, E.R.; Breckpot, B.J.; Charron, P.; Chockalingam, P.; et al. N EHRA/HRS/APHRS/LAHRS. Expert Consensus Statement on the state of genetic testing for cardiac diseases. Europace 2022, 24, 1307–1367. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Genetic testing strategy in HCM. Abbreviations: HCM: hypertrophic cardiomyopathy; VUS: variant of uncertain significance.
Figure 1. Genetic testing strategy in HCM. Abbreviations: HCM: hypertrophic cardiomyopathy; VUS: variant of uncertain significance.
Jcm 12 02489 g001
Table 1. Recommendations for the cardiologist on genetic testing in HCM.
Table 1. Recommendations for the cardiologist on genetic testing in HCM.
Genetic Testing in HCM: Tips for the Cardiologist
It is important to inform the patient that genetic testing:
Usually needs a long time (up to three months) before results are ready.
Has a chance of finding a causative mutation ranging from 30 to 60%, depending on the cardiomyopathy and its presence in other family members.
Can be extended to other family members ONLY in the presence of a pathogenic/likely pathogenic variant in the proband.
It is undesirable to suggest:
Performing the test as a matter of urgency.
Waiting for the test result to start treatment.
Submitting the entire family to genetic testing before determining the proband’s mutation.
Advising a prenatal diagnosis before genetic counselling.
Waiting for test results to decide on physical and sport activity.
Performing genetic testing for individuals in the family without phenotypic evidence of HCM, including relatives of sudden cardiac death victims.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Girolami, F.; Gozzini, A.; Pálinkás, E.D.; Ballerini, A.; Tomberli, A.; Baldini, K.; Marchi, A.; Zampieri, M.; Passantino, S.; Porcedda, G.; et al. Genetic Testing and Counselling in Hypertrophic Cardiomyopathy: Frequently Asked Questions. J. Clin. Med. 2023, 12, 2489. https://doi.org/10.3390/jcm12072489

AMA Style

Girolami F, Gozzini A, Pálinkás ED, Ballerini A, Tomberli A, Baldini K, Marchi A, Zampieri M, Passantino S, Porcedda G, et al. Genetic Testing and Counselling in Hypertrophic Cardiomyopathy: Frequently Asked Questions. Journal of Clinical Medicine. 2023; 12(7):2489. https://doi.org/10.3390/jcm12072489

Chicago/Turabian Style

Girolami, Francesca, Alessia Gozzini, Eszter Dalma Pálinkás, Adelaide Ballerini, Alessia Tomberli, Katia Baldini, Alberto Marchi, Mattia Zampieri, Silvia Passantino, Giulio Porcedda, and et al. 2023. "Genetic Testing and Counselling in Hypertrophic Cardiomyopathy: Frequently Asked Questions" Journal of Clinical Medicine 12, no. 7: 2489. https://doi.org/10.3390/jcm12072489

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop