1
|
Bains S, Giudicessi JR, Odening KE, Ackerman MJ. Gene therapy for cardiac arrhythmias. Nat Rev Cardiol 2025:10.1038/s41569-025-01168-5. [PMID: 40410593 DOI: 10.1038/s41569-025-01168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/25/2025]
Abstract
Cardiovascular diseases are the leading cause of death globally, with cardiac arrhythmias contributing substantially to this burden. Gene therapy, which directly targets the underlying disease pathobiology, offers an appealing treatment strategy for cardiac arrhythmias owing to its potential as a one-time, curative solution. Over the past two decades, substantial efforts have been made to develop new gene therapy approaches that overcome the limitations of conventional treatments. In this Review, we describe the rationale for gene therapy to treat cardiac arrhythmias; discuss advantages and disadvantages of gene silencing, gene replacement, gene suppression-and-replacement and gene editing technologies; summarize vector modalities and delivery approaches used in the field; present examples of gene therapy strategies used for atrial and ventricular arrhythmias; and highlight the current challenges and limitations in the gene therapy field.
Collapse
Affiliation(s)
- Sahej Bains
- Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - John R Giudicessi
- Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Michael J Ackerman
- Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA.
- Department of Paediatric and Adolescent Medicine, Division of Paediatric Cardiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Dicorato MM, Citarelli G, Mangini F, Alemanni R, Albanese M, Cicco S, Greco CA, Forleo C, Basile P, Carella MC, Ciccone MM, Guaricci AI, Dentamaro I. Integrative Approaches in the Management of Hypertrophic Cardiomyopathy: A Comprehensive Review of Current Therapeutic Modalities. Biomedicines 2025; 13:1256. [PMID: 40427081 PMCID: PMC12108688 DOI: 10.3390/biomedicines13051256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is often associated with left ventricular outflow tract (LVOT) obstruction, which affects a substantial proportion of patients. This obstruction results from a range of anatomical abnormalities involving both the valvular and subvalvular structures. Pharmacological therapies play a pivotal role in the management of LVOT obstruction, with a range of drug classes exhibiting distinct mechanisms of action. Beta-blockers, including atenolol and nadolol, are considered the first-line treatment due to their ability to reduce heart rate and myocardial contractility and enhance diastolic filling. Non-dihydropyridine calcium channel blockers, such as verapamil and diltiazem, are utilized as second-line agents when beta-blockers are ineffective or contraindicated. Disopyramid, a Class 1A antiarrhythmic agent, is employed for patients who do not respond to initial therapeutic interventions and can reduce LVOT gradients. Recent advancements in cardiac myosin modulators, such as Mavacamten and Aficamten, offer targeted therapies by modulating myosin-actin interactions to reduce LVOT gradients and improve symptoms, with promising results from clinical trials. Although gene therapy is still in its nascent stages, it has the potential to address the genetic basis of HCM by employing techniques such as genome editing, gene replacement, and the modulation of signaling pathways. For patients exhibiting severe symptoms or demonstrating unresponsiveness to medical treatment, invasive therapies, such as septal reduction therapy and alcohol septal ablation, are considered. Ultimately, the treatment and prevention of atrial fibrillation and sudden cardiac death are two key points of HCM management in both obstructive and non-obstructive forms. This review aims to provide an overview of current pharmacological and invasive strategies, as well as emerging therapies, in the management of HCM.
Collapse
Affiliation(s)
- Marco Maria Dicorato
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | | | - Francesco Mangini
- Cardiology Division, Miulli Hospital, 70021 Acquaviva delle Fonti, Italy;
| | - Rossella Alemanni
- Cardiac Surgery Division, Casa Sollievo Della Sofferenza Hospital, 71013 San Giovanni Rotondo, Italy;
| | - Miriam Albanese
- Division of Cardiology, V. Fazzi Hospital, 73100 Lecce, Italy;
| | - Sebastiano Cicco
- Internal Medicine Unit “Guido Baccelli”-Arterial Hypertension Unit “Anna Maria Pirrelli”, Department of Precision and Regenerative Medicine and Jonic Area (DiMePReJ), Polyclinic University Hospital, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Cinzia Forleo
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | - Paolo Basile
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | - Maria Cristina Carella
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | - Marco Matteo Ciccone
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | - Andrea Igoren Guaricci
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| | - Ilaria Dentamaro
- Cardiology Unit, Interdisciplinary Department of Medicine, Polyclinic University Hospital, University of Bari “Aldo Moro”, 70121 Bari, Italy; (C.F.); (P.B.); (M.C.C.); (M.M.C.); (A.I.G.); (I.D.)
| |
Collapse
|
3
|
Khattab E, Myrianthefs MM, Sakellaropoulos S, Alexandrou K, Mitsis A. Precision medicine applications in dilated cardiomyopathy: Advancing personalized care. Curr Probl Cardiol 2025; 50:103076. [PMID: 40381754 DOI: 10.1016/j.cpcardiol.2025.103076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Dilated cardiomyopathy (DCM) is a prevalent cardiac disorder affecting 1 in 250-500 individuals, characterized by ventricular dilation and impaired systolic function, leading to heart failure and increased mortality, including sudden cardiac death. DCM arises from genetic and environmental factors, such as drug-induced, inflammatory, and viral causes, resulting in diverse yet overlapping phenotypes. Advances in precision medicine are revolutionizing DCM management by leveraging genetic and molecular profiling for tailored diagnostic and therapeutic approaches. This review highlights comprehensive diagnostic evaluations, genetic discoveries, and multi-omics approaches integrating genomic, transcriptomic, proteomic, and metabolomic data to enhance understanding of DCM pathophysiology. Innovative risk stratification methods, including machine learning, are improving predictions of disease progression. Despite these advancements, the current one-size-fits-all management strategy contributes to persistently high morbidity and mortality. Emerging targeted therapies, such as CRISPR/Cas9 genome editing, aetiology-specific interventions, and pharmacogenomics, are reshaping treatment paradigms. Precision medicine holds promise for optimizing DCM diagnosis, treatment, and outcomes, aiming to reduce the burden of this debilitating condition.
Collapse
Affiliation(s)
- Elina Khattab
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Michael M Myrianthefs
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus
| | - Stefanos Sakellaropoulos
- Department of Internal Medicine, Cardiology Clinic, Kantonsspital Baden, Baden 5404, Switzerland
| | - Kyriakos Alexandrou
- Department of Nursing, School of Health Sciences, Cyprus University of Technology; Archiepiskopou Kyprianou 30, Limassol 3036, Cyprus
| | - Andreas Mitsis
- Cardiology Department, Consultant Interventional Cardiologist, Nicosia General Hospital, State Health Services Organization, 215, Old Road Nicosia-Limassol, Nicosia 2029, Cyprus.
| |
Collapse
|
4
|
Colombo G, Biering‐Sorensen T, Ferreira JP, Lombardi CM, Bonelli A, Garascia A, Metra M, Inciardi RM. Cardiac remodelling in the era of the recommended four pillars heart failure medical therapy. ESC Heart Fail 2025; 12:1029-1044. [PMID: 39600110 PMCID: PMC11911582 DOI: 10.1002/ehf2.15095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 11/29/2024] Open
Abstract
Cardiac remodelling is a key determinant of worse cardiovascular outcome in patients with heart failure (HF) and reduced ejection fraction (HFrEF). It affects both the left ventricle (LV) structure and function as well as the left atrium (LA) and the right ventricle (RV). Guideline recommended medical therapy for HF, including angiotensin-converting enzyme inhibitors/angiotensin receptors II blockers/angiotensin receptor blocker-neprilysin inhibitors (ACE-I/ARB/ARNI), beta-blockers, mineralocorticoid receptor antagonists (MRA) and sodium-glucose transport protein 2 inhibitors (SGLT2i), have shown to improve morbidity and mortality in patients with HFrEF. By targeting multiple pathophysiological pathways, foundational HF therapies are supposed to drive their beneficial clinical effects by a direct myocardial effect. Simultaneous initiation of guideline directed medical therapy (GDMT) through a synergistic effect promotes a 'reverse remodelling', leading to a full or partial recovered structure and function by enhancing systemic neurohumoral regulation and energy metabolism, reducing cardiomyocyte apoptosis, lowering oxidative stress and inflammation and adverse extracellular matrix deposition. The aim of this review is to describe how these classes of drugs can drive reverse remodelling in the LV, LA and RV and improve prognosis in patients with HFrEF.
Collapse
Affiliation(s)
- Giada Colombo
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili di BresciaUniversity of BresciaBresciaItaly
- Division of CardiovascularASST Grande Ospedale Metropolitano di NiguardaMilanItaly
| | - Tor Biering‐Sorensen
- Department of Cardiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Herlev and Gentofte HospitalUniversity of CopenhagenCopenhagenDenmark
| | - Joao P. Ferreira
- Department of Surgery and Physiology, Faculty of Medicine Cardiovascular Research and Development CenterUniversity of PortoPortoPortugal
| | - Carlo Mario Lombardi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili di BresciaUniversity of BresciaBresciaItaly
| | - Andrea Bonelli
- Division of CardiovascularASST Grande Ospedale Metropolitano di NiguardaMilanItaly
| | - Andrea Garascia
- Division of CardiovascularASST Grande Ospedale Metropolitano di NiguardaMilanItaly
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili di BresciaUniversity of BresciaBresciaItaly
| | - Riccardo M. Inciardi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili di BresciaUniversity of BresciaBresciaItaly
| |
Collapse
|
5
|
Greer-Short A, Greenwood A, Leon EC, Qureshi TN, von Kraut K, Wong J, Tsui JH, Reid CA, Cheng Z, Easter E, Yang J, Ho J, Steltzer S, Budan A, Cho M, Chandrakumar R, Cisne-Thompson O, Feathers C, Chung TW, Rodriguez N, Jones S, Alleyne-Levy C, Liu J, Jing F, Prince WS, Lin J, Ivey KN, Tingley WG, Hoey T, Lombardi LM. AAV9-mediated MYBPC3 gene therapy with optimized expression cassette enhances cardiac function and survival in MYBPC3 cardiomyopathy models. Nat Commun 2025; 16:2196. [PMID: 40038304 PMCID: PMC11880196 DOI: 10.1038/s41467-025-57481-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) affects approximately 600,000 people in the United States. Loss-of-function mutations in Myosin Binding Protein C3, MYBPC3, are the most common genetic cause of HCM, with the majority of mutations resulting in haploinsufficiency. To restore cardiac MYBPC3, we use an adeno-associated virus (AAV9) vector and engineer an optimized expression cassette with a minimal promoter and cis-regulatory elements (TN-201) to enhance packaging efficiency and cardiomyocyte expression. Rather than simply preventing cardiac dysfunction preclinically, we demonstrate in a symptomatic MYBPC3-deficient murine model the ability of AAV gene therapy to reverse cardiac hypertrophy and systolic dysfunction, improve diastolic dysfunction, and prolong survival. Dose-ranging efficacy studies exhibit restoration of wild-type MYBPC3 protein levels and saturation of cardiac improvement at the clinically relevant dose of 3E13 vg/kg, outperforming a previously published construct. These findings suggest that TN-201 may offer therapeutic benefits in MYBPC3-associated cardiomyopathy, pending further validation in clinical settings.
Collapse
Affiliation(s)
| | | | - Elena C Leon
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | | | - Justin Wong
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | | | - Ze Cheng
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | - Jin Yang
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Jaclyn Ho
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | - Ana Budan
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Marie Cho
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | | | | | | | | | | | | | - Jun Liu
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Frank Jing
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | - JianMin Lin
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | | | | - Timothy Hoey
- Tenaya Therapeutics, South San Francisco, CA, USA
| | | |
Collapse
|
6
|
Gigli M, Stolfo D, Merlo M, Sinagra G, Taylor MRG, Mestroni L. Pathophysiology of dilated cardiomyopathy: from mechanisms to precision medicine. Nat Rev Cardiol 2025; 22:183-198. [PMID: 39394525 PMCID: PMC12046608 DOI: 10.1038/s41569-024-01074-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/13/2024]
Abstract
Dilated cardiomyopathy (DCM) is a complex disease with multiple causes and various pathogenic mechanisms. Despite improvements in the prognosis of patients with DCM in the past decade, this condition remains a leading cause of heart failure and premature death. Conventional treatment for DCM is based on the foundational therapies for heart failure with reduced ejection fraction. However, increasingly, attention is being directed towards individualized treatments and precision medicine. The ability to confirm genetic causality is gradually being complemented by an increased understanding of genotype-phenotype correlations. Non-genetic factors also influence the onset of DCM, and growing evidence links genetic background with concomitant non-genetic triggers or precipitating factors, increasing the extreme complexity of the pathophysiology of DCM. This Review covers the spectrum of pathophysiological mechanisms in DCM, from monogenic causes to the coexistence of genetic abnormalities and triggering environmental factors (the 'two-hit' hypothesis). The roles of common genetic variants in the general population and of gene modifiers in disease onset and progression are also discussed. Finally, areas for future research are highlighted, particularly novel therapies, such as small molecules, RNA and gene therapy, and measures for the prevention of arrhythmic death.
Collapse
Affiliation(s)
- Marta Gigli
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Davide Stolfo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marco Merlo
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Centre for Diagnosis and Treatment of Cardiomyopathies, European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luisa Mestroni
- Molecular Genetics Program, Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
7
|
Protonotarios A, Asimaki A, Basso C, Xylouri Z, Monda E, Protonotarios I, Crisci G, Abrahms DJR, Anastasakis A, Antoniades L, Bakalakos A, Carbone A, S. Coonar A, Gimeno JR, Lazaros G, Lerakis S, Mestroni L, Papadopoulos G, Pecchia L, Prandi FR, Syrris P, Cadrin-Turigny J, Vasilakis A, Saffitz JE, Gaetano Thiene S, Elliott PM, Kaski JP, McKenna WJ, Bossone E, Limongelli G, Tsatsopoulou A. Naxos Disease and Related Cardio-Cutaneous Syndromes. JACC. ADVANCES 2025; 4:101547. [PMID: 39877668 PMCID: PMC11773020 DOI: 10.1016/j.jacadv.2024.101547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 01/31/2025]
Abstract
Naxos disease is a rare autosomal recessive condition combining arrhythmogenic right ventricular cardiomyopathy, woolly hair, and palmoplantar keratoderma. The first identified causative variant was in the gene encoding the desmosomal protein plakoglobin. Naxos disease exhibits fibro-fatty myocardial replacement with immunohistological abnormalities in cardiac protein and signaling pathways, highlighting the role of inflammation and potential anti-inflammatory treatments. Childhood cutaneous signs precede cardiac features, which are diagnosed by familial and genetic evaluation, electrocardiography and cardiac imaging. Disease progression necessitates holistic care with risk management and lifestyle adjustments, often needing treatment for arrhythmia and heart failure. Similar phenotypes have been linked to desmoplakin and rarely desmocollin2 gene variants, highlighting the importance of familial and genetic evaluation. This document summarizes current knowledge on Naxos disease and related cardiocutaneous syndromes and initiates an international endeavor to collect and study all global cases, aiming to improve understanding, treatment, and patient care through shared data and research.
Collapse
Affiliation(s)
| | - Angeliki Asimaki
- Cardiovascular and Genomics Research Institute of City, St George’s University of London, London, UK
| | | | | | - Emanuele Monda
- Department of Traslational Sciences, University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | - Ioannis Protonotarios
- Cardiovascular and Genomics Research Institute of City, St George’s University of London, London, UK
| | - Giulia Crisci
- University of Naples Federico II, Naples, Italy
- Department of Cardiology, San Paolo Hospital, University of Milan, Milan, Italy
| | - Dominic JR. Abrahms
- Center for Cardiovascular Genetics, Boston Children’s Hospital, Harvard Medical School, Boston, USA
| | - Aris Anastasakis
- Inherited Cardiovascular Diseases, Onassis Cardiac Surgery Centre, Athens, Greece
| | | | - Athanasios Bakalakos
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, UK
| | | | - Aman S. Coonar
- Cardiothoracic Surgery, Royal Papworth Hospital, Cambridge University Health Partners, Cambridge, UK
| | - Juan Ramon Gimeno
- Inherited Cardiac Disease Department (CSUR/ ERN Guard Heart), Virgen de la Arrixaca University Hospital, Murcia, Spain
| | - George Lazaros
- George Lazaros, First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stamatis Lerakis
- Department of Cardiology, Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Luisa Mestroni
- Medicine/Cardiology, Genetics Program, University of Colorado Cardiovascular Institute, Aurora, USA
| | | | | | | | - Petros Syrris
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Julia Cadrin-Turigny
- Cardiovascular Genetics Center, Montréal Heart Institute, Faculty of Medicine, University of Montréal, Montréal, Québec, Canada
| | | | - Jeffrey E. Saffitz
- Harvard Medical School, Department of Pathology, Beth Israel Deaconess Medical Center, Boston, USA
| | | | - Perry M. Elliott
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, UK
| | - Juan Pablo Kaski
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, UK
| | - William J. McKenna
- Centre for Heart Muscle Disease, UCL Institute of Cardiovascular Science, London, UK
| | | | - Giuseppe Limongelli
- Department of Traslational Sciences, University of Campania ‘Luigi Vanvitelli’, Naples, Italy
| | - Adalena Tsatsopoulou
- Precision Medicine Network in Cardiology Onassis Cardiac Surgery Center, Athens, Greece
- Pediatric Clinic, Naxos, Cyclades, Greece
| |
Collapse
|
8
|
Aimo A, Olivotto I, Todiere G, Barison A, Panichella G, Fiuzat M, Linde C, Lakdawala NK, Desai M, Zannad F, Maron MS. Endpoint Selection in Randomized Clinical Trials for Hypertrophic Cardiomyopathy. JACC. HEART FAILURE 2025; 13:200-212. [PMID: 39708028 DOI: 10.1016/j.jchf.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 10/02/2024] [Indexed: 12/23/2024]
Abstract
Randomized clinical trials (RCTs) for hypertrophic cardiomyopathy (HCM) have long been challenging caused by the condition's rarity, low event rates, and diverse clinical presentations. However, recent advances in targeted therapies have sparked increased interest in HCM research. Despite this, designing effective RCTs remains complex, particularly in identifying clinically meaningful endpoints. HCM, often linked to sequence variation in sarcomeric protein genes like MYH7 and MYBPC3, exhibits varied phenotypic expressions that influence disease progression and treatment responses. This genetic variability underscores the need for personalized approaches in clinical trials. Emerging gene therapies, such as CRISPR/Cas9, show promise in addressing these genetic factors. A major challenge in HCM RCTs is ensuring that endpoints are both statistically and clinically significant, given issues like test-retest variability and missing data. Primary endpoints often focus on symptom relief and functional improvement, while secondary and exploratory endpoints provide broader insights into treatment effects. Regulatory authorities are increasingly open to a wider range of endpoints, including patient-reported outcomes and functional measures, although the cost-risk balance is crucial, especially for high-risk interventions. Future HCM RCTs may incorporate hard clinical endpoints such as heart failure hospitalization, atrial fibrillation recurrence, and all-cause mortality, offering a more comprehensive evaluation of treatment efficacy. Integrating genetic insights and advanced technologies will be essential to improving trial design and enhancing patient outcomes in HCM.
Collapse
Affiliation(s)
- Alberto Aimo
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | | | - Giancarlo Todiere
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Andrea Barison
- Health Sciences Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy; Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giorgia Panichella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mona Fiuzat
- Duke University and Duke Clinical Research Institute, Durham, NC, USA
| | - Cecilia Linde
- Karolinska Institutet, Department of Medicine, Cardiology Unit, Stockholm, Sweden; Karolinska University Hospital, Department of Cardiology, Stockholm, Sweden
| | - Neal K Lakdawala
- Heart and Vascular Center, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Milind Desai
- Hypertrophic Cardiomyopathy Center, Heart Vascular Thoracic Institute Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Martin S Maron
- Hypertrophic Cardiomyopathy Center, Lahey Hospital and Medical Center, Burlington, Massachusetts, USA
| |
Collapse
|
9
|
Zhang L, Ding F, Ren Z, Cheng W, Dai H, Liang Q, Kong F, Xu W, Wang M, Zhang Y, Tao Q. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TNNI3 c.235C > T variant. Int J Cardiol 2025; 419:132627. [PMID: 39426416 DOI: 10.1016/j.ijcard.2024.132627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is typically manifested as a hereditary disorder, with 30 %-60 % of cases linked to cardiac sarcomere gene mutations. Despite numerous identified TNNI3 mutations associated with HCM, their severity, prevalence, and disease progression vary. The link between TNNI3 variants and phenotypes remains largely unexplored. This study aims to elucidate the impact of the TNNI3 c.235C > T mutation on HCM through clinical research and cell experiments and to explore its mechanism in HCM development. METHODS We screened an HCM family for pathogenic gene mutations using gene sequencing. The proband and family members were assessed through electrocardiography, echocardiography, and cardiac MRI, and a pedigree map was created for disease prediction analysis. Mutant plasmids were constructed with the TNNI3 c.235C > T mutation and transfected into the AC16 human cardiomyocyte cell line to investigate the mutation's effects. RESULTS The TNNI3 c.235C > T mutation was identified as the disease-causing variant in the family. This mutation led to the upregulation of hypertrophy-associated genes ANP, BNP, and MYH7, increased cardiomyocyte size, and activation of the ERK signaling pathway. Further investigations revealed that the TNNI3 c.235C > T mutation impaired mitochondrial function, disrupted cardiomyocyte metabolism, and increased cellular autophagy and apoptosis. CONCLUSIONS The TNNI3 c.235C > T gene mutation may be a pathogenic factor for HCM, showing heterogeneous features and clinical phenotypes. This mutation induces myocardial hypertrophy, activates the ERK signaling pathway, and exacerbates mitochondrial dysfunction, apoptosis, and autophagy in cardiomyocytes. These findings provide insights into the mechanism of HCM caused by gene mutations and may inform HCM treatment strategies.
Collapse
Affiliation(s)
- Lai Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fengzhi Ding
- Department of Physiology, Wannan Medical College, Wuhu, Anhui, 241000, China
| | - Zhongyuan Ren
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Weili Cheng
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - He Dai
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Qing Liang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fanling Kong
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Wenjing Xu
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Minghui Wang
- College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yuqing Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| | - Qin Tao
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
10
|
Zhang H, Ma X, Zhao L. Editorial for "Second-Order Motion-Compensated Echo-Planar Cardiac Diffusion-Weighted MRI: Usefulness of Compressed Sensitivity Encoding". J Magn Reson Imaging 2025; 61:319-320. [PMID: 38662936 DOI: 10.1002/jmri.29410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 12/15/2024] Open
Affiliation(s)
- Hongbo Zhang
- Department of Interventional Diagnosis and Treatment, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaohai Ma
- Department of Interventional Diagnosis and Treatment, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lei Zhao
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Jordan E, Ni H, Parker P, Kinnamon DD, Owens A, Lowes B, Shenoy C, Martin CM, Judge DP, Fishbein DP, Stoller D, Minami E, Kransdorf E, Smart F, Haas GJ, Huggins GS, Ewald GA, Diamond J, Wilcox JE, Jimenez J, Wang J, Tallaj J, Drazner MH, Hofmeyer M, Wheeler MT, Pinzon OW, Shah P, Gottlieb SS, Katz S, Shore S, Tang WHW, Hershberger RE, DCM Precision Medicine study of the DCM Consortium. Implementing Precision Medicine for Dilated Cardiomyopathy: Insights from The DCM Consortium. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.22.24317816. [PMID: 39649582 PMCID: PMC11623749 DOI: 10.1101/2024.11.22.24317816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Clinical genetic evaluation of dilated cardiomyopathy (DCM) is implemented variably or not at all. Identifying needs and barriers to genetic evaluations will enable strategies to enhance precision medicine care. Methods An online survey was conducted in June 2024 among cardiologist investigators of the DCM Consortium from US advanced heart failure/transplant (HF/TX) programs to collect demographics, training, program characteristics, genetic evaluation practices for DCM, and implementation needs. An in-person discussion followed. Results Twenty-five cardiologists (28% female, 12% Hispanic, 68% White) participated in the survey and 15 in the discussion; genetics training backgrounds varied greatly. Clinical genetic testing for DCM was conducted by all programs with annual uptake ranging from 5%-70% (median 25%). Thirteen respondents (52%) did not use selection criteria for testing whereas others selected patients based on specific clinical and family history data. Eight (32%) ordered testing by themselves, and the remainder had testing managed mostly by a genetic counselor or others with genetic expertise (16/17; 94%). Six themes were distilled from open-ended responses regarding thoughts for the future and included access to genetics services, navigating uncertainty, knowledge needs, cost concerns, family-based care barriers, and institutional infrastructure limitations. Following an in-person discussion, four areas were identified for focused effort: improved reimbursement for genetic services, genetic counselor integration with HF/TX teams, improved provider education resources, and more research to find missing heritability and to resolve uncertain results. Conclusions HF/TX programs have implementation challenges in the provision of DCM genetic evaluations; targeted plans to facilitate precision medicine for DCM are needed.
Collapse
Affiliation(s)
- Elizabeth Jordan
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Hanyu Ni
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Patricia Parker
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Daniel D. Kinnamon
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Anjali Owens
- Center for Inherited Cardiovascular Disease, Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brian Lowes
- University of Nebraska Medical Center, Omaha, NE
| | | | | | | | | | | | | | - Evan Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Frank Smart
- Louisiana State University Health Sciences Center, New Orleans, LA
| | - Garrie J. Haas
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Gordon S. Huggins
- Cardiology Division, Tufts Medical Center and Tufts University School of Medicine, Boston, MA
| | | | | | - Jane E. Wilcox
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Javier Jimenez
- Miami Cardiac & Vascular Institute, Baptist Health South, Miami, FL
| | - Jessica Wang
- University of California Los Angeles Medical Center, Los Angeles, CA
| | - Jose Tallaj
- University of Alabama at Birmingham; Washington, DC
| | - Mark H. Drazner
- University of Texas Southwestern Medical Center; Washington, DC
| | - Mark Hofmeyer
- MedStar Health Research Institute, Medstar Washington Hospital Center, Washington, DC
| | - Matthew T. Wheeler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Palak Shah
- Inova Schar Heart and Vascular, Falls Church, VA
| | | | - Stuart Katz
- New York University Langone Medical Center, New York, NY
| | | | - W. H. Wilson Tang
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| | - Ray E. Hershberger
- The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University, Columbus, OH
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | | |
Collapse
|
12
|
Grisorio L, Bongianino R, Gianeselli M, Priori SG. Gene therapy for cardiac diseases: methods, challenges, and future directions. Cardiovasc Res 2024; 120:1664-1682. [PMID: 39302117 DOI: 10.1093/cvr/cvae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024] Open
Abstract
Gene therapy is advancing at an unprecedented pace, and the recent success of clinical trials reinforces optimism and trust among the scientific community. Recently, the cardiac gene therapy pipeline, which had progressed more slowly than in other fields, has begun to advance, overcoming biological and technical challenges, particularly in treating genetic heart pathologies. The primary rationale behind the focus on monogenic cardiac diseases is the well-defined molecular mechanisms driving their phenotypes, directly linked to the pathogenicity of single genetic mutations. This aspect makes these conditions a remarkable example of 'genetically druggable' diseases. Unfortunately, current treatments for these life-threatening disorders are few and often poorly effective, underscoring the need to develop therapies to modulate or correct their molecular substrates. In this review we examine the latest advancements in cardiac gene therapy, discussing the pros and cons of different molecular approaches and delivery vectors, with a focus on their therapeutic application in cardiac inherited diseases. Additionally, we highlight the key factors that may enhance clinical translation, drawing insights from previous trials and the current prospects of cardiac gene therapy.
Collapse
Affiliation(s)
- Luca Grisorio
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
| | - Rossana Bongianino
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
| | - Matteo Gianeselli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Silvia Giuliana Priori
- Department of Internal Medicine, University of Pavia, Via Golgi 19, Pavia, 27100, Italy
- Molecular Cardiology, IRCCS Istituti Clinici Scientifici Maugeri, Via Maugeri 10, Pavia, 27100, Italy
- Molecular Cardiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), C/ Melchor Fernandez Almagro 3, Madrid, 28029, Spain
| |
Collapse
|
13
|
Torbey AFM, Couto RGT, Grippa A, Maia EC, Miranda SA, Santos MACD, Peres ET, Costa OPS, Oliveira EMD, Mesquita ET. Cardiomyopathy in Children and Adolescents in the Era of Precision Medicine. Arq Bras Cardiol 2024; 121:e20230154. [PMID: 39442130 PMCID: PMC11634207 DOI: 10.36660/abc.20230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/18/2024] [Accepted: 05/15/2024] [Indexed: 10/25/2024] Open
Abstract
In childhood and adolescence, cardiomyopathies have their own characteristics and are an important cause of heart failure, arrhythmias, sudden death, and indication for heart transplantation. Diagnosis is a challenge in daily practice due to its varied clinical presentation, heterogeneous etiologies, and limited knowledge of tools related to clinical and molecular genetics. However, it is essential to recognize the different phenotypes and prioritize the search for the etiology. Recent advances in precision medicine have made molecular diagnosis accessible, which makes it possible to individualize therapeutic approaches, stratify the prognosis, and identify individuals in the family who are at risk of developing the disease. The objective of this review is to emphasize the particularities of cardiomyopathies in pediatrics and how the individualized approach impacts the therapy and prognosis of the patient. Through a systematized approach, the five-stage protocol used in our service is presented. These stages bring together clinical evaluation for determining the morphofunctional phenotype, identification of etiology, classification, establishment of prognosis, and the search for personalized therapies.
Collapse
Affiliation(s)
- Ana Flávia Mallheiros Torbey
- Universidade Federal Fluminense, Niterói, RJ - Brasil
- Programa de Pós-Graduação em Ciências Cardiovasculares da Universidade Federal Fluminense, Niterói, RJ - Brasil
| | - Raquel Germer Toja Couto
- Universidade Federal Fluminense Hospital Universitário Antônio Pedro (EBSERH), Niterói, RJ - Brasil
| | - Aurea Grippa
- Universidade Federal Fluminense, Niterói, RJ - Brasil
| | | | | | | | | | | | | | - Evandro Tinoco Mesquita
- Universidade Federal Fluminense, Niterói, RJ - Brasil
- Programa de Pós-Graduação em Ciências Cardiovasculares da Universidade Federal Fluminense, Niterói, RJ - Brasil
- Complexo Hospitalar de Niteroi, Niterói, RJ - Brasil
| |
Collapse
|
14
|
Ormondroyd E, Grace C, Borsari W, Goel A, McDonough B, Rose J, Seidman C, Watkins H. Genetic therapies for cardiomyopathy: survey of attitudes of the patient community for the CureHeart project. Eur J Hum Genet 2024; 32:1045-1052. [PMID: 38972962 PMCID: PMC11368914 DOI: 10.1038/s41431-024-01660-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Cardiomyopathies are a group of inherited heart muscle disorders. Expressivity is variable and while sometimes mild, complications can result in sudden cardiac death (SCD) at any age, heart failure and stroke. In around a third of patients a monogenic cause is identifiable, and development of genetic therapies that aim to correct the underlying genetic defect is underway. Here we describe results of a survey designed to understand preliminary views of the patient community about genetic therapies in the context of disease burden. The internet survey was publicized with a bespoke information video via patient support groups in the UK and USA; 634 people responded of whom 96% had a personal and/or family history of cardiomyopathy. Findings show that concern about cardiomyopathy-related issues with a future dimension, such as disease progression, is significantly greater than concern about current issues. A total of 93.6% thought that genetic therapies should be developed for cardiomyopathy. A majority would consider participation in a genetic therapy trial in six scenarios varying by age and clinical situation significantly more in the scenario of an adult with symptomatic disease and evident progression than an asymptomatic adult with SCD risk, or a child. In all scenarios, a majority said that the chance genetic therapy would stop or slow progression, and risk of serious adverse and unintended effects, were important considerations. Qualitative analysis of free-text responses found that concern was often informed by family experience. Patient consideration of genetic therapy is likely to require individualized assessment of the benefits and risks.
Collapse
Affiliation(s)
- Elizabeth Ormondroyd
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- NIHR Biomedical Research Centre, Oxford, UK.
| | - Christopher Grace
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Barbara McDonough
- Harvard Medical School Department of Genetics, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Christine Seidman
- Harvard Medical School Department of Genetics, Boston, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, Oxford, UK
| |
Collapse
|
15
|
Johnston JR, Adler ED. Precision Genetic Therapies: Balancing Risk and Benefit in Patients with Heart Failure. Curr Cardiol Rep 2024; 26:973-983. [PMID: 39110386 PMCID: PMC11379760 DOI: 10.1007/s11886-024-02096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE OF REVIEW Precision genetic medicine is evolving at a rapid pace and bears significant implications for clinical cardiology. Herein, we discuss the latest advancements and emerging strategies in gene therapy for cardiomyopathy and heart failure. RECENT FINDINGS Elucidating the genetic architecture of heart failure has paved the way for precision therapies in cardiovascular medicine. Recent preclinical studies and early-phase clinical trials have demonstrated encouraging results that support the development of gene therapies for heart failure arising from a variety of etiologies. In addition to the discovery of new therapeutic targets, innovative delivery platforms are being leveraged to improve the safety and efficacy of cardiac gene therapies. Precision genetic therapy represents a potentially safe and effective approach for improving outcomes in patients with heart failure. It holds promise for radically transforming the treatment paradigm for heart failure by directly targeting the underlying etiology. As this new generation of cardiovascular medicines progress to the clinic, it is especially important to carefully evaluate the benefits and risks for patients.
Collapse
Affiliation(s)
- Jamie R Johnston
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, 32306, USA
| | - Eric D Adler
- Division of Cardiology, Department of Internal Medicine, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
16
|
Lopes LR, Ho CY, Elliott PM. Genetics of hypertrophic cardiomyopathy: established and emerging implications for clinical practice. Eur Heart J 2024; 45:2727-2734. [PMID: 38984491 PMCID: PMC11313585 DOI: 10.1093/eurheartj/ehae421] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/05/2023] [Accepted: 06/19/2024] [Indexed: 07/11/2024] Open
Abstract
Pathogenic variation in genes encoding proteins of the cardiac sarcomere is responsible for 30%-40% of cases of hypertrophic cardiomyopathy. The main clinical utility of genetic testing is to provide diagnostic confirmation and facilitation of family screening. It also assists in the detection of aetiologies, which require distinct monitoring and treatment approaches. Other clinical applications, including the use of genetic information to inform risk prediction models, have been limited by the challenge of establishing robust genotype-phenotype correlations with actionable consequences, but new data on the interaction between rare and common genetic variation, as well as the emergence of therapies targeting disease-specific pathogenic mechanisms, herald a new era for genetic testing in routine practice.
Collapse
Affiliation(s)
- Luis R Lopes
- Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 5 University St, London WC1E 6JF, UK
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Perry M Elliott
- Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 5 University St, London WC1E 6JF, UK
| |
Collapse
|
17
|
Neufeldt D, Schmidt A, Mohr E, Lu D, Chatterjee S, Fuchs M, Xiao K, Pan W, Cushman S, Jahn C, Juchem M, Hunkler HJ, Cipriano G, Jürgens B, Schmidt K, Groß S, Jung M, Hoepfner J, Weber N, Foo R, Pich A, Zweigerdt R, Kraft T, Thum T, Bär C. Circular RNA circZFPM2 regulates cardiomyocyte hypertrophy and survival. Basic Res Cardiol 2024; 119:613-632. [PMID: 38639887 PMCID: PMC11319402 DOI: 10.1007/s00395-024-01048-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) constitutes the most common genetic cardiac disorder. However, current pharmacotherapeutics are mainly symptomatic and only partially address underlying molecular mechanisms. Circular RNAs (circRNAs) are a recently discovered class of non-coding RNAs and emerged as specific and powerful regulators of cellular functions. By performing global circRNA-specific next generation sequencing in cardiac tissue of patients with hypertrophic cardiomyopathy compared to healthy donors, we identified circZFPM2 (hsa_circ_0003380). CircZFPM2, which derives from the ZFPM2 gene locus, is a highly conserved regulatory circRNA that is strongly induced in HCM tissue. In vitro loss-of-function experiments were performed in neonatal rat cardiomyocytes, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), and HCM-patient-derived hiPSC-CMs. A knockdown of circZFPM2 was found to induce cardiomyocyte hypertrophy and compromise mitochondrial respiration, leading to an increased production of reactive oxygen species and apoptosis. In contrast, delivery of recombinant circZFPM2, packaged in lipid-nanoparticles or using AAV-based overexpression, rescued cardiomyocyte hypertrophic gene expression and promoted cell survival. Additionally, HCM-derived cardiac organoids exhibited improved contractility upon CM-specific overexpression of circZFPM2. Multi-Omics analysis further promoted our hypothesis, showing beneficial effects of circZFPM2 on cardiac contractility and mitochondrial function. Collectively, our data highlight that circZFPM2 serves as a promising target for the treatment of cardiac hypertrophy including HCM.
Collapse
MESH Headings
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Humans
- Animals
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/pathology
- Cardiomyopathy, Hypertrophic/metabolism
- Induced Pluripotent Stem Cells/metabolism
- Rats
- Cell Survival
- Apoptosis/genetics
- Cells, Cultured
- Reactive Oxygen Species/metabolism
- RNA/genetics
- Animals, Newborn
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Dimyana Neufeldt
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Arne Schmidt
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Elisa Mohr
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Dongchao Lu
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Ke Xiao
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Wen Pan
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Sarah Cushman
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Christopher Jahn
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Malte Juchem
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Hannah Jill Hunkler
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Giuseppe Cipriano
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Bjarne Jürgens
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Sonja Groß
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Jeannine Hoepfner
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Roger Foo
- Institute of Molecular and Cell Biology, A*Star, Singapore, Singapore
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
- Core Facility Proteomics, Institute of Toxicology, Hannover, Germany
| | - Robert Zweigerdt
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute for Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany.
| |
Collapse
|
18
|
Falcão-Pires I, Ferreira AF, Trindade F, Bertrand L, Ciccarelli M, Visco V, Dawson D, Hamdani N, Van Laake LW, Lezoualc'h F, Linke WA, Lunde IG, Rainer PP, Abdellatif M, Van der Velden J, Cosentino N, Paldino A, Pompilio G, Zacchigna S, Heymans S, Thum T, Tocchetti CG. Mechanisms of myocardial reverse remodelling and its clinical significance: A scientific statement of the ESC Working Group on Myocardial Function. Eur J Heart Fail 2024; 26:1454-1479. [PMID: 38837573 DOI: 10.1002/ejhf.3264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/22/2024] [Accepted: 04/18/2024] [Indexed: 06/07/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbimortality in Europe and worldwide. CVD imposes a heterogeneous spectrum of cardiac remodelling, depending on the insult nature, that is, pressure or volume overload, ischaemia, arrhythmias, infection, pathogenic gene variant, or cardiotoxicity. Moreover, the progression of CVD-induced remodelling is influenced by sex, age, genetic background and comorbidities, impacting patients' outcomes and prognosis. Cardiac reverse remodelling (RR) is defined as any normative improvement in cardiac geometry and function, driven by therapeutic interventions and rarely occurring spontaneously. While RR is the outcome desired for most CVD treatments, they often only slow/halt its progression or modify risk factors, calling for novel and more timely RR approaches. Interventions triggering RR depend on the myocardial insult and include drugs (renin-angiotensin-aldosterone system inhibitors, beta-blockers, diuretics and sodium-glucose cotransporter 2 inhibitors), devices (cardiac resynchronization therapy, ventricular assist devices), surgeries (valve replacement, coronary artery bypass graft), or physiological responses (deconditioning, postpartum). Subsequently, cardiac RR is inferred from the degree of normalization of left ventricular mass, ejection fraction and end-diastolic/end-systolic volumes, whose extent often correlates with patients' prognosis. However, strategies aimed at achieving sustained cardiac improvement, predictive models assessing the extent of RR, or even clinical endpoints that allow for distinguishing complete from incomplete RR or adverse remodelling objectively, remain limited and controversial. This scientific statement aims to define RR, clarify its underlying (patho)physiologic mechanisms and address (non)pharmacological options and promising strategies to promote RR, focusing on the left heart. We highlight the predictors of the extent of RR and review the prognostic significance/impact of incomplete RR/adverse remodelling. Lastly, we present an overview of RR animal models and potential future strategies under pre-clinical evaluation.
Collapse
Affiliation(s)
- Inês Falcão-Pires
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Ana Filipa Ferreira
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Fábio Trindade
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Luc Bertrand
- Université Catholique de Louvain, Institut de Recherche Expérimentale et Clinique, Pôle of Cardiovascular Research, Brussels, Belgium
- WELBIO, Department, WEL Research Institute, Wavre, Belgium
| | - Michele Ciccarelli
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Valeria Visco
- Cardiovascular Research Unit, Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Dana Dawson
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Department of Physiology, Cardiovascular Research Institute Maastricht University Maastricht, Maastricht, the Netherlands
| | - Linda W Van Laake
- Division Heart and Lungs, Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank Lezoualc'h
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm, Université Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway
- KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| | - Mahmoud Abdellatif
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | | | - Nicola Cosentino
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Cardiovascular Section, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alessia Paldino
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Giulio Pompilio
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Serena Zacchigna
- Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Stephane Heymans
- Department of Cardiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
- Centre of Cardiovascular Research, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| |
Collapse
|
19
|
Mundisugih J, Ravindran D, Kizana E. Exploring the Therapeutic Potential of Gene Therapy in Arrhythmogenic Right Ventricular Cardiomyopathy. Biomedicines 2024; 12:1351. [PMID: 38927558 PMCID: PMC11201581 DOI: 10.3390/biomedicines12061351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Right dominant arrhythmogenic cardiomyopathy, commonly known as Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), represents a formidable challenge in cardiovascular medicine, as conventional therapies are commonly ineffective in impeding disease progression and the development of end-stage heart failure. Recombinant adeno-associated virus (AAV)-mediated gene therapy presents a promising avenue for targeted therapeutic interventions, potentially revolutionising treatment approaches for ARVC patients. Encouraging results from preclinical studies have sparked optimism about the possibility of curing specific subtypes of ARVC in the near future. This narrative review delves into the dynamic landscape of genetic therapy for ARVC, elucidating its underlying mechanisms and developmental stages, and providing updates on forthcoming trials. Additionally, it examines the hurdles and complexities impeding the successful translation of ARVC genetic therapies into clinical practice. Despite notable scientific advancements, the journey towards implementing genetic therapies for ARVC patients in real-world clinical settings is still in its early phases.
Collapse
Affiliation(s)
- Juan Mundisugih
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Dhanya Ravindran
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
20
|
Bains S, Giudicessi JR, Odening KE, Ackerman MJ. State of Gene Therapy for Monogenic Cardiovascular Diseases. Mayo Clin Proc 2024; 99:610-629. [PMID: 38569811 DOI: 10.1016/j.mayocp.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024]
Abstract
Over the past 2 decades, significant efforts have been made to advance gene therapy into clinical practice. Although successful examples exist in other fields, gene therapy for the treatment of monogenic cardiovascular diseases lags behind. In this review, we (1) highlight a brief history of gene therapy, (2) distinguish between gene silencing, gene replacement, and gene editing technologies, (3) discuss vector modalities used in the field with a special focus on adeno-associated viruses, (4) provide examples of gene therapy approaches in cardiomyopathies, channelopathies, and familial hypercholesterolemia, and (5) present current challenges and limitations in the gene therapy field.
Collapse
Affiliation(s)
- Sahej Bains
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN
| | - John R Giudicessi
- Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine (Division of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo Clinic, Rochester, MN
| | - Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Michael J Ackerman
- Department of Molecular Pharmacology and Experimental Therapeutics (Windland Smith Rice Sudden Death Genomics Laboratory), Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine (Division of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo Clinic, Rochester, MN; Department of Pediatric and Adolescent Medicine (Division of Pediatric Cardiology), Mayo Clinic, Rochester, MN.
| |
Collapse
|
21
|
Silver E, Argiro A, Hong K, Adler E. Gene therapy vector-related myocarditis. Int J Cardiol 2024; 398:131617. [PMID: 38030043 DOI: 10.1016/j.ijcard.2023.131617] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Gene therapy is a technique to correct genetic abnormalities, through introduction of a functional gene or through direct genome editing. Adeno-associated virus (AAV)-mediated gene replacement shows promise for targeted therapies in treatment of inherited cardiomyopathies and is the most used approach in clinical trials. However, immune responses from the host to the virus and gene product pose delivery and safety challenges. This review explores the immunological reactions to AAV-based gene therapy, their potential toxic effects, with a focus on myocarditis, and future directions for gene therapy.
Collapse
Affiliation(s)
- Elizabeth Silver
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, San Diego, CA, United States; School of Medicine, University of Connecticut Health Center, Farmington, CT, United States.
| | - Alessia Argiro
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Kimberly Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, San Diego, CA, United States
| | - Eric Adler
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
22
|
Abstract
Dilated cardiomyopathy (DCM) is defined as dilation and/or reduced function of one or both ventricles and remains a common disease worldwide. An estimated 40% of cases of familial DCM have an identifiable genetic cause. Accordingly, there is a fast-growing interest in the field of molecular genetics as it pertains to DCM. Many gene mutations have been identified that contribute to phenotypically significant cardiomyopathy. DCM genes can affect a variety of cardiomyocyte functions, and particular genes whose function affects the cell-cell junction and cytoskeleton are associated with increased risk of arrhythmias and sudden cardiac death. Through advancements in next-generation sequencing and cardiac imaging, identification of genetic DCM has improved over the past couple decades, and precision medicine is now at the forefront of treatment for these patients and their families. In addition to standard treatment of heart failure and prevention of arrhythmias and sudden cardiac death, patients with genetic cardiomyopathy stand to benefit from gene mechanism-specific therapies.
Collapse
Affiliation(s)
- Ramone Eldemire
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA;
| | - Luisa Mestroni
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA;
- Cardiovascular Institute, University of Colorado, Aurora, Colorado, USA
| | - Matthew R G Taylor
- Cardiovascular Institute, University of Colorado, Aurora, Colorado, USA
- Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
23
|
Al-Aidarous S, Protonotarios A, Elliott PM, Lambiase PD. Management of arrhythmogenic right ventricular cardiomyopathy. Heart 2024; 110:156-162. [PMID: 37433658 DOI: 10.1136/heartjnl-2023-322612] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/26/2023] [Indexed: 07/13/2023] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a disease characterised by fibrofatty replacement of the ventricular myocardium due to specific mutations, leading to ventricular arrhythmias and sudden cardiac death. Treating this condition can be challenging due to progressive fibrosis, phenotypic variations and small patient cohorts limiting the feasibility of conducting meaningful clinical trials. Although widely used, the evidence base for anti-arrhythmic drugs is limited. Beta-blockers are theoretically sound, yet their efficacy in reducing arrhythmic risk is not robust. Additionally, the impact of sotalol and amiodarone is inconsistent with studies reporting contradictory results. Emerging evidence suggests that combining flecainide and bisoprolol may be efficacious.Radiofrequency ablation has shown some potential in disrupting ventricular tachycardia circuits, with combined endo and epicardial ablation yielding better results which could be considered at the index procedure. In addition, stereotactic radiotherapy may be a future option that can decrease arrhythmias beyond simple scar formation by altering levels of Nav1.5 channels, Connexin 43 and Wnt signalling, potentially modifying myocardial fibrosis.Future therapies, such as adenoviruses and GSk3b modulation, are still in early-stage research. While implantable cardioverter-defibrillator implantation is a key intervention for reducing arrhythmic death, the risks of inappropriate shocks and device complications must be carefully considered.
Collapse
Affiliation(s)
- Sayed Al-Aidarous
- Institute of Cardiovascular Science, University College London, London, UK
| | - Alexandros Protonotarios
- Institute of Cardiovascular Science, University College London, London, UK
- St Bartholomew's Hospital, London, UK
| | - Perry M Elliott
- Institute of Cardiovascular Science, University College London, London, UK
| | - Pier D Lambiase
- Institute of Cardiovascular Science, University College London, London, UK
- Department of Cardiology, Saint Bartholomew's Hospital, Barts Heart Centre, London, UK
| |
Collapse
|
24
|
Curran L, de Marvao A, Inglese P, McGurk KA, Schiratti PR, Clement A, Zheng SL, Li S, Pua CJ, Shah M, Jafari M, Theotokis P, Buchan RJ, Jurgens SJ, Raphael CE, Baksi AJ, Pantazis A, Halliday BP, Pennell DJ, Bai W, Chin CW, Tadros R, Bezzina CR, Watkins H, Cook SA, Prasad SK, Ware JS, O’Regan DP. Genotype-Phenotype Taxonomy of Hypertrophic Cardiomyopathy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:e004200. [PMID: 38014537 PMCID: PMC10729901 DOI: 10.1161/circgen.123.004200] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/25/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is an important cause of sudden cardiac death associated with heterogeneous phenotypes, but there is no systematic framework for classifying morphology or assessing associated risks. Here, we quantitatively survey genotype-phenotype associations in HCM to derive a data-driven taxonomy of disease expression. METHODS We enrolled 436 patients with HCM (median age, 60 years; 28.8% women) with clinical, genetic, and imaging data. An independent cohort of 60 patients with HCM from Singapore (median age, 59 years; 11% women) and a reference population from the UK Biobank (n=16 691; mean age, 55 years; 52.5% women) were also recruited. We used machine learning to analyze the 3-dimensional structure of the left ventricle from cardiac magnetic resonance imaging and build a tree-based classification of HCM phenotypes. Genotype and mortality risk distributions were projected on the tree. RESULTS Carriers of pathogenic or likely pathogenic variants for HCM had lower left ventricular mass, but greater basal septal hypertrophy, with reduced life span (mean follow-up, 9.9 years) compared with genotype negative individuals (hazard ratio, 2.66 [95% CI, 1.42-4.96]; P<0.002). Four main phenotypic branches were identified using unsupervised learning of 3-dimensional shape: (1) nonsarcomeric hypertrophy with coexisting hypertension; (2) diffuse and basal asymmetrical hypertrophy associated with outflow tract obstruction; (3) isolated basal hypertrophy; and (4) milder nonobstructive hypertrophy enriched for familial sarcomeric HCM (odds ratio for pathogenic or likely pathogenic variants, 2.18 [95% CI, 1.93-2.28]; P=0.0001). Polygenic risk for HCM was also associated with different patterns and degrees of disease expression. The model was generalizable to an independent cohort (trustworthiness, M1: 0.86-0.88). CONCLUSIONS We report a data-driven taxonomy of HCM for identifying groups of patients with similar morphology while preserving a continuum of disease severity, genetic risk, and outcomes. This approach will be of value in understanding the causes and consequences of disease diversity.
Collapse
Affiliation(s)
- Lara Curran
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - Antonio de Marvao
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
- Department of Women and Children’s Health (A.d.M.)
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine and Sciences, King’s College London, United Kingdom (A.d.M.)
| | - Paolo Inglese
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Kathryn A. McGurk
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Pierre-Raphaël Schiratti
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Adam Clement
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Sean L. Zheng
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Surui Li
- Biomedical Image Analysis Group, Department of Computing (S.L., W.B.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Chee Jian Pua
- National Heart Research Institute Singapore, Singapore, PRC (C.J.P., C.W.L.C., S.A.C.)
| | - Mit Shah
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Mina Jafari
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Biomedical Image Analysis Group, Department of Computing (S.L., W.B.)
- Department of Brain Sciences, Imperial College London, London, United Kingdom (W.B.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
- Department of Women and Children’s Health (A.d.M.)
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine and Sciences, King’s College London, United Kingdom (A.d.M.)
- National Heart Research Institute Singapore, Singapore, PRC (C.J.P., C.W.L.C., S.A.C.)
- Department of Cardiology, National Heart Center Singapore, Singapore, PRC (C.W.L.C.)
- Cardiovascular Sciences ACP, Duke NUS Medical School, Singapore (C.W.L.C.)
- Mayo Clinic Rochester, MN (C.E.R.)
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (S.J.J., C.R.B.)
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.J.J.)
- Cardiovascular Genetics Centre, Montreal Heart Institute (R.T.)
- Faculty of Medicine, Université de Montréal, QC, Canada (R.T.)
- Radcliffe Department of Medicine, University of Oxford, United Kingdom (H.W.)
| | - Pantazis Theotokis
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Rachel J. Buchan
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Sean J. Jurgens
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (S.J.J., C.R.B.)
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA (S.J.J.)
| | - Claire E. Raphael
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Mayo Clinic Rochester, MN (C.E.R.)
| | - Arun John Baksi
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - Antonis Pantazis
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - Brian P. Halliday
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - Dudley J. Pennell
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing (S.L., W.B.)
- Department of Brain Sciences, Imperial College London, London, United Kingdom (W.B.)
| | - Calvin W.L. Chin
- National Heart Research Institute Singapore, Singapore, PRC (C.J.P., C.W.L.C., S.A.C.)
- Department of Cardiology, National Heart Center Singapore, Singapore, PRC (C.W.L.C.)
- Cardiovascular Sciences ACP, Duke NUS Medical School, Singapore (C.W.L.C.)
| | - Rafik Tadros
- Cardiovascular Genetics Centre, Montreal Heart Institute (R.T.)
- Faculty of Medicine, Université de Montréal, QC, Canada (R.T.)
| | - Connie R. Bezzina
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (S.J.J., C.R.B.)
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, United Kingdom (H.W.)
| | - Stuart A. Cook
- Department of Women and Children’s Health (A.d.M.)
- National Heart Research Institute Singapore, Singapore, PRC (C.J.P., C.W.L.C., S.A.C.)
| | - Sanjay K. Prasad
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
| | - James S. Ware
- National Heart and Lung Institute (L.C., K.A.M., S.L.Z., P.T., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust (L.C., R.J.B., C.E.R., A.J.B., A.P., B.P.H., D.J.P., S.K.P., J.S.W.)
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| | - Declan P. O’Regan
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, United Kingdom (A.d.M., P.I., K.A.M., P.-R.S., A.C., S.L.Z., S.L., M.S., M.J., P.T., R.J.B., S.A.C., J.S.W., D.P.O.)
| |
Collapse
|
25
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
26
|
Zhang B, Wu Y, Yang X, Xiang Y, Yang B. Molecular insight into arrhythmogenic cardiomyopathy caused by DSG2 mutations. Biomed Pharmacother 2023; 167:115448. [PMID: 37696084 DOI: 10.1016/j.biopha.2023.115448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Mutant desmoglein 2 (DSG2) is the second most common pathogenic gene in arrhythmogenic cardiomyopathy (ACM), accounting for approximately 10% of ACM cases. In addition to common clinical and pathological features, ACM caused by mutant DSG2 has specific characteristics, manifesting as left ventricle involvement and a high risk of heart failure. Pathological studies have shown extensive cardiomyocyte necrosis, infiltration of immune cells, and fibrofatty replacement in both ventricles, as well as abnormal desmosome structures in the hearts of humans and mice with mutant DSG2-related ACM. Although desmosome dysfunction is a common pathway in the pathogenesis of mutant DSG2-related ACM, the mechanisms underlying this dysfunction vary among mutations. Desmosome dysfunction induces cardiomyocyte injury, plakoglobin dislocation, and gap junction dysfunction, all of which contribute to the initiation and progression of ACM. Additionally, dysregulated inflammation, overactivation of transforming growth factor-beta-1 signaling and endoplasmic reticulum stress, and cardiac metabolic dysfunction contribute to the pathogenesis of ACM caused by mutant DSG2. These features demonstrate that patients with mutant DSG2-related ACM should be managed individually and precisely based on the genotype and phenotype. Further studies are needed to investigate the underlying mechanisms and to identify novel therapies to reverse or attenuate the progression of ACM caused by mutant DSG2.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| |
Collapse
|
27
|
Schröder LC, Frank D, Müller OJ. Transcriptional Targeting Approaches in Cardiac Gene Transfer Using AAV Vectors. Pathogens 2023; 12:1301. [PMID: 38003766 PMCID: PMC10675517 DOI: 10.3390/pathogens12111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiac-targeted transgene delivery offers new treatment opportunities for cardiovascular diseases, which massively contribute to global mortality. Restricted gene transfer to cardiac tissue might protect extracardiac organs from potential side-effects. This could be mediated by using cis-regulatory elements, including promoters and enhancers that act on the transcriptional level. Here, we discuss examples of tissue-specific promoters for targeted transcription in myocytes, cardiomyocytes, and chamber-specific cardiomyocytes. Some promotors are induced at pathological states, suggesting a potential use as "induction-by-disease switches" in gene therapy. Recent developments have resulted in the identification of novel enhancer-elements that could further pave the way for future refinement of transcriptional targeting, for example, into the cardiac conduction system.
Collapse
Affiliation(s)
- Lena C. Schröder
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (L.C.S.); (D.F.)
| | - Derk Frank
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (L.C.S.); (D.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany; (L.C.S.); (D.F.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| |
Collapse
|
28
|
Longoni M, Bhasin K, Ward A, Lee D, Nisson M, Bhatt S, Rodriguez F, Dash R. Real-world utilization of guideline-directed genetic testing in inherited cardiovascular diseases. Front Cardiovasc Med 2023; 10:1272433. [PMID: 37915745 PMCID: PMC10616303 DOI: 10.3389/fcvm.2023.1272433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Background Cardiovascular disease continues to be the leading cause of death globally. Clinical practice guidelines aimed at improving disease management and positively impacting major cardiac adverse events recommend genetic testing for inherited cardiovascular conditions such as dilated cardiomyopathy (DCM), hypertrophic cardiomyopathy (HCM), long QT syndrome (LQTS), hereditary amyloidosis, and familial hypercholesterolemia (FH); however, little is known about how consistently practitioners order genetic testing for these conditions in routine clinical practice. This study aimed to assess the adoption of guideline-directed genetic testing for patients diagnosed with DCM, HCM, LQTS, hereditary amyloidosis, or FH. Methods This retrospective cohort study captured real-world evidence of genetic testing from ICD-9-CM and ICD-10-CM codes, procedure codes, and structured text fields of de-identified patient records in the Veradigm Health Insights Ambulatory EHR Research Database linked with insurance claims data. Data analysis was conducted using an automated electronic health record analysis engine. Patient records in the Veradigm database were sourced from more than 250,000 clinicians serving over 170 million patients in outpatient primary care and specialty practice settings in the United States and linked insurance claims data from public and private insurance providers. The primary outcome measure was evidence of genetic testing within six months of condition diagnosis. Results Between January 1, 2017, and December 31, 2021, 224,641 patients were newly diagnosed with DCM, HCM, LQTS, hereditary amyloidosis, or FH and included in this study. Substantial genetic testing care gaps were identified. Only a small percentage of patients newly diagnosed with DCM (827/101,919; 0.8%), HCM (253/15,507; 1.6%), LQTS (650/56,539; 1.2%), hereditary amyloidosis (62/1,026; 6.0%), or FH (718/49,650; 1.5%) received genetic testing. Conclusions Genetic testing is underutilized across multiple inherited cardiovascular conditions. This real-world data analysis provides insights into the delivery of genomic healthcare in the United States and suggests genetic testing guidelines are rarely followed in practice.
Collapse
Affiliation(s)
- Mauro Longoni
- Global Medical Affairs Organization, Illumina, Inc., San Diego, CA, United States
| | | | | | | | | | - Sucheta Bhatt
- Global Medical Affairs Organization, Illumina, Inc., San Diego, CA, United States
| | - Fatima Rodriguez
- HealthPals Inc., Redwood, CA, United States
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Rajesh Dash
- HealthPals Inc., Redwood, CA, United States
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
29
|
Heymans S, Lakdawala NK, Tschöpe C, Klingel K. Dilated cardiomyopathy: causes, mechanisms, and current and future treatment approaches. Lancet 2023; 402:998-1011. [PMID: 37716772 DOI: 10.1016/s0140-6736(23)01241-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 09/18/2023]
Abstract
Dilated cardiomyopathy is conventionally defined as the presence of left ventricular or biventricular dilatation or systolic dysfunction in the absence of abnormal loading conditions (eg, primary valve disease) or significant coronary artery disease sufficient to cause ventricular remodelling. This definition has been recognised as overly restrictive, as left ventricular hypokinesis without dilation could be the initial presentation of dilated cardiomyopathy. The causes of dilated cardiomyopathy comprise genetic (primary dilated cardiomyopathy) or acquired factors (secondary dilated cardiomyopathy). Acquired factors include infections, toxins, cancer treatment, endocrinopathies, pregnancy, tachyarrhythmias, and immune-mediated diseases. 5-15% of patients with acquired dilated cardiomyopathy harbour a likely pathogenic or pathogenic gene variant (ie, gene mutation). Therefore, the diagnostic tests and therapeutic approach should always consider both genetic and acquired factors. This Seminar will focus on the current multidimensional diagnostic and therapeutic approach and discuss the underlying pathophysiology that could drive future treatments aiming to repair or replace the existing gene mutation, or target the specific inflammatory, metabolic, or pro-fibrotic drivers of genetic or acquired dilated cardiomyopathy.
Collapse
Affiliation(s)
- Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht, University of Maastricht & Maastricht University Medical Centre, Maastricht, Netherlands; Department of Cardiovascular Sciences, Centre for Vascular and Molecular Biology, KU Leuven, Leuven, Belgium
| | - Neal K Lakdawala
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carsten Tschöpe
- Department of Cardiology, Angiology, and Intensive Medicine (CVK), German Heart Center of the Charité (DHZC), Charité Universitätsmedizin, Berlin, Germany; Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
30
|
Chiswell K, Zaininger L, Semsarian C. Evolution of genetic testing and gene therapy in hypertrophic cardiomyopathy. Prog Cardiovasc Dis 2023; 80:38-45. [PMID: 37137376 DOI: 10.1016/j.pcad.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023]
Abstract
Studies over the last 30 years have identified hypertrophic cardiomyopathy (HCM) as predominantly an autosomal dominant disorder caused by disease-causing variants in genes encoding the sarcomere proteins critical for contractile function. The two most common disease genes implicated are the MYBPC3 and MYH7 genes, with disease-causing variants in these two genes accounting for 70-80% of all genotype-positive HCM patients. This increased knowledge of the genetic basis of HCM has heralded the era of precision medicine, with genetic testing leading to more improved and precise diagnosis, effective cascade genetic testing in at-risk family members, assistance with reproductive decisions, targeted therapeutics guided by both phenotype and genotype, and providing important insights into risk stratification and prognosis. Most recently, novel insights into genetic mechanisms have been elucidated, spanning non-Mendelian aetiologies, non-familial forms of HCM, and development of polygenic risk scores. These advances have laid the platform for exciting future endeavours such as newer gene therapy approaches in HCM, including gene replacement studies and genome editing approaches to ultimately cure disease. This brief review summarises the current role of genetic testing in HCM patients and families, and introduces some new mechanistic insights leading to the consideration of gene therapy approaches for HCM.
Collapse
Affiliation(s)
- Katherine Chiswell
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Louisa Zaininger
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia; Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia
| | - Christopher Semsarian
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia; Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
31
|
Brown EE, Murray B. A Practical Guide to Genetic Testing in Inherited Heart Disease. Card Electrophysiol Clin 2023; 15:241-247. [PMID: 37558295 DOI: 10.1016/j.ccep.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Genetic testing has increasingly been shown to provide critical information regarding the treatment and management of patients with hereditary cardiomyopathies and arrhythmias and is available for a wide variety of conditions. It can provide information regarding arrhythmia risk, lifestyle recommendations, such as exercise avoidance, pharmaceutical therapies, and prognosis. Beyond the proband, genetic testing can be a valuable tool for cascade screening in the family. Genetic testing should be accompanied with genetic counseling, as genetic tests should be accompanied by expert interpretation, support in cascade family evaluation, and psychosocial considerations. Overall, it should be routinely implemented in arrhythmia and cardiomyopathy clinics.
Collapse
Affiliation(s)
- Emily E Brown
- Division of Cardiology, Johns Hopkins University, 600 North Wolfe Street, Blalock 572, Baltimore, MD 21287, USA.
| | - Brittney Murray
- Division of Cardiology, Johns Hopkins University, 600 North Wolfe Street, Blalock 572, Baltimore, MD 21287, USA
| |
Collapse
|
32
|
Tikunova SB, Thuma J, Davis JP. Mouse Models of Cardiomyopathies Caused by Mutations in Troponin C. Int J Mol Sci 2023; 24:12349. [PMID: 37569724 PMCID: PMC10419064 DOI: 10.3390/ijms241512349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiac muscle contraction is regulated via Ca2+ exchange with the hetero-trimeric troponin complex located on the thin filament. Binding of Ca2+ to cardiac troponin C, a Ca2+ sensing subunit within the troponin complex, results in a series of conformational re-arrangements among the thin filament components, leading to an increase in the formation of actomyosin cross-bridges and muscle contraction. Ultimately, a decline in intracellular Ca2+ leads to the dissociation of Ca2+ from troponin C, inhibiting cross-bridge cycling and initiating muscle relaxation. Therefore, troponin C plays a crucial role in the regulation of cardiac muscle contraction and relaxation. Naturally occurring and engineered mutations in troponin C can lead to altered interactions among components of the thin filament and to aberrant Ca2+ binding and exchange with the thin filament. Mutations in troponin C have been associated with various forms of cardiac disease, including hypertrophic, restrictive, dilated, and left ventricular noncompaction cardiomyopathies. Despite progress made to date, more information from human studies, biophysical characterizations, and animal models is required for a clearer understanding of disease drivers that lead to cardiomyopathies. The unique use of engineered cardiac troponin C with the L48Q mutation that had been thoroughly characterized and genetically introduced into mouse myocardium clearly demonstrates that Ca2+ sensitization in and of itself should not necessarily be considered a disease driver. This opens the door for small molecule and protein engineering strategies to help boost impaired systolic function. On the other hand, the engineered troponin C mutants (I61Q and D73N), genetically introduced into mouse myocardium, demonstrate that Ca2+ desensitization under basal conditions may be a driving factor for dilated cardiomyopathy. In addition to enhancing our knowledge of molecular mechanisms that trigger hypertrophy, dilation, morbidity, and mortality, these cardiomyopathy mouse models could be used to test novel treatment strategies for cardiovascular diseases. In this review, we will discuss (1) the various ways mutations in cardiac troponin C might lead to disease; (2) relevant data on mutations in cardiac troponin C linked to human disease, and (3) all currently existing mouse models containing cardiac troponin C mutations (disease-associated and engineered).
Collapse
Affiliation(s)
- Svetlana B. Tikunova
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA (J.P.D.)
| | | | | |
Collapse
|
33
|
Olivotto I, Udelson JE, Pieroni M, Rapezzi C. Genetic causes of heart failure with preserved ejection fraction: emerging pharmacological treatments. Eur Heart J 2023; 44:656-667. [PMID: 36582184 DOI: 10.1093/eurheartj/ehac764] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/13/2022] [Accepted: 11/26/2022] [Indexed: 12/31/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major driver of cardiac morbidity and mortality in developed countries, due to ageing populations and the increasing prevalence of comorbidities. While heart failure with reduced ejection fraction is dominated by left ventricular impairment, HFpEF results from a complex interplay of cardiac remodelling, peripheral circulation, and concomitant features including age, hypertension, obesity, and diabetes. In an important subset, however, HFpEF is subtended by specific diseases of the myocardium that are genetically determined, have distinct pathophysiology, and are increasingly amenable to targeted, innovative treatments. While each of these conditions is rare, they collectively represent a relevant subset within HFpEF cohorts, and their prompt recognition has major consequences for clinical practice, as access to dedicated, disease-specific treatments may radically change the quality of life and outcome. Furthermore, response to standard heart failure treatment will generally be modest for these individuals, whose inclusion in registries and trials may dilute the perceived efficacy of treatments targeting mainstream HFpEF. Finally, a better understanding of the molecular underpinnings of monogenic myocardial disease may help identify therapeutic targets and develop innovative treatments for selected HFpEF phenotypes of broader epidemiological relevance. The field of genetic cardiomyopathies is undergoing rapid transformation due to recent, groundbreaking advances in drug development, and deserves greater awareness within the heart failure community. The present review addressed existing and developing therapies for genetic causes of HFpEF, including hypertrophic cardiomyopathy, cardiac amyloidosis, and storage diseases, discussing their potential impact on management and their broader implications for our understanding of HFpEF at large.
Collapse
Affiliation(s)
- Iacopo Olivotto
- Department of Experimental and Clinical Medicine, University of Florence, Meyer University Children Hospital and Careggi University Hospital, Viale Pieraccini 24, 50139 Florence, Italy
| | - James E Udelson
- Division of Cardiology and The CardioVascular Center, Tufts Medical Center, and the Tufts University School of Medicine, 800 Washington St, Boston, MA 02111, USA
| | - Maurizio Pieroni
- Cardiology Department, Hospital San Donato, Via Pietro Nenni, 20 - 52100 Arezzo, Italy
| | - Claudio Rapezzi
- Cardiology Centre, University of Ferrara, Via Fossato di Mortara, 64/B - 44121 Ferrara, Italy.,Maria Cecilia Hospital, GVM Care & Research, Via Corriera, 1, 48033 Cotignola, Emilia-Romagna, Italy
| |
Collapse
|
34
|
Dominguez F, Cabrera E. Mavacamten in obstructive hypertrophic cardiomyopathy - Are beta-blockers blocking part of its shine? Eur J Heart Fail 2023; 25:271-273. [PMID: 36597820 DOI: 10.1002/ejhf.2768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Affiliation(s)
- Fernando Dominguez
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Eva Cabrera
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Madrid, Madrid, Spain
| |
Collapse
|
35
|
Chumakova OS, Milovanova NV, Bychkov IO, Zakharova EY, Mershina EA, Sinitsin VE, Zateyshchikov DA. Overlapping Phenotype of Adult-Onset ALPK3-Cardiomyopathy in the Setting of Two Novel Variants. Cardiol Res 2022; 13:398-404. [PMID: 36660067 PMCID: PMC9822668 DOI: 10.14740/cr1449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Inherited cardiomyopathies (CMPs) are fairly common causes of morbidity and mortality, particularly, in young individuals. In substantial number of cases, only morphological diagnostic criteria cannot distinguish one CMP from another because of incomplete penetrance, advanced stage of the disease, or overlapping phenotypes. Genetic testing has become a mandatory tool for definite diagnosis that is required for family screening, individual prognosis, and personalized treatment strategy in routine practice. In parallel, accumulation of genotype-phenotype correlations, especially for rare genes, promotes the deciphering of underling molecular mechanisms and the development of targeting treatment of CMPs. Here we present an adult-onset case comprised morphological features of several CMPs: asymmetric left ventricle (LV) hypertrophy, severe systolic dysfunction, LV hypertrabeculation and restrictive physiology. Using next-generation sequencing, two novel variants (NM_020778.5:c.1958C>G:p.Ser653* and c.3491G>A:p.Arg1164Gln) in alpha-protein kinase 3 (ALPK3) gene were identified and confirmed with Sanger sequencing. The trans-position (location on different alleles) of identified ALPK3 variants was established by plasmid cloning method. The ALPK3 gene, encoding nuclear alpha-protein kinase 3, has only recently been associated with CMPs and there are still few clinical data on ALPK3 variant carriers. To date, only five affected individuals with adult-onset CMPs in the setting of biallelic variants of ALPK3 gene have been reported.
Collapse
Affiliation(s)
- Olga S. Chumakova
- Moscow Healthcare Department, City Clinical Hospital 17, 119620 Moscow, Russia
- E.I. Chazov National Medical Research Center for Cardiology, 121552 Moscow, Russia
| | | | | | | | - Elena A. Mershina
- Medical Research and Educational Center, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Valentin E. Sinitsin
- Medical Research and Educational Center, Lomonosov Moscow State University, 119991 Moscow, Russia
| | | |
Collapse
|