1
|
Nappi F. Myocarditis and Inflammatory Cardiomyopathy in Dilated Heart Failure. Viruses 2025; 17:484. [PMID: 40284927 PMCID: PMC12031395 DOI: 10.3390/v17040484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory cardiomyopathy is a condition that is characterised by the presence of inflammatory cells in the myocardium, which can lead to a significant deterioration in cardiac function. The etiology of this condition involves multiple factors, both infectious and non-infectious causes. While it is primarily associated with viral infections, other potential causes include bacterial, protozoal, or fungal infections, as well as a wide variety of toxic substances and drugs, and systemic immune-mediated pathological conditions. In spite of comprehensive investigation, the presence of inflammatory cardiomyopathy accompanied by left ventricular dysfunction, heart failure or arrhythmia is indicative of an unfavourable outcome. The reasons for the occurrence of either favourable outcomes, characterised by the absence of residual myocardial injury, or unfavourable outcomes, marked by the development of dilated cardiomyopathy, in patients afflicted by the condition remain to be elucidated. The relative contributions of pathogenic agents, genomic profiles of the host, and environmental factors in disease progression and resolution remain subjects of ongoing discourse. This includes the determination of which viruses function as active inducers and which merely play a bystander role. It remains unknown which changes in the host immune profile are critical in determining the outcome of myocarditis caused by various viruses, including coxsackievirus B3 (CVB3), adenoviruses, parvoviruses B19 and SARS-CoV-2. The objective of this review is unambiguous: to provide a concise summary and comprehensive assessment of the extant evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy. Its focus is exclusively on virus-induced and virus-associated myocarditis. In addition, the extant lacunae of knowledge in this field are identified and the extant experimental models are evaluated, with the aim of proposing future directions for the research domain. This includes differential gene expression that regulates iron and lipid and metabolic remodelling. Furthermore, the current state of knowledge regarding the cardiovascular implications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is also discussed, along with the open questions that remain to be addressed.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
2
|
Drazner MH, Bozkurt B, Cooper LT, Aggarwal NR, Basso C, Bhave NM, Caforio ALP, Ferreira VM, Heidecker B, Kontorovich AR, Martín P, Roth GA, Van Eyk JE. 2024 ACC Expert Consensus Decision Pathway on Strategies and Criteria for the Diagnosis and Management of Myocarditis: A Report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2025; 85:391-431. [PMID: 39665703 DOI: 10.1016/j.jacc.2024.10.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
|
3
|
Vicenzetto C, Giordani AS, Menghi C, Baritussio A, Scognamiglio F, Pontara E, Bison E, Peloso-Cattini MG, Marcolongo R, Caforio ALP. Cellular Immunology of Myocarditis: Lights and Shades-A Literature Review. Cells 2024; 13:2082. [PMID: 39768171 PMCID: PMC11674465 DOI: 10.3390/cells13242082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium with heterogeneous etiology, clinical presentation, and prognosis; when it is associated with myocardial dysfunction, this identifies the entity of inflammatory cardiomyopathy. In the last few decades, the relevance of the immune system in myocarditis onset and progression has become evident, thus having crucial clinical relevance in terms of treatment and prognostic stratification. In fact, the advances in cardiac immunology have led to a better characterization of the cellular subtypes involved in the pathogenesis of inflammatory cardiomyopathy, whether the etiology is infectious or autoimmune/immune-mediated. The difference in the clinical course between spontaneous recovery to acute, subacute, or chronic progression to end-stage heart failure may be explained not only by classical prognostic markers but also through immune-pathological mechanisms at a cellular level. Nevertheless, much still needs to be clarified in terms of immune characterization and molecular mechanisms especially in biopsy-proven myocarditis. The aims of this review are to (1) describe inflammatory cardiomyopathy etiology, especially immune-mediated/autoimmune forms, (2) analyze recent findings on the role of different immune cells subtypes in myocarditis, (3) illustrate the potential clinical relevance of such findings, and (4) highlight the need of further studies in pivotal areas of myocarditis cellular immunology.
Collapse
Affiliation(s)
- Cristina Vicenzetto
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Caterina Menghi
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anna Baritussio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Federico Scognamiglio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elena Pontara
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elisa Bison
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Maria Grazia Peloso-Cattini
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Renzo Marcolongo
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Cardioimmunology Laboratory, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
4
|
Giordani AS, Pontara E, Vicenzetto C, Baritussio A, Peloso Cattini MG, Bison E, Re F, Marcolongo R, Joseph S, Chatterjee D, Fatah M, Hamilton RM, Caforio ALP. Prevalence and Correlates of Anti-DSG2 Antibodies in Arrhythmogenic Right Ventricular Cardiomyopathy and Myocarditis: Immunological Insights from a Multicenter Study. J Clin Med 2024; 13:6736. [PMID: 39597880 PMCID: PMC11594951 DOI: 10.3390/jcm13226736] [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: 10/21/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Autoantibodies against Desmoglein-2 desmosomal protein (anti-DSG2-ab) were identified in Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) by Enzyme-Linked ImmunoSorbent Assay (ELISA); anti-intercalated disk autoantibodies (AIDAs) were identified in myocarditis and (ARVC) by indirect immunofluorescence (IFL). We aim to assess: (1) anti-DSG2-ab specificity in ARVC and myocarditis, (2) accuracy of anti-DSG2-ab detection by ELISA versus AIDA by IFL, and (3) clinical correlates of anti-DSG2-ab in ARVC. Methods: We included 77 patients with ARVC, 91 with myocarditis/dilated cardiomyopathy (DCM), 27 with systemic immune-mediated diseases, and 50 controls. Anti-heart antibodies (AHAs) and AIDAs were assessed by IFL, and anti-DSG2-ab by ELISA (assessed both by optical density, OD, and U/L). Receiving operator curve (ROC) analysis was used to assess ELISA diagnostic accuracy. Results: A relevant proportion (56%) of ARVC patients was anti-DSG2-ab-positive, with higher anti-DSG2-ab levels than controls. Anti-DSG2-ab titer was not different between ARVC and myocarditis/DCM patients (48% anti-DSG-ab positive). Frequency of anti-DSG2 positivity by ELISA was higher in AIDA-positive cases by IFL than AIDA-negative cases (p = 0.039 for OD, p = 0.023 for U/L). In ARVC, AIDA-positive patients were more likely to be AHA-positive (p < 0.001), had pre-syncope (p = 0.025), and abnormalities in cardiac rhythm (p = 0.03) than ARVC AIDA-negative patients, while anti-DSG2-ab positivity did not have clinical correlates. Conclusions: Anti-DG2-ab detection in ARVC and myocarditis/DCM reflects immune-mediated pathogenesis to desmosomal proteins. Higher frequency of anti-DSG2-ab positivity by ELISA by U/L was higher in AIDA-positive cases by IFL than AIDA-negative cases, in keeping with the hypothesis that DSG2 is one of AIDA autoantigens. In ARVC, AIDA status but not anti-DSG2-ab showed distinct clinical correlates, possibly reflecting a wider AIDA autoantigenic spectrum.
Collapse
Affiliation(s)
- Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elena Pontara
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Cristina Vicenzetto
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anna Baritussio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Maria Grazia Peloso Cattini
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Elisa Bison
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Federica Re
- Cardiology Division, San Camillo Hospital, 00152 Rome, Italy
| | - Renzo Marcolongo
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Shaylyn Joseph
- Department of Pediatrics, The Labatt Family Heart Centre and Translational Medicine, The Hospital for Sick Children & Research Institute, The University of Toronto, Room 1725D, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Diptendu Chatterjee
- Department of Pediatrics, The Labatt Family Heart Centre and Translational Medicine, The Hospital for Sick Children & Research Institute, The University of Toronto, Room 1725D, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Meena Fatah
- Department of Pediatrics, The Labatt Family Heart Centre and Translational Medicine, The Hospital for Sick Children & Research Institute, The University of Toronto, Room 1725D, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Robert M. Hamilton
- Department of Pediatrics, The Labatt Family Heart Centre and Translational Medicine, The Hospital for Sick Children & Research Institute, The University of Toronto, Room 1725D, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
5
|
Rolfs N, Huber C, Opgen-Rhein B, Altmann I, Anderheiden F, Hecht T, Fischer M, Wiegand G, Reineker K, Voges I, Kiski D, Frede W, Boehne M, Khedim M, Messroghli D, Klingel K, Schwarzkopf E, Pickardt T, Schubert S, Lunze FI, Seidel F. Prognostic Value of Speckle Tracking Echocardiography-Derived Strain in Unmasking Risk for Arrhythmias in Children with Myocarditis. Biomedicines 2024; 12:2369. [PMID: 39457681 PMCID: PMC11505463 DOI: 10.3390/biomedicines12102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Risk assessment in pediatric myocarditis is challenging, particularly when left ventricular ejection fraction (LVEF) is preserved. This study aimed to evaluate LV myocardial deformation using speckle-tracking echocardiography (STE)-derived longitudinal +strain (LS) and assessed its diagnostic and prognostic value in children with myocarditis. Methods: Retrospective STE-derived layer-specific LV LS analysis was performed on echocardiograms from patients within the multicenter, prospective registry for pediatric myocarditis "MYKKE". Age- and sex-adjusted logistic regression and ROC analysis identified predictors of cardiac arrhythmias (ventricular tachycardia, ventricular fibrillation, atrioventricular blockage III°) and major adverse cardiac events (MACE: need for mechanical circulatory support (MCS), cardiac transplantation, and/or cardiac death). Results: Echocardiograms from 175 patients (median age 15 years, IQR 7.9-16.5 years; 70% male) across 13 centers were included. Cardiac arrhythmias occurred in 36 patients (21%), and MACE in 28 patients (16%). Impaired LV LS strongly correlated with reduced LVEF (r > 0.8). Impaired layer-specific LV LS, reduced LVEF, LV dilatation, and increased BSA-indexed LV mass, were associated with the occurrence of MACE and cardiac arrhythmias. In patients with preserved LVEF, LV LS alone predicted cardiac arrhythmias (p < 0.001), with optimal cutoff values of -18.0% for endocardial LV LS (sensitivity 0.69, specificity 0.94) and -17.0% for midmyocardial LV LS (sensitivity 0.81, specificity 0.75). Conclusions: In pediatric myocarditis, STE-derived LV LS is not only a valuable tool for assessing systolic myocardial dysfunction and predicting MACE but also identifies patients at risk for cardiac arrhythmias, even in the context of preserved LVEF.
Collapse
Affiliation(s)
- Nele Rolfs
- Department of Congenital Heart Disease—Pediatric Cardiology; Deutsches Herzzentrum der Charité, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Cynthia Huber
- Department of Medical Statistics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Bernd Opgen-Rhein
- Department of Congenital Heart Disease—Pediatric Cardiology; Deutsches Herzzentrum der Charité, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Isabell Altmann
- Clinic for Pediatric Cardiology, Heart Centre, University of Leipzig, 04109 Leipzig, Germany
| | - Felix Anderheiden
- Pediatric Cardiology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Tobias Hecht
- Center for Congenital Heart Disease, Heart and Diabetes Center NRW, University Hospital of the Ruhr University Bochum, Medical Faculty OWL (University of Bielefeld), 32345 Bad Oeynhausen, Germany
| | - Marcus Fischer
- Department of Pediatric Cardiology and Pediatric Intensive Care, Ludwig Maximilians University of Munich, 80336 Munich, Germany
| | - Gesa Wiegand
- Pediatric Cardiology, University Hospital Tübingen, 72076 Tuebingen, Germany
| | - Katja Reineker
- Department of Congenital Heart Disease and Pediatric Cardiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Inga Voges
- Department for Congenital Heart Disease and Pediatric Cardiology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 24105 Kiel, Germany
| | - Daniela Kiski
- Pediatric Cardiology, University Hospital Münster, 48149 Muenster, Germany
| | - Wiebke Frede
- Pediatric Cardiology and Congenital Heart Defects, Center for Pediatrics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Martin Boehne
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Malika Khedim
- Pediatric Cardiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Daniel Messroghli
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 10117 Berlin, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Eicke Schwarzkopf
- Department of Congenital Heart Disease—Pediatric Cardiology; Deutsches Herzzentrum der Charité, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Thomas Pickardt
- Competence Network for Congenital Heart Defects, 13353 Berlin, Germany
| | - Stephan Schubert
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Center for Congenital Heart Disease, Heart and Diabetes Center NRW, University Hospital of the Ruhr University Bochum, Medical Faculty OWL (University of Bielefeld), 32345 Bad Oeynhausen, Germany
| | - Fatima I. Lunze
- Department of Congenital Heart Disease—Pediatric Cardiology; Deutsches Herzzentrum der Charité, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Franziska Seidel
- Department of Congenital Heart Disease—Pediatric Cardiology; Deutsches Herzzentrum der Charité, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| |
Collapse
|
6
|
Frustaci A, Letizia C, Alfarano M, Marchionni G, Verardo R, Chimenti C. Immunomodulating and Immunosuppressive Therapy for Virus-Negative Immune-Mediated Myocarditis. Biomedicines 2024; 12:1565. [PMID: 39062138 PMCID: PMC11274480 DOI: 10.3390/biomedicines12071565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium caused by infectious and noninfectious agents. Clinical manifestations range from mildly symptomatic forms to acute heart failure, cardiogenic shock, life-threatening arrhythmias and sudden death. Myocarditis is still a challenging diagnosis because of its wide variability in clinical presentation and unpredictable course. Moreover, a standardized, specific treatment in not yet available. Immunosuppressive treatment for virus-negative lymphocytic myocarditis is still controversial. Conversely, immunosuppression is well established in sarcoidosis, eosinophilic, giant-cell, drug hypersensitivity, and trauma-related myocarditis as well as lymphocytic myocarditis associated with connective tissue diseases or with the rejection of a transplanted heart. Recently, immunosuppressive therapy has been also recognized as an effective treatment in virus-negative inflammatory cardiomyopathy. The aim of this review is to underline the role of immunomodulating and immunosuppressive therapies in patients with immune-mediated myocarditis and illustrate the different treatment strategies depending on the etiology. An endomyocardial biopsy remains the gold standard for the diagnosis of myocarditis as well as for a tailored treatment.
Collapse
Affiliation(s)
- Andrea Frustaci
- Cellular and Molecular Cardiology Laboratory, IRCCS Lazzaro Spallanzani, 00149 Rome, Italy;
- IRCCS San Raffaele, 00163 Rome, Italy
| | - Claudio Letizia
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| | - Maria Alfarano
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| | - Giulia Marchionni
- Policlinico San Matteo Pavia IRCCS Foundation, University of Pavia, 27100 Pavia, Italy;
| | - Romina Verardo
- Cellular and Molecular Cardiology Laboratory, IRCCS Lazzaro Spallanzani, 00149 Rome, Italy;
| | - Cristina Chimenti
- Department of Clinical, Internal, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.L.); (M.A.); (C.C.)
| |
Collapse
|
7
|
Vicenzetto C, Giordani AS, Menghi C, Baritussio A, Peloso Cattini MG, Pontara E, Bison E, Rizzo S, De Gaspari M, Basso C, Thiene G, Iliceto S, Marcolongo R, Caforio ALP. The Role of the Immune System in Pathobiology and Therapy of Myocarditis: A Review. Biomedicines 2024; 12:1156. [PMID: 38927363 PMCID: PMC11200507 DOI: 10.3390/biomedicines12061156] [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: 03/29/2024] [Revised: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The role of the immune system in myocarditis onset and progression involves a range of complex cellular and molecular pathways. Both innate and adaptive immunity contribute to myocarditis pathogenesis, regardless of its infectious or non-infectious nature and across different histological and clinical subtypes. The heterogeneity of myocarditis etiologies and molecular effectors is one of the determinants of its clinical variability, manifesting as a spectrum of disease phenotype and progression. This spectrum ranges from a fulminant presentation with spontaneous recovery to a slowly progressing, refractory heart failure with ventricular dysfunction, to arrhythmic storm and sudden cardiac death. In this review, we first examine the updated definition and classification of myocarditis at clinical, biomolecular and histopathological levels. We then discuss recent insights on the role of specific immune cell populations in myocarditis pathogenesis, with particular emphasis on established or potential therapeutic applications. Besides the well-known immunosuppressive agents, whose efficacy has been already demonstrated in human clinical trials, we discuss the immunomodulatory effects of other drugs commonly used in clinical practice for myocarditis management. The immunological complexity of myocarditis, while presenting a challenge to simplistic understanding, also represents an opportunity for the development of different therapeutic approaches with promising results.
Collapse
Affiliation(s)
- Cristina Vicenzetto
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Andrea Silvio Giordani
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Caterina Menghi
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Anna Baritussio
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Maria Grazia Peloso Cattini
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Elena Pontara
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Elisa Bison
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Stefania Rizzo
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Monica De Gaspari
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Gaetano Thiene
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy (G.T.)
| | - Sabino Iliceto
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Renzo Marcolongo
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| | - Alida Linda Patrizia Caforio
- Cardiology and Cardioimmunology Laboratory, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (C.V.); (R.M.)
| |
Collapse
|
8
|
Scheel PJ, Cartella I, Murray B, Gilotra NA, Ammirati E. Role of genetics in inflammatory cardiomyopathy. Int J Cardiol 2024; 400:131777. [PMID: 38218248 DOI: 10.1016/j.ijcard.2024.131777] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Traditional cardiomyopathy paradigms segregate inflammatory etiologies from those caused by genetic variants. An identified or presumed trigger is implicated in acute myocarditis or chronic inflammatory cardiomyopathy but growing evidence suggests a significant proportion of patients have an underlying cardiomyopathy-associated genetic variant often even when a clear inflammatory trigger is identified. Recognizing a possible genetic contribution to inflammatory cardiomyopathy may have major downstream implications for both the patient and family. The presenting features of myocarditis (i.e. chest pain, arrhythmia, and/or heart failure) may provide insight into diagnostic considerations. One example is isolated cardiac sarcoidosis, a distinct inflammatory cardiomyopathy that carries diagnostic challenges and clinical overlap; genetic testing has increasingly reclassified cases of isolated cardiac sarcoidosis as genetic cardiomyopathy, notably altering management. On the other side, inflammatory presentations of genetic cardiomyopathies are likewise underappreciated and a growing area of investigation. Inflammation plays an important role in the pathogenesis of several familial cardiomyopathies, especially arrhythmogenic phenotypes. Given these clinical scenarios, and the implications on clinical decision making such as initiation of immunosuppression, sudden cardiac death prevention, and family screening, it is important to recognize when genetics may be playing a role.
Collapse
Affiliation(s)
- Paul J Scheel
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA.
| | - Iside Cartella
- De Gasperis Cardio Center, Transplant Center, Niguarda Hospital, Milano, Italy; Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA
| | - Nisha A Gilotra
- Division of Cardiology, Department of Medicine, Johns Hopkins University, USA
| | - Enrico Ammirati
- De Gasperis Cardio Center, Transplant Center, Niguarda Hospital, Milano, Italy; Department of Health Sciences, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
9
|
Weisleder H, Jacobson E, Frishman WH, Dhand A. Cardiac Manifestations of Viral Infections, Including COVID-19: A Review. Cardiol Rev 2024; 32:124-130. [PMID: 36730913 DOI: 10.1097/crd.0000000000000481] [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] [Indexed: 02/04/2023]
Abstract
Viral infections have been linked to a variety of cardiac pathology, which may include acute myocarditis, dilated cardiomyopathy, heart failure, cardiogenic shock, pericarditis, acute coronary syndromes, and arrhythmias. We performed a systematic review of literature focusing on the cardiovascular effects of various viral infections, as well as providing an update on the current understanding of the pathophysiology of Coronavirus disease-2019 (COVID-19). Cardiac manifestations of viral illnesses are usually self-limiting, have variable clinical presentations, and require sufficient clinical suspicion for diagnosis and optimal management.
Collapse
Affiliation(s)
| | | | | | - Abhay Dhand
- From the New York Medical College, Valhalla, NY
- Department of Medicine and Surgery, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
10
|
Musigk N, Suwalski P, Golpour A, Fairweather D, Klingel K, Martin P, Frustaci A, Cooper LT, Lüscher TF, Landmesser U, Heidecker B. The inflammatory spectrum of cardiomyopathies. Front Cardiovasc Med 2024; 11:1251780. [PMID: 38464847 PMCID: PMC10921946 DOI: 10.3389/fcvm.2024.1251780] [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: 07/02/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Infiltration of the myocardium with various cell types, cytokines and chemokines plays a crucial role in the pathogenesis of cardiomyopathies including inflammatory cardiomyopathies and myocarditis. A more comprehensive understanding of the precise immune mechanisms involved in acute and chronic myocarditis is essential to develop novel therapeutic approaches. This review offers a comprehensive overview of the current knowledge of the immune landscape in cardiomyopathies based on etiology. It identifies gaps in our knowledge about cardiac inflammation and emphasizes the need for new translational approaches to improve our understanding thus enabling development of novel early detection methods and more effective treatments.
Collapse
Affiliation(s)
- Nicolas Musigk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Phillip Suwalski
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Ainoosh Golpour
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Karin Klingel
- Cardiopathology Institute for Pathology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Pilar Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Thomas F. Lüscher
- GZO-Zurich Regional Health Centre, Wetzikon & Cardioimmunology, Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton & Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Bettina Heidecker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
11
|
Sur M, Rasquinha MT, Mone K, Massilamany C, Lasrado N, Gurumurthy C, Sobel RA, Reddy J. Investigation into Cardiac Myhc-α 334-352-Specific TCR Transgenic Mice Reveals a Role for Cytotoxic CD4 T Cells in the Development of Cardiac Autoimmunity. Cells 2024; 13:234. [PMID: 38334626 PMCID: PMC10854502 DOI: 10.3390/cells13030234] [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: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Myocarditis is one of the major causes of heart failure in children and young adults and can lead to dilated cardiomyopathy. Lymphocytic myocarditis could result from autoreactive CD4+ and CD8+ T cells, but defining antigen specificity in disease pathogenesis is challenging. To address this issue, we generated T cell receptor (TCR) transgenic (Tg) C57BL/6J mice specific to cardiac myosin heavy chain (Myhc)-α 334-352 and found that Myhc-α-specific TCRs were expressed in both CD4+ and CD8+ T cells. To investigate if the phenotype is more pronounced in a myocarditis-susceptible genetic background, we backcrossed with A/J mice. At the fourth generation of backcrossing, we observed that Tg T cells from naïve mice responded to Myhc-α 334-352, as evaluated by proliferation assay and carboxyfluorescein succinimidyl ester staining. The T cell responses included significant production of mainly pro-inflammatory cytokines, namely interferon (IFN)-γ, interleukin-17, and granulocyte macrophage-colony stimulating factor. While the naïve Tg mice had isolated myocardial lesions, immunization with Myhc-α 334-352 led to mild myocarditis, suggesting that further backcrossing to increase the percentage of A/J genome close to 99.99% might show a more severe disease phenotype. Further investigations led us to note that CD4+ T cells displayed the phenotype of cytotoxic T cells (CTLs) akin to those of conventional CD8+ CTLs, as determined by the expression of CD107a, IFN-γ, granzyme B natural killer cell receptor (NKG)2A, NKG2D, cytotoxic and regulatory T cell molecules, and eomesodermin. Taken together, the transgenic system described in this report may be a helpful tool to distinguish the roles of cytotoxic cardiac antigen-specific CD4+ T cells vs. those of CD8+ T cells in the pathogenesis of myocarditis.
Collapse
Affiliation(s)
- Meghna Sur
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
| | - Mahima T. Rasquinha
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
| | - Kiruthiga Mone
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
| | - Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
- CRISPR Therapeutics, Boston, MA 02127, USA
| | - Ninaad Lasrado
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
- Center for Virology and Vaccine Research, Harvard Medical School, Boston, MA 02115, USA
| | - Channabasavaiah Gurumurthy
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Raymond A. Sobel
- Department of Pathology, Stanford University, Stanford, CA 94305, USA;
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (M.S.); (M.T.R.); (K.M.); (C.M.); (N.L.)
| |
Collapse
|
12
|
Baumeier C, Harms D, Aleshcheva G, Gross U, Escher F, Schultheiss HP. Advancing Precision Medicine in Myocarditis: Current Status and Future Perspectives in Endomyocardial Biopsy-Based Diagnostics and Therapeutic Approaches. J Clin Med 2023; 12:5050. [PMID: 37568452 PMCID: PMC10419903 DOI: 10.3390/jcm12155050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The diagnosis and specific and causal treatment of myocarditis and inflammatory cardiomyopathy remain a major clinical challenge. Despite the rapid development of new imaging techniques, endomyocardial biopsies remain the gold standard for accurate diagnosis of inflammatory myocardial disease. With the introduction and continued development of immunohistochemical inflammation diagnostics in combination with viral nucleic acid testing, myocarditis diagnostics have improved significantly since their introduction. Together with new technologies such as miRNA and gene expression profiling, quantification of specific immune cell markers, and determination of viral activity, diagnostic accuracy and patient prognosis will continue to improve in the future. In this review, we summarize the current knowledge on the pathogenesis and diagnosis of myocarditis and inflammatory cardiomyopathies and highlight future perspectives for more in-depth and specialized biopsy diagnostics and precision, personalized medicine approaches.
Collapse
Affiliation(s)
- Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (D.H.); (G.A.); (U.G.); (H.-P.S.)
| | - Dominik Harms
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (D.H.); (G.A.); (U.G.); (H.-P.S.)
- Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany
| | - Ganna Aleshcheva
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (D.H.); (G.A.); (U.G.); (H.-P.S.)
| | - Ulrich Gross
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (D.H.); (G.A.); (U.G.); (H.-P.S.)
| | - Felicitas Escher
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Campus Virchow Klinikum, 13353 Berlin, Germany;
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Heinz-Peter Schultheiss
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (D.H.); (G.A.); (U.G.); (H.-P.S.)
| |
Collapse
|
13
|
Blagova OV, Alieva IN, Kulikova VA, Nedostup AV, Kogan EA, Sedov VP, Parfenov DA, Volovchenko AN, Sarkisova ND. [Long-term treatment of morphologically verified myocarditis: successes and probable errors. Case report]. TERAPEVT ARKH 2023; 95:327-334. [PMID: 38158981 DOI: 10.26442/00403660.2023.04.202156] [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: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 01/03/2024]
Abstract
Diagnosis and treatment of myocarditis can be challenging, including determining indications for heart transplantation. We present a 6-year medical history of a 54 years old patient with severe morphologically verified viral-negative lymphocytic myocarditis and systemic manifestations (onset of hemorrhagic vasculitis) combined with moderate coronary atherosclerosis, which regressed according to repeated coronary angiography. For 5 years, the patient received immunosuppressive therapy with methylprednisolone and azathioprine with a significant improvement. Repeated relapses of atrial fibrillation required correction of basic therapy and plasmapheresis. The disease was complicated by thyrotoxicosis and multi-organ dysfunction; the autopsy showed persistent myocarditis activity. The myocarditis is a chronic condition and requires a review of the treatment strategy at each stage.
Collapse
Affiliation(s)
- O V Blagova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - I N Alieva
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V A Kulikova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - A V Nedostup
- Sechenov First Moscow State Medical University (Sechenov University)
| | - E A Kogan
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V P Sedov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - D A Parfenov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - A N Volovchenko
- Sechenov First Moscow State Medical University (Sechenov University)
| | - N D Sarkisova
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
14
|
Paul T, Klingel K, Tschöpe C, Bertram H, Seidel F. Leitlinie Myokarditis der Deutschen Gesellschaft für
Pädiatrische Kardiologie. KLINISCHE PADIATRIE 2023; 235:e1-e15. [PMID: 37094605 PMCID: PMC10191740 DOI: 10.1055/a-2039-2604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
This consensus statement presents updated recommendations on diagnosis and treatment of myocarditis in childhood.
Collapse
Affiliation(s)
- Thomas Paul
- Universitätsmedizin Göttingen Klinik für
Pädiatrische Kardiologie und Intensivmedizin, Göttingen,
Deutschland
| | - Karin Klingel
- Universitätshospital Tübingen, Institut für
Pathologie und Neuropathologie, Tübingen, Deutschland
| | - Carsten Tschöpe
- Charité Universitätsmedizin Berlin, Kardiologie,
Berlin, Deutschland
| | - Harald Bertram
- Medizinische Hochschule Hannover, Klinik für
Pädiatrische Kardiologie und Pädiatrische Intensivmedizin,
Hannover, Deutschland
| | - Franziska Seidel
- Charité Universitätsmedizn Berlin, Pädiatrische
Kardiologie, Berlin, Deutschland
| |
Collapse
|
15
|
Grzechocińska J, Tymińska A, Giordani AS, Wysińska J, Ostrowska E, Baritussio A, Caforio ALP, Grabowski M, Marcolongo R, Ozierański K. Immunosuppressive Therapy of Biopsy-Proven, Virus-Negative, Autoimmune/Immune-Mediated Myocarditis-Focus on Azathioprine: A Review of Existing Evidence and Future Perspectives. BIOLOGY 2023; 12:356. [PMID: 36979048 PMCID: PMC10044979 DOI: 10.3390/biology12030356] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 02/26/2023]
Abstract
The use of immunosuppressive therapy (IT) in biopsy-proven, autoimmune/immune-mediated (AI), virus-negative myocarditis has become the standard of care. In particular, according to recent guidelines, azathioprine (AZA), in association with steroids, is a cornerstone of first-line therapy regimens. IT may have a crucial impact on the natural history of AI myocarditis, preventing its progression to end-stage heart failure, cardiovascular death, or heart transplantation, provided that strict appropriateness and safety criteria are observed. In particular, AZA treatment for AI virus-negative myocarditis requires the consideration of some crucial aspects regarding its pharmacokinetics and pharmacodynamics, as well as a high index of suspicion to detect its overt and/or subclinical side effects. Importantly, besides a tight teamwork with a clinical immunologist/immuno-rheumatologist, before starting IT, it is also necessary to carry out a careful "safety check-list" in order to rule out possible contraindications to IT and minimize patient's risk. The aim of this review is to describe the pharmacological properties of AZA, as well as to discuss practical aspects of its clinical use, in the light of existing evidence, with particular regard to the new field of cardioimmunology.
Collapse
Affiliation(s)
- Justyna Grzechocińska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| | - Agata Tymińska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| | - Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy
| | - Julia Wysińska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| | - Ewa Ostrowska
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| | - Anna Baritussio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| | - Renzo Marcolongo
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy
| | - Krzysztof Ozierański
- First Department of Cardiology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland
| |
Collapse
|
16
|
Frustaci A, Russo MA, Chimenti C. Individualized immunosuppression in virus-negative inflammatory cardiomyopathy. Eur Heart J 2022; 43:4760. [PMID: 36263787 DOI: 10.1093/eurheartj/ehac559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Andrea Frustaci
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, La Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy.,Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Rome, Italy
| | - Matteo A Russo
- MEBIC Consortium, San Raffaele Open University and IRCCS San Raffaele, 00166 Rome, Italy
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, La Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
17
|
Affiliation(s)
- Cristina Basso
- From the Cardiovascular Pathology Unit, Azienda Ospedaliera, Department of Cardiac, Thoracic, and Vascular Sciences and Public Health, University of Padua, Padua, Italy
| |
Collapse
|
18
|
Schultheiss HP, Escher F. Advanced diagnostics in inflammatory cardiomyopathy for personalized therapeutic decision-making. Eur Heart J 2022; 43:3474-3476. [PMID: 35920154 DOI: 10.1093/eurheartj/ehac412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Heinz-Peter Schultheiss
- Department of Cardiology Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Moltkestr. 31, D-12203, Berlin, Germany
| | - Felicitas Escher
- Department of Cardiology Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Moltkestr. 31, D-12203, Berlin, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité-Universitaetsmedizin Berlin, corporate member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
19
|
Chimenti C, Russo MA, Frustaci A. Immunosuppressive therapy in virus-negative inflammatory cardiomyopathy: 20-year follow-up of the TIMIC trial. Eur Heart J 2022; 43:3463-3473. [PMID: 35831932 PMCID: PMC9492235 DOI: 10.1093/eurheartj/ehac348] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/26/2022] [Accepted: 06/07/2022] [Indexed: 01/05/2023] Open
Abstract
AIMS Long-term results of the Tailored IMmunosuppression in virus-negative Inflammatory Cardiomyopathy (TIMIC) trial protocol have been evaluated. METHODS AND RESULTS Eighty-five patients with endomyocardial biopsy-proven virus-negative chronic inflammatory cardiomyopathy were enrolled in the randomized, double-blind, placebo-controlled TIMIC trial and received prednisone and azathioprine (n = 43) vs. placebo (n = 42) for 6 months. Immunosuppressive treatment promoted an improvement in cardiac function in 88% of the cases compared with none of the patients in the placebo group, which were switched to a 6-month immunosuppressive therapy at the end of the 6-month study period. Long-term (up to 20 years) clinical outcomes of the whole cohort of 85 patients originally enrolled in the TIMIC trial (Group A) were compared with those of a 1:2 propensity score-matched control cohort of patients untreated with the TIMIC protocol (Group B) and followed for a comparable period of time. The primary outcome was a composite of cardiovascular death and heart transplantation. At long-term follow-up, the risk of cardiovascular death [hazard ratio (HR) 6.77; 95% confidence interval (CI) 2.36-19.45] and heart transplantation (HR 7.92; 95% CI 1.80-34.88) was significantly higher in Group B patients. Group A showed a persistent improvement in the left ventricular ejection fraction compared with Group B (HR 7.24; 95% CI 3.05-17.18). A higher number of Group B patients underwent implantable cardioverter defibrillator implantation. The incidence of recurrent myocarditis was similar between groups, and patients with evidence of a recurrent cardiac inflammatory process promptly responded to a TIMIC protocol application. CONCLUSION Virus-negative inflammatory cardiomyopathy benefits from immunosuppressive therapy even after long-term follow-up. Recurrence appears to respond to a new TIMIC protocol application.
Collapse
Affiliation(s)
- Cristina Chimenti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy,Molecular and Cellular Cardiology Lab, IRCCS ‘L. Spallanzani’, Rome, Italy
| | | | | |
Collapse
|
20
|
Sozzi FB, Gherbesi E, Faggiano A, Gnan E, Maruccio A, Schiavone M, Iacuzio L, Carugo S. Viral Myocarditis: Classification, Diagnosis, and Clinical Implications. Front Cardiovasc Med 2022; 9:908663. [PMID: 35795363 PMCID: PMC9250986 DOI: 10.3389/fcvm.2022.908663] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Myocarditis is an inflammatory disease of the myocardium with focal or diffuse involvement. Viral infections are the most common cause of myocarditis, especially in Western countries. A recent viral illness with gastroenteric or upper respiratory symptoms often precedes myocarditis. The absence of specific pathognomonic features in conjunction with the wide spectrum of clinical manifestations that range from subclinical cases to sudden cardiac death (SCD) makes myocarditis diagnosis particularly challenging. Moreover, myocarditis might represent a cause of initially unexplained dilated cardiomyopathy (DCM) and heart failure (HF), especially among children and young adults. Cardiac magnetic resonance imaging (CMR) is crucial for myocarditis diagnosis, because of its ability to detect interstitial edema during acute inflammation. Assessment of subepicardial or mid-myocardial fibrosis by late gadolinium enhancement (LGE) is typical for myocarditis. Cardiac arrhythmias are frequent events that may arise especially in more severe myocarditis cases. The most common form of arrhythmia is atrial fibrillation, followed by ventricular tachycardia. Documented arrhythmias have been reported more commonly with HIV myocarditis than other more common infections such as Adenovirus, Parvovirus B19, human Herpes virus 6, and Enterovirus. The mechanisms of arrhythmogenesis in myocardial inflammation are not fully understood; in the acute phase, the spectrum of arrhythmogenesis ranges from a direct effect on cardiomyocytes that leads to electrical instability and ion channel impairment to ischemia from coronary macro- or microvascular disease. In chronic myocarditis, instead, myocardial replacement with fibrosis promotes scar-mediated re-entrant ventricular arrhythmias. Observational data suggested the important role of CMR, with LGE being the strongest independent predictor of SCD, cardiac, and all-cause mortality. In acute myocarditis, the most common localization of subepicardial LGE dwells in the lateral wall. Patients with myocarditis that develop HF and arrhythmias usually show a larger LGE distribution involving several myocardial segments. Moreover, a mid-layer LGE in the interventricular septum is more frequent in acute myocarditis than in acute coronary syndromes cases. The risk of SCD in patients with wide areas of LGE is significant, and a shared decision-making approach is warranted. Nevertheless, there is no formal consensus about the extension of LGE to justify implantable cardioverter defibrillator (ICD) implantation in primary prevention.
Collapse
Affiliation(s)
- Fabiola B. Sozzi
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Elisa Gherbesi
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Andrea Faggiano
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Eleonora Gnan
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Alessio Maruccio
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| | - Marco Schiavone
- Cardiology Unit, Luigi Sacco University Hospital, Milan, Italy
| | | | - Stefano Carugo
- Cardiology Unit, Internal Medicine Department, Fondazione Ospedale Maggiore Policlinico IRCCS Cà Granda, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Venkataraman S, Bhardwaj A, Belford PM, Morris BN, Zhao DX, Vallabhajosyula S. Veno-Arterial Extracorporeal Membrane Oxygenation in Patients with Fulminant Myocarditis: A Review of Contemporary Literature. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:215. [PMID: 35208538 PMCID: PMC8876206 DOI: 10.3390/medicina58020215] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/12/2022] [Accepted: 01/27/2022] [Indexed: 11/16/2022]
Abstract
Fulminant myocarditis is characterized by life threatening heart failure presenting as cardiogenic shock requiring inotropic or mechanical circulatory support to maintain tissue perfusion. There are limited data on the role of veno-arterial extracorporeal membrane oxygenation (VA-ECMO) in the management of fulminant myocarditis. This review seeks to evaluate the management of fulminant myocarditis with a special emphasis on the role and outcomes with VA-ECMO use.
Collapse
Affiliation(s)
- Shreyas Venkataraman
- Department of Medicine, Barnes-Jewish Hospital, Washington University of Saint Louis, St. Louis, MO 63110, USA;
| | - Abhishek Bhardwaj
- Respiratory Institute, Cleveland Clinic Foundation, Cleveland, OH 44106, USA;
| | - Peter Matthew Belford
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (P.M.B.); (D.X.Z.)
| | - Benjamin N. Morris
- Section of Cardiovascular and Critical Care Anesthesia, Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA;
| | - David X. Zhao
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (P.M.B.); (D.X.Z.)
| | - Saraschandra Vallabhajosyula
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (P.M.B.); (D.X.Z.)
| |
Collapse
|
22
|
Myocardial inflammation and sudden death in the inherited cardiomyopathies. Can J Cardiol 2022; 38:427-438. [DOI: 10.1016/j.cjca.2022.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/08/2022] [Accepted: 01/08/2022] [Indexed: 12/20/2022] Open
|
23
|
Escher F, Aleshcheva G, Pietsch H, Baumeier C, Gross UM, Schrage BN, Westermann D, Bock CT, Schultheiss HP. Transcriptional Active Parvovirus B19 Infection Predicts Adverse Long-Term Outcome in Patients with Non-Ischemic Cardiomyopathy. Biomedicines 2021; 9:1898. [PMID: 34944716 PMCID: PMC8698988 DOI: 10.3390/biomedicines9121898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022] Open
Abstract
Parvovirus B19 (B19V) is the predominant cardiotropic virus currently found in endomyocardial biopsies (EMBs). However, direct evidence showing a causal relationship between B19V and progression of inflammatory cardiomyopathy are still missing. The aim of this study was to analyze the impact of transcriptionally active cardiotropic B19V infection determined by viral RNA expression upon long-term outcomes in a large cohort of adult patients with non-ischemic cardiomyopathy in a retrospective analysis from a prospective observational cohort. In total, the analyzed study group comprised 871 consecutive B19V-positive patients (mean age 50.0 ± 15.0 years) with non-ischemic cardiomyopathy who underwent EMB. B19V-positivity was ascertained by routine diagnosis of viral genomes in EMBs. Molecular analysis of EMB revealed positive B19V transcriptional activity in n = 165 patients (18.9%). Primary endpoint was all-cause mortality in the overall cohort. The patients were followed up to 60 months. On the Cox regression analysis, B19V transcriptional activity was predictive of a worse prognosis compared to those without actively replicating B19V (p = 0.01). Moreover, multivariable analysis revealed transcriptional active B19V combined with inflammation [hazard ratio 4.013, 95% confidence interval 1.515-10.629 (p = 0.005)] as the strongest predictor of impaired survival even after adjustment for age and baseline LVEF (p = 0.005) and independently of viral load. The study demonstrates for the first time the pathogenic clinical importance of B19V with transcriptional activity in a large cohort of patients. Transcriptionally active B19V infection is an unfavourable prognostic trigger of adverse outcome. Our findings are of high clinical relevance, indicating that advanced diagnostic differentiation of B19V positive patients is of high prognostic importance.
Collapse
Affiliation(s)
- Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Ganna Aleshcheva
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
| | - Heiko Pietsch
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
| | - Ulrich M. Gross
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
| | - Benedikt Norbert Schrage
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20246 Hamburg, Germany; (B.N.S.); (D.W.)
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 20246 Hamburg, Germany
| | - Dirk Westermann
- Department of Cardiology, University Heart and Vascular Center Hamburg, 20246 Hamburg, Germany; (B.N.S.); (D.W.)
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, 20246 Hamburg, Germany
| | - Claus-Thomas Bock
- Department of Infectious Diseases, Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Robert Koch Institute, 13353 Berlin, Germany;
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany
| | - Heinz-Peter Schultheiss
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (G.A.); (H.P.); (C.B.); (U.M.G.); (H.-P.S.)
| |
Collapse
|
24
|
Schultheiss HP, Baumeier C, Pietsch H, Bock CT, Poller W, Escher F. Cardiovascular consequences of viral infections: from COVID to other viral diseases. Cardiovasc Res 2021; 117:2610-2623. [PMID: 34609508 PMCID: PMC8500164 DOI: 10.1093/cvr/cvab315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Infection of the heart muscle with cardiotropic viruses is one of the major aetiologies of myocarditis and acute and chronic inflammatory cardiomyopathy (DCMi). However, viral myocarditis and subsequent dilated cardiomyopathy is still a challenging disease to diagnose and to treat and is therefore a significant public health issue globally. Advances in clinical examination and thorough molecular genetic analysis of intramyocardial viruses and their activation status have incrementally improved our understanding of molecular pathogenesis and pathophysiology of viral infections of the heart muscle. To date, several cardiotropic viruses have been implicated as causes of myocarditis and DCMi. These include, among others, classical cardiotropic enteroviruses (Coxsackieviruses B), the most commonly detected parvovirus B19, and human herpes virus 6. A newcomer is the respiratory virus that has triggered the worst pandemic in a century, SARS-CoV-2, whose involvement and impact in viral cardiovascular disease is under scrutiny. Despite extensive research into the pathomechanisms of viral infections of the cardiovascular system, our knowledge regarding their treatment and management is still incomplete. Accordingly, in this review, we aim to explore and summarize the current knowledge and available evidence on viral infections of the heart. We focus on diagnostics, clinical relevance and cardiovascular consequences, pathophysiology, and current and novel treatment strategies.
Collapse
Affiliation(s)
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
| | - Heiko Pietsch
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - C -Thomas Bock
- Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Department of Infectious Diseases, Robert Koch Institute, Berlin 13353 Germany
- Institute of Tropical Medicine, University of Tübingen, Tübingen 72074, Germany
| | - Wolfgang Poller
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Department of Cardiology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin12203, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
| | - Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
25
|
Schultheiss HP, Baumeier C, Aleshcheva G, Bock CT, Escher F. Viral Myocarditis-From Pathophysiology to Treatment. J Clin Med 2021; 10:5240. [PMID: 34830522 PMCID: PMC8623269 DOI: 10.3390/jcm10225240] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of acute and chronic myocarditis remains a challenge for clinicians. Characterization of this disease has been hampered by its diverse etiologies and heterogeneous clinical presentations. Most cases of myocarditis are caused by infectious agents. Despite successful research in the last few years, the pathophysiology of viral myocarditis and its sequelae leading to severe heart failure with a poor prognosis is not fully understood and represents a significant public health issue globally. Most likely, at a certain point, besides viral persistence, several etiological types merge into a common pathogenic autoimmune process leading to chronic inflammation and tissue remodeling, ultimately resulting in the clinical phenotype of dilated cardiomyopathy. Understanding the underlying molecular mechanisms is necessary to assess the prognosis of patients and is fundamental to appropriate specific and personalized therapeutic strategies. To reach this clinical prerequisite, there is the need for advanced diagnostic tools, including an endomyocardial biopsy and guidelines to optimize the management of this disease. The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has currently led to the worst pandemic in a century and has awakened a special sensitivity throughout the world to viral infections. This work aims to summarize the pathophysiology of viral myocarditis, advanced diagnostic methods and the current state of treatment options.
Collapse
Affiliation(s)
- Heinz-Peter Schultheiss
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (C.B.); (G.A.); (F.E.)
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (C.B.); (G.A.); (F.E.)
| | - Ganna Aleshcheva
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (C.B.); (G.A.); (F.E.)
| | - C.-Thomas Bock
- Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany;
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany
| | - Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, 12203 Berlin, Germany; (C.B.); (G.A.); (F.E.)
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
26
|
Italia L, Ingallina G, Napolano A, Boccellino A, Belli M, Cannata F, Rolando M, Ancona F, Melillo F, Stella S, Ripa M, Scarpellini P, Tresoldi M, Ortalda A, Righetti B, De Cobelli F, Esposito A, Ciceri F, Castagna A, Rovere PQ, Fragasso G, Agricola E. Subclinical myocardial dysfunction in patients recovered from COVID-19. Echocardiography 2021; 38:1778-1786. [PMID: 34672005 PMCID: PMC8652678 DOI: 10.1111/echo.15215] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/29/2021] [Accepted: 09/22/2021] [Indexed: 01/15/2023] Open
Abstract
Background Myocardial injury (MI) can be detected during the acute phase of Coronavirus disease 19 (COVID‐19) and is associated with a dismal prognosis. Recent imaging studies described the persistence of cardiac abnormalities after the recovery. The aim of the study was to investigate the spectrum of cardiac abnormalities at mid‐term follow‐up in patients recovered from COVID‐19 using clinical assessment, laboratory tests, and imaging evaluation with comprehensive echocardiography. Methods This is an observational, cross‐sectional study assessing an unselected cohort of consecutive patients recovered from COVID‐19. MI was defined by elevated plasma levels of high sensitive troponin T (hsTnT). At the follow‐up, a complete examination including echocardiography was performed. Results The 123 patients included were divided into two groups according to the presence of MI during hospitalization: group A (without MI) and group B (with MI). After a median of 85 days, group B patients were more frequently symptomatic for dyspnea and had significantly higher values of hsTnT and N‐Terminal prohormone of Brain Natriuretic Peptide (NT‐proBNP), compared to Group A. No differences between the two groups in left nor right ventricle dimension and ejection fraction were found. However, in group B a significant reduction of mean left ventricle global longitudinal strain was observed (‐15.7±.7 vs ‐18.1± .3 in group A, p < 0.001), together with higher frequency of impaired diastolic function and higher values of pulmonary pressure. Conclusions In patients recovered from COVID‐19, echocardiography with speckle‐tracking analysis may be an useful imaging tool to identify subclinical myocardial dysfunction and potentially guide management strategies.
Collapse
Affiliation(s)
- Leonardo Italia
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Ingallina
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Napolano
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Boccellino
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Martina Belli
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Cannata
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Rolando
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Ancona
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Melillo
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Stella
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Ripa
- Department of Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Scarpellini
- Department of Infectious Disease, San Raffaele Scientific Institute, Milan, Italy
| | - Moreno Tresoldi
- Department of General Medicine and Advanced Care, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Ortalda
- Department of Anesthesia and Intensive Care, San Raffaele Scientific Institute, Milan, Italy
| | - Beatrice Righetti
- Department of Anesthesia and Intensive Care, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco De Cobelli
- Vita-Salute San Raffaele University, Milan, Italy.,Experimental Imaging Center, Radiology Department, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Esposito
- Vita-Salute San Raffaele University, Milan, Italy.,Experimental Imaging Center, Radiology Department, San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Castagna
- Department of Infectious Disease, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Patrizia Querini Rovere
- Vita-Salute San Raffaele University, Milan, Italy.,Internal Medicine, Diabetes and Endocrinology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Fragasso
- Heart Failure Clinic, Clinical Cardiology, San Raffaele Scientific Institute, Milan, Italy
| | - Eustachio Agricola
- Cardiovascular Imaging Unit, San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
27
|
Hirono K, Takarada S, Okabe M, Miyao N, Nakaoka H, Ibuki K, Ozawa S, Origasa H, Ichida F, Imanaka-Yoshida K. Clinical significance of chronic myocarditis: systematic review and meta-analysis. Heart Vessels 2021; 37:300-314. [PMID: 34365565 DOI: 10.1007/s00380-021-01914-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 11/30/2022]
Abstract
Chronic myocarditis is a prolonged inflammatory condition in the myocardium and its histological manifestation is defined by the presence of an inflammatory infiltrate. Chronic myocarditis has not been well known and its treatment of chronic myocarditis has not been established. Primary outcome of this study was to assess the efficacy of immunomodulatory treatment in addition to conventional treatment, and secondary outcomes were to clarity the prognosis of natural history of chronic myocarditis and incidence of chronic myocarditis in patients with dilated cardiomyopathy (DCM). We searched for studies in Medline, Embase, Cochrane Central Register of Controlled Trials, and Igaku Chuo Zasshi published between January 1946 and June 2020. Sixteen studies met the inclusion criteria. A meta-analysis revealed that patients receiving immunomodulatory treatment showed an improvement in left ventricular ejection fraction after immunomodulatory treatment compared to the control group (hazard ratio, 16.65; confidence interval, 4.55-28.74; p = 0.007). Five-year survival rate of the patients with inflammatory DCM (iDCM) and DCM was 52.7-70.3% and 51.9-91.1%, respectively. Moreover, 51.5%-62.7% of patients with DCM met the criteria of iDCM. Our systematic review revealed that patients with chronic myocarditis had poor prognosis and immunomodulatory treatment was significantly effective in addition to conventional treatment.
Collapse
Affiliation(s)
- Keiichi Hirono
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan.
| | - Shinya Takarada
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Mako Okabe
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Nariaki Miyao
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Hideyuki Nakaoka
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Keijiro Ibuki
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Sayaka Ozawa
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama, 930-0194, Japan
| | - Hideki Origasa
- Biostatistics and Clinical Epidemiology, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Fukiko Ichida
- Department of Pediatrics, International University of Health&Welfare, Tokyo, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
28
|
Wu L, Wang W, Leng Q, Tang N, Zhou N, Wang Y, Wang DW. Focus on Autoimmune Myocarditis in Graves' Disease: A Case-Based Review. Front Cardiovasc Med 2021; 8:678645. [PMID: 34307494 PMCID: PMC8292634 DOI: 10.3389/fcvm.2021.678645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
The manifestations of hyperthyroidism-related myocardial damage are multitudinous, including arrhythmia, dilated cardiomyopathy, valvular diseases, and even cardiogenic shock. Acute myocarditis induced by thyrotoxicosis had been reported in a few studies. However, attention on its prevalence and underlying mechanisms is sorely lacking. Its long-term harm is often ignored, and it may eventually develop into dilated cardiomyopathy and heart failure. We report a case of Graves' disease with a progressive elevation of hypersensitive cardiac troponin-I at several days after discontinuation of the patient's anti-thyroid drugs. Cardiac magnetic resonance imaging (CMRI) showed inflammatory edema of some cardiomyocytes (stranded enhanced signals under T2 mapping), myocardial necrosis (scattered enhanced signals under T1 late gadolinium enhancement) in the medial and inferior epicardial wall, with a decreased left ventricular systolic function (48%), which implied a possibility of acute myocarditis induced by thyrotoxicosis. The patient was then given a transient glucocorticoid (GC) treatment and achieved a good curative effect. Inspired by this case, we aim to systematically elaborate the pathogenesis, diagnosis, and treatment of hyperthyroidism-induced autoimmune myocarditis. Additionally, we emphasize the importance of CMRI and GC therapy in the diagnosis and treatment of hyperthyroidism-related myocarditis.
Collapse
Affiliation(s)
- Lujin Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Qianru Leng
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Nana Tang
- Nursing Teaching Office of Internal Medicine, Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science & Technology, Wuhan, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Rroku A, Kottwitz J, Heidecker B. Update on myocarditis - what we know so far and where we may be heading. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2021; 10:455–467. [PMID: 32319308 DOI: 10.1177/2048872620910109] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Affiliation(s)
- Andi Rroku
- Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Germany
| | | | - Bettina Heidecker
- Charite Universitätsmedizin Berlin, Campus Benjamin Franklin, Germany
| |
Collapse
|
30
|
Blagova OV, Nedostup AV, Sedov VP, Zaitsev AY, Novosadov VM, Kogan EA. Effectiveness of myocarditis therapy depending on the diagnosis approach (with or without myocardial biopsy). КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-2637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim. To evaluate the effectiveness of myocarditis therapy depending on the diagnosis approach (with or without myocardial biopsy).Material and methods. The study included 83 patients ≥18 years old with severe and moderate myocarditis (25 women and 58 men; mean age, 45,7±11,7 years), established by myocardial biopsy (group 1, n=36) or by a non-invasive diagnostic algorithm (group 2, n=47), for which immunosuppressive therapy (IST) was carried out. Inclusion criteria were left ventricular (LV) end-diastolic dimension >5,5 cm and ejection fraction (EF) <50%. An endomyocardial (n=31) or intraoperative (n=5) biopsy with a study of the viral genome and level of anticardiac antibodies were performed. Coronary angiography (29%), cardiac multislice computed tomography (75%), cardiac magnetic resonance imaging (41%), and 99mTc-MIBI scintigraphy (35%) were also carried out. The mean follow-up period was 3 years (36 [12; 65] months). The study was approved by the Intercollegiate Ethics Committee.Results. The groups were completely comparable in age, baseline parameters (class III [2,25; 3] and III [2; 3] heart failure (HF); end-diastolic LV dimension, 6,7±0,7 and 6,4±0,7 cm; EF, 29,9±8,7 and 31,4±9,3%), the extent of cardiac therapy (excluding the administration rate of в-blockers — 94,4 and 78,7%, p<0,05) and 1ST (methylprednisolone in 91,7 and 89,4% of patients at a mean dose of 24 [16; 32] and 20 [15; 32] mg/day, azathioprine in 50,0 and 46,8% of patients at a mean dose of 150 mg/day or mycophenolate mofetil 2,0 g/day in 30,6% in group 1, hydroxychloroquine 0,2 g/day in 27,8 and 23,4%). Biopsy in group 1 revealed active/borderline (61/39%) myocarditis, in 8 patients — viral genome in the myocardium, including parvovirus B19 in 7 of them. Both groups showed a comparable significant increase in EF after 6 months up to 37,6±8,1 and 42,6±11,5% (p<0,001) and after 27 [12; 54] months up to 43,4±9,6 and 45,5±12,3% (p<0,001), as well as a significant decrease in HF class to 2 [1; 2] in both groups. An increase in EF by ≥10% was recorded in 70 and 72% of patients, respectively. The mortality rate was 13,9 and 12,8%. Taking into account the only transplantation in group 2, the death+transplantation endpoints reached 13,9 and 14,9% of patients (without significant differences between the groups).Conclusion. In patients with severe and moderate myocarditis diagnosed with and without myocardial biopsy, the effectiveness of combined therapy, including IST, was comparable. If it is impossible to perform a biopsy, complex non-invasive strategy makes it possible to diagnose myocarditis with different probability rate and conduct an effective IST, the refusal of which mostly is not justified.
Collapse
Affiliation(s)
| | | | - V. P. Sedov
- I.M. Sechenov First Moscow State Medical University
| | | | | | - E. A. Kogan
- I.M. Sechenov First Moscow State Medical University
| |
Collapse
|
31
|
Schultheiss HP, Bock T, Pietsch H, Aleshcheva G, Baumeier C, Fruhwald F, Escher F. Nucleoside Analogue Reverse Transcriptase Inhibitors Improve Clinical Outcome in Transcriptional Active Human Parvovirus B19-Positive Patients. J Clin Med 2021; 10:1928. [PMID: 33946917 PMCID: PMC8125167 DOI: 10.3390/jcm10091928] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/24/2021] [Accepted: 04/25/2021] [Indexed: 01/01/2023] Open
Abstract
Human parvovirus B19 (B19V) is the predominant cardiotropic virus associated with dilated inflammatory cardiomyopathy (DCMi). Transcriptionally active cardiotropic B19V infection is clinically relevant and triggers adverse long-term mortality. During the study; we evaluated whether antiviral treatment with the nucleoside analogue telbivudine (LTD) is effective in suppressing transcriptional active B19V in endomyocardial biopsies (EMBs) of B19V positive patients and improving clinical outcomes. Seventeen B19V-positive patients (13 male; mean age 45.7 ± 13.9 years; mean left ventricular ejection fraction (LVEF) 37.7 ± 13.5%) with positive B19V DNA and transcriptional activity (B19V mRNA) in EMBs were treated with 600 mg/d LTD over a period of six months. Patients underwent EMBs before and after termination of the LTD treatment. B19V RNA copy numbers remained unchanged in 3/17 patients (non-responder) and declined or disappeared completely in the remaining 14/17 patients (responder) (p ≤ 0.0001). Notably; LVEF improvement was more significant in patients who reduced or lost B19V RNA (responder; p = 0.02) in contrast to non-responders (p = 0.7). In parallel; responder patients displayed statistically significant improvement in quality of life (QoL) questionnaires (p = 0.03) and dyspnea on exertion (p = 0.0006), reflecting an improvement in New York Heart Association (NYHA) Classification (p = 0.001). Our findings demonstrated for the first time that suppression of B19V transcriptional activity by LTD treatment improved hemodynamic and clinical outcome significantly. Thus; the present study substantiates the clinical relevance of detecting B19V transcriptional activity of the myocardium.
Collapse
Affiliation(s)
- Heinz-Peter Schultheiss
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
- Department of Cardiology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 12200 Berlin, Germany
| | - Thomas Bock
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
- Institute of Tropical Medicine, University of Tübingen, 72074 Tübingen, Germany
| | - Heiko Pietsch
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Ganna Aleshcheva
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
| | - Christian Baumeier
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
| | | | - Felicitas Escher
- Institute of Cardiac Diagnostics and Therapy, 12203 Berlin, Germany; (T.B.); (H.P.); (G.A.); (C.B.); (F.E.)
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
32
|
Kadhi A, Mohammed F, Nemer G. The Genetic Pathways Underlying Immunotherapy in Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:613295. [PMID: 33937353 PMCID: PMC8079649 DOI: 10.3389/fcvm.2021.613295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is a global public health threat affecting 26 million individuals worldwide with an estimated prevalence increase of 46% by 2030. One of the main causes of HF and sudden death in children and adult is Dilated Cardiomyopathy (DCM). DCM is characterized by dilation and systolic dysfunction of one or both ventricles. It has an underlying genetic basis or can develop subsequent to various etiologies that cause myocardium inflammation (secondary causes). The morbidity and mortality rates of DCM remains high despite recent advancement to manage the disease. New insights have been dedicated to better understand the pathogenesis of DCM in respect to genetic and inflammatory basis by linking the two entities together. This cognizance in the field of cardiology might have an innovative approach to manage DCM through targeted treatment directed to the causative etiology. The following review summarizes the genetical and inflammatory causes underlying DCM and the pathways of the novel precision-medicine-based immunomodulatory strategies to salvage and prevent the associated heart failure linked to the disease.
Collapse
Affiliation(s)
- Ayat Kadhi
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Fathima Mohammed
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Georges Nemer
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
33
|
Aleshcheva G, Pietsch H, Escher F, Schultheiss HP. MicroRNA profiling as a novel diagnostic tool for identification of patients with inflammatory and/or virally induced cardiomyopathies. ESC Heart Fail 2020; 8:408-422. [PMID: 33215881 PMCID: PMC7835602 DOI: 10.1002/ehf2.13090] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 01/03/2023] Open
Abstract
AIMS MicroRNAs (miRNAs) might be used as prospective biomarkers for the identification of unexplained heart failure caused by a viral and/or inflammatory process. The aim of this study was to identify and to evaluate prognostic miRNAs in serum of patients with inflammatory heart diseases diagnosed by endomyocardial biopsies. METHODS AND RESULTS After TaqMan® OpenArray® screening of 754 unique circulating miRNAs in serum of biopsy-proven patients [184 patients with inflammatory and/or virally induced myocardial diseases (DCMi), 25 patients with dilated cardiomyopathy (DCM), and 25 healthy donors], we identified seven miRNAs of interest (P < 0.05). These data have been verified by single qRT-PCR assays in other biopsy-proven patients (159 patients with viral and/or inflammatory myocardial diseases, 46 patients with DCM, and 60 healthy donors). The expression of let-7f, miR-197, miR-223, miR-93, and miR-379 allowed us to differentiate between patients with a virus and/or inflammation and healthy donors (P < 0.05) with the specificity over 93%. Based on the expression of miR-21 and miR-30a-5p, we could sort out patients with DCM from all other study groups (P < 0.05) with the specificity over 95%. CONCLUSIONS This miRNA profile provides for the first time a new non-invasive diagnostic perspective to identify patients with intramyocardial inflammation and/or viral persistence only from single serum sample, independently of prescribed therapy and time of symptoms onset. It allows the early finding of those patients relevant for myocardial biopsy for exact diagnosis and further proscription of causal aetiology-driven specific treatment.
Collapse
Affiliation(s)
- Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany
| | - Heiko Pietsch
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, 12203, Germany.,Department of Cardiology, Campus Virchow, Charité - University Hospital Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | | |
Collapse
|
34
|
Efficacy of immunosuppressive therapy in myocarditis: A 30-year systematic review and meta analysis. Autoimmun Rev 2020; 20:102710. [PMID: 33197576 DOI: 10.1016/j.autrev.2020.102710] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/01/2020] [Indexed: 12/20/2022]
Abstract
AIMS Myocarditis is an inflammation of the heart muscle, due to infectious, toxic or autoimmune causes. Literature reported controversial results in relation to the effect of immunosuppression (IS)/immunomodulation (IM). We aimed at assessing the effect of IS/IM by meta analysis. METHODS AND RESULTS Using the P.R.I.S.M.A. approach, two researchers searched for relevant studies on PubMed, Embase, and the Central Registry of Controlled Trials of the Cochrane Library. Proposed MeSH terms were: "immunotherapy OR immune therapy OR immune modeling OR Immunosuppressive Agents" AND "combination OR combined with OR plus" AND "myocarditis OR cardiomyopathies OR inflammatory cardiomyopathy". The language was restricted to English. Reference lists of included articles and those relevant to the topic were hand searched for the identification of additional, potentially relevant articles. The cutoff date was from 1987 until 30th Nov 2019. Reported survival or mortality events or change of left ventricular ejection fraction (LVEF) after IS/IT were primary outcomes of the study; in addition, improvement of New York Heart Association class, follow-up biopsy (Bx) findings, viral genome clearance on Bx and recurrence of myocarditis were recorded if reported. Statistical analysis was conducted using Review Manager 5.3; 5452 studies were screened, of these 73 were assessed for eligibility, including 8 randomized control studies, 26 retrospective studies, 2 prospective studies and 1 case control study, 34 case reports and 2 case series. In prospective studies, the difference in mortality between the IS and control groups tended to be lower in the combined IS groups (12.5% vs. 18.2%) (95% CI of odds ratio 0.7(0.3, 1.64)) and the pooled difference of the increase of LVEF between the IS and control groups tended to be higher in the combined IS groups (95% CI 7.26 (-2.29, 16.81)). In retrospective studies, the difference of survival between the IS and control group was significantly in favor of IS (95%CI Hazard ratio 0.82(0.69, 0.96)). CONCLUSIONS A tailored IS may be considered in myocarditis, depending on the phase of the disease, and the type of underlying autoimmune or immune-mediated form.
Collapse
|
35
|
Fang Y, Chang Z, Xu Z, Hu J, Zhou H, Yu S, Wan X. Osteoglycin silencing exerts inhibitory effects on myocardial fibrosis and epithelial/endothelial-mesenchymal transformation in a mouse model of myocarditis. Biofactors 2020; 46:1018-1030. [PMID: 33141515 DOI: 10.1002/biof.1683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022]
Abstract
Osteoglycin (Ogn), a class III SLRP member with multiple glycosylation sites, has been proposed to be engaged in cardiac dysfunction and adverse remodeling in human heart failure following myocardial infarction. However, the underlying mechanism remains to be elucidated. Thus, we sought to define the role of Ogn in regulation of the Wnt pathway on myocardial fibrosis and epithelial/endothelial-mesenchymal transformation (EMT/EndMT) in mice with myocarditis. The pathological changes are observed, while hematoxylin-eosin staining and picric acid Sirius red staining were conducted in successfully constructed myocarditis mouse models. Immunohistochemistry and enzyme-linked immunosorbent assay were adopted to determine Ogn and β-catenin levels and serum procollagen propeptide concentrations in the mouse myocardial tissues, respectively. Expression of Ogn and Wnt signaling pathway-related factors were measured by reverse transcription quantitative polymerase chain reaction and western blot assay, cell viability by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and cell cycle distribution and apoptosis by flow cytometry. We saw indicative pathological changes accompanied by many Ogn and β-catenin positive cells and increased serum procollagen propeptide, in the mouse myocardial tissues. Loss function assays showed reduced levels of Ogn, β-catenin, LRP6, TGF-β1, Twist, FSP-1, α-SMA and higher levels of E-cadherin and VE-cadherin, together with decreased proliferation rate, as well as increased apoptosis rate, indicating that the Wnt signaling pathway, proliferation were inhibited while apoptosis was enhanced with upon gene silencing. Coherently, depletion of Ogn inhibits myocardial fibroblasts proliferation and EMT/EndMT while facilitating myocardial fibroblasts apoptosis in myocarditis through the Wnt signaling pathway, thus serving as an intervention target for the molecular treatment of myocarditis.
Collapse
Affiliation(s)
- Yan Fang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Zhitang Chang
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Zhicheng Xu
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Jing Hu
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Haiwen Zhou
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Songping Yu
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Xuan Wan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
36
|
Abstract
Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.
Collapse
|
37
|
Evaluation of Myocardial Gene Expression Profiling for Superior Diagnosis of Idiopathic Giant-Cell Myocarditis and Clinical Feasibility in a Large Cohort of Patients with Acute Cardiac Decompensation. J Clin Med 2020; 9:jcm9092689. [PMID: 32825201 PMCID: PMC7563288 DOI: 10.3390/jcm9092689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/31/2022] Open
Abstract
Aims: The diagnostic approach to idiopathic giant-cell myocarditis (IGCM) is based on identifying various patterns of inflammatory cell infiltration and multinucleated giant cells (GCs) in histologic sections taken from endomyocardial biopsies (EMBs). The sampling error for detecting focally located GCs by histopathology is high, however. The aim of this study was to demonstrate the feasibility of gene profiling as a new diagnostic method in clinical practice, namely in a large cohort of patients suffering from acute cardiac decompensation. Methods and Results: In this retrospective multicenter study, EMBs taken from n = 427 patients with clinically acute cardiac decompensation and suspected acute myocarditis were screened (mean age: 47.03 ± 15.69 years). In each patient, the EMBs were analyzed on the basis of histology, immunohistology, molecular virology, and gene-expression profiling. Out of the total of n = 427 patient samples examined, GCs could be detected in 26 cases (6.1%) by histology. An established myocardial gene profile consisting of 27 genes was revealed; this was narrowed down to a specified profile of five genes (CPT1, CCL20, CCR5, CCR6, TLR8) which serve to identify histologically proven IGCM with high specificity in 25 of the 26 patients (96.2%). Once this newly established profiling approach was applied to the remaining patient samples, an additional n = 31 patients (7.3%) could be identified as having IGCM without any histologic proof of myocardial GCs. In a subgroup analysis, patients diagnosed with IGCM using this gene profiling respond in a similar fashion to immunosuppressive therapy as patients diagnosed with IGCM by conventional histology alone. Conclusions: Myocardial gene-expression profiling is a promising new method in clinical practice, one which can predict IGCM even in the absence of any direct histologic proof of GCs in EMB sections. Gene profiling is of great clinical relevance in terms of (a) overcoming the sampling error associated with purely histologic examinations and (b) monitoring the effectiveness of therapy.
Collapse
|
38
|
Blagova O, Nedostup A, Kogan E, Zaitsev A, Fomin V. Immunosuppressive therapy of biopsy proved immune-mediated lymphocytic myocarditis in the virus-negative and virus-positive patients. Cardiovasc Pathol 2020; 49:107260. [PMID: 32683240 DOI: 10.1016/j.carpath.2020.107260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/18/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE to study the effect of immunosupressive therapy (IST) in the virus-negative and virus-positive patients with immune-mediated myocarditis. METHODS in 60 patients (45 male, 46.7 ± 11.8 years, mean LV EDD, 6.7 ± 0.7 cm, EF 26.2 ± 9.1%) active/borderline myocarditis was verified by endomyocardial biopsy (n = 38), intraoperative biopsy (n = 10), examination of explanted heart (n = 3) and autopsy (n = 9). Indications for IST determined based on histological, immune activity. The follow-up was 19.0 [7.25; 40.25] months. RESULTS The viral genome in the myocardium was detected in 32 patients (V+ group), incl. parvovirus B19 in 23. The anti-heart antibody level was equally high in the V+ and V- patients. Antiviral therapy was administered in 24 patients. IST (in 22 V+ and 24 V- patients) include steroids (n = 40), hydroxychloroquine (n = 20), azathioprine (n = 21). The significant decrease of LV EDD (6.7 ± 0.7 to 6.4 ± 0.8), PAP (48.9 ± 15.5 to 39.4 ± 11.5 mm Hg, р<0,01), increase of EF (26.5 ± 0.9 to 36.0 ± 10.8), and lower lethality (23.9% and 64.3%; RR 0.37, 95% CI 0.19-0.71), p<0.01, were found only in IST group. Significant improvement due to IST were achieved not only in V-, but also in V+ patients. CONCLUSIONS IST in patients with immune-mediated lymphocytic myocarditis is effective and is associated with lower lethality both in virus-negative and virus-positive patients.
Collapse
Affiliation(s)
- Olga Blagova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation.
| | - Alexander Nedostup
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Evgeniya Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Alexander Zaitsev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Victor Fomin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| |
Collapse
|
39
|
De Luca G, Campochiaro C, Sartorelli S, Peretto G, Dagna L. Therapeutic strategies for virus-negative myocarditis: a comprehensive review. Eur J Intern Med 2020; 77:9-17. [PMID: 32402564 DOI: 10.1016/j.ejim.2020.04.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
Virus-negative or autoimmune myocarditis(VNM) is an inflammatory disease affecting the myocardium that may occur as a distinct disease with exclusive cardiac involvement, or in the context of systemic autoimmune or inflammatory disorders. The pathogenesis of VNM involves both innate and acquired immunity and is not completely elucidated: an early immune-mediated pathogenic process lead to subacute and chronic stages and eventually results in tissue remodeling, fibrosis, contractile dysfunction, dilated cardiomyopathy and arrhythmic burden, accounting for a dismal prognosis. Treatment interventions effectively curbing the acute inflammatory process at an early stage can prevent late cardiac remodeling and improve patient's outcome. The mainstay of treatment of VNM remains symptomatic therapy of heart failure and arrhythmia, while the use of immunosuppressive treatments has long been considered controversial until recently, and strategies effectively targeting the inflammatory and immune-mediated substrate of the disease remain elusive. Only steroids and azathioprine have been tested in clinical trials, and nowadays represent the therapy of choice. A substantial proportion of patients are resistant to first line strategies, suggesting that some critical inflammatory mechanisms are not responsive to conventional immunosuppression with steroids and azathioprine, or experience drug-related adverse events. Thus, second-line targeted therapeutic strategies to treat VNM are eagerly awaited. Recent data on the pathogenic mechanisms underlying myocardial inflammation are paving the way to novel, promising treatment strategies for myocarditis, which could reformulate future treatment strategies for VNM. In this review, we summarize the current therapeutic opportunities, beyond corticosteroids, to treat VNM, including conventional and biologic immunosuppressive drugs and cytokine blocking agents.
Collapse
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Sartorelli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy
| | - Giovanni Peretto
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
40
|
Abstract
Inflammatory dilated cardiomyopathy (DCMi) is a syndrome, not an etiological disease entity. The infective etiology and the immunopathology can be best determined through endomyocardial biopsy with a complete work-up by light microscopy, immunohistology, and polymerase chain reaction for microbial agents. This review focuses on the methodological advances in diagnosis in the past few years and exemplifies the importance of an etiology-orientated treatment in different case scenarios. In fulminant nonviral myocarditis, immunosuppressive treatment together with hemodynamic stabilization of the patient via mechanical circulatory support (e.g., microaxial pumps, extracorporeal membrane oxygenation, left ventricular assist device) can be life-saving. For viral inflammatory cardiomyopathy, intravenous immunoglobulin treatment can resolve inflammation and often eradicate the virus.
Collapse
Affiliation(s)
- Bernhard Maisch
- Medical Faculty, Philipps University, Marburg, Germany.
- Heart and Vessel Centre, Marburg, Germany.
| | - Sabine Pankuweit
- Department of Internal Medicine and Cardiology, Philipps University and UKGM, Marburg, Germany
| |
Collapse
|
41
|
Elsanhoury A, Tschöpe C, Van Linthout S. A Toolbox of Potential Immune-Related Therapies for Inflammatory Cardiomyopathy. J Cardiovasc Transl Res 2020; 14:75-87. [PMID: 32440911 PMCID: PMC7892499 DOI: 10.1007/s12265-020-10025-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022]
Abstract
Myocarditis is a multifactorial disorder, characterized by an inflammatory reaction in the myocardium, predominantly triggered by infectious agents, but also by antigen mimicry or autoimmunity in susceptible individuals. Unless spontaneously resolved, a chronic inflammatory course concludes with cardiac muscle dysfunction portrayed by ventricular dilatation, clinically termed inflammatory cardiomyopathy (Infl-CM). Treatment strategies aim to resolve chronic inflammation and preserve cardiac function. Beside standard heart failure treatments, which only play a supportive role in this condition, systemic immunosuppressants are used to diminish inflammatory cell function at the cost of noxious side effects. To date, the treatment protocols are expert-based without large clinical evidence. This review describes concept and contemporary strategies to alleviate myocardial inflammation and sheds light on potential inflammatory targets in an evidence-based order.
Collapse
Affiliation(s)
- Ahmed Elsanhoury
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK), Föhrerstrasse 15, 13353, Berlin, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany.
| |
Collapse
|
42
|
Abstract
Myocarditis is generally a mild and self-limited consequence of systemic infection of cardiotropic viruses. However, patients can develop a temporary or permanent impairment of cardiac function including acute cardiomyopathy with hemodynamic compromise or severe arrhythmias. In this setting, specific causes of inflammation are associated with variable risks of death and transplantation. Recent translational studies suggest that treatments tailored to specific causes of myocarditis may impact clinical outcomes when added to guideline-directed medical care. This review summarizes recent advances in translational research that influence the utility of endomyocardial biopsy for the management of inflammatory cardiomyopathies. Emerging therapies for myocarditis based on these mechanistic hypotheses are entering clinical trials and may add to the benefits of established heart failure treatment.
Collapse
Affiliation(s)
- Carsten Tschöpe
- From the Charité, University Medicine Berlin, Campus Virchow Klinikum (CVK), Department of Cardiology, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin-Brandenburg Centrum für Regenerative Therapien, Germany (C.T., S.V.L.).,Deutsches Zentrum für Herz Kreislauf Forschung (DZHK)-Standort Berlin/Charité, Germany (C.T., S.V.L.)
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL (L.T.C.)
| | - Guillermo Torre-Amione
- Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (G.T.-A.).,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Vascular, Monterrey, Nuevo León, Mexico (G.T.-A.)
| | - Sophie Van Linthout
- From the Charité, University Medicine Berlin, Campus Virchow Klinikum (CVK), Department of Cardiology, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, Germany (C.T., S.V.L.).,Charité-Universitätsmedizin Berlin, BCRT-Berlin-Brandenburg Centrum für Regenerative Therapien, Germany (C.T., S.V.L.).,Deutsches Zentrum für Herz Kreislauf Forschung (DZHK)-Standort Berlin/Charité, Germany (C.T., S.V.L.)
| |
Collapse
|
43
|
Degener F, Salameh A, Manuylova T, Pickardt T, Kostelka M, Daehnert I, Berger F, Messroghli D, Schubert S, Klingel K. First paediatric cohort for the evaluation of inflammation in endomyocardial biopsies derived from congenital heart surgery. Int J Cardiol 2020; 303:36-40. [DOI: 10.1016/j.ijcard.2019.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 09/08/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022]
|
44
|
De Luca G, Campochiaro C, Sartorelli S, Peretto G, Sala S, Palmisano A, Esposito A, Candela C, Basso C, Rizzo S, Thiene G, Della Bella P, Dagna L. Efficacy and safety of mycophenolate mofetil in patients with virus-negative lymphocytic myocarditis: A prospective cohort study. J Autoimmun 2020; 106:102330. [DOI: 10.1016/j.jaut.2019.102330] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 12/12/2022]
|
45
|
|
46
|
Merken J, Hazebroek M, Van Paassen P, Verdonschot J, Van Empel V, Knackstedt C, Abdul Hamid M, Seiler M, Kolb J, Hoermann P, Ensinger C, Brunner-La Rocca HP, Poelzl G, Heymans S. Immunosuppressive Therapy Improves Both Short- and Long-Term Prognosis in Patients With Virus-Negative Nonfulminant Inflammatory Cardiomyopathy. Circ Heart Fail 2019; 11:e004228. [PMID: 29449368 DOI: 10.1161/circheartfailure.117.004228] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inflammatory cardiomyopathy (infl-CMP) is characterized by increased cardiac inflammation in the absence of viruses, ischemia, valvular disease, or other apparent causes. Studies addressing the efficacy of immunosuppressive therapy in patients with infl-CMP are sparse. This study retrospectively investigates whether immunosuppressive agents on top of heart failure therapy according to current guidelines improves cardiac function and long-term outcome in patients with infl-CMP. METHODS AND RESULTS Within the Innsbruck and Maastricht Cardiomyopathy Registry, a total of 209 patients fulfilled the criteria for infl-CMP using endomyocardial biopsy (≥14 infiltrating inflammatory cells/mm2). A total of 110 (53%) patients received immunosuppressive therapy and 99 (47%) did not. To correct for potential selection bias, 1:1 propensity score matching was used on all significant baseline parameters, resulting in a total of 90 patients per group. Baseline characteristics did not significantly differ between both patient groups, reflecting optimal propensity score matching. After a median follow-up of 31 (15-47) months, immunosuppressive therapy resulted in an improved long-term outcome (eg, heart transplantation-free survival) as compared with standard heart failure therapy alone (Log-rank P=0.043; hazard ratio, 0.34 [95% CI, 0.17-0.92]) and in a significant larger increase of left ventricular ejection fraction after a mean of 12 months follow-up, as compared with patients receiving standard heart failure treatment only (12.2% versus 7.3%, respectively; P=0.036). CONCLUSIONS To conclude, this study suggests that immunosuppressive therapy in infl-CMP patients results in an improved heart transplantation-free survival as compared with standard heart failure therapy alone, underscoring the urgent need for a large prospective multicenter trial.
Collapse
Affiliation(s)
- Jort Merken
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria.
| | - Mark Hazebroek
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Pieter Van Paassen
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Job Verdonschot
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Vanessa Van Empel
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Christian Knackstedt
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Myrurgia Abdul Hamid
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Michael Seiler
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Julian Kolb
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Philipp Hoermann
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Christian Ensinger
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Hans-Peter Brunner-La Rocca
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Gerhard Poelzl
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| | - Stephane Heymans
- From the Cardiology Department (J.M., M.H., J.V., V.V.E., C.K., H.-P.B.-L.R., S.H.), Immunology Department (P.V.P.), and Pathology Department (M.A.H.), Maastricht University Medical Center, The Netherlands; and Clinical Division of Cardiology and Angiology (M.S., J.K., P.H., G.P.) and Institute of Pathology (C.E.), Innsbruck Medical University, Austria
| |
Collapse
|
47
|
Seferović PM, Polovina M, Bauersachs J, Arad M, Gal TB, Lund LH, Felix SB, Arbustini E, Caforio AL, Farmakis D, Filippatos GS, Gialafos E, Kanjuh V, Krljanac G, Limongelli G, Linhart A, Lyon AR, Maksimović R, Miličić D, Milinković I, Noutsias M, Oto A, Oto Ö, Pavlović SU, Piepoli MF, Ristić AD, Rosano GM, Seggewiss H, Ašanin M, Seferović JP, Ruschitzka F, Čelutkiene J, Jaarsma T, Mueller C, Moura B, Hill L, Volterrani M, Lopatin Y, Metra M, Backs J, Mullens W, Chioncel O, Boer RA, Anker S, Rapezzi C, Coats AJ, Tschöpe C. Heart failure in cardiomyopathies: a position paper from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2019; 21:553-576. [DOI: 10.1002/ejhf.1461] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/20/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Petar M. Seferović
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Serbian Academy of Sciences and Arts Belgrade Serbia
| | - Marija Polovina
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Department of CardiologyClinical Center of Serbia Belgrade Serbia
| | - Johann Bauersachs
- Department of Cardiology and AngiologyMedical School Hannover Hannover Germany
| | - Michael Arad
- Cardiomyopathy Clinic and Heart Failure Institute, Leviev Heart Center, Sheba Medical Center and Sackler School of Medicine, Tel Aviv University Tel Aviv Israel
| | - Tuvia Ben Gal
- Department of CardiologyRabin Medical Center, Sackler Faculty of Medicine, Tel Aviv University Tel Aviv Israel
| | - Lars H. Lund
- Department of MedicineKarolinska Institutet, and Heart and Vascular Theme, Karolinska University Hospital Stockholm Sweden
| | - Stephan B. Felix
- Department of Internal Medicine BUniversity Medicine Greifswald Greifswald Germany
| | - Eloisa Arbustini
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation, University Hospital Policlinico San Matteo Pavia Italy
| | - Alida L.P. Caforio
- Division of Cardiology, Department of Cardiological, Thoracic and Vascular SciencesUniversity of Padua Padua Italy
| | - Dimitrios Farmakis
- University of Cyprus Medical School, Nicosia, Cyprus; Heart Failure Unit, Department of CardiologyAthens University Hospital Attikon, National and Kapodistrian University of Athens Athens Greece
| | - Gerasimos S. Filippatos
- University of Cyprus Medical School, Nicosia, Cyprus; Heart Failure Unit, Department of CardiologyAthens University Hospital Attikon, National and Kapodistrian University of Athens Athens Greece
| | - Elias Gialafos
- Second Department of CardiologyHeart Failure and Preventive Cardiology Section, Henry Dunant Hospital Athens Greece
| | | | - Gordana Krljanac
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Department of CardiologyClinical Center of Serbia Belgrade Serbia
| | - Giuseppe Limongelli
- Department of Cardiothoracic Sciences, Università della Campania ‘Luigi VanvitellI’Monaldi Hospital, AORN Colli, Centro di Ricerca Cardiovascolare, Ospedale Monaldi, AORN Colli, Naples, Italy, and UCL Institute of Cardiovascular Science London UK
| | - Aleš Linhart
- Second Department of Medicine, Department of Cardiovascular MedicineGeneral University Hospital, Charles University in Prague Prague Czech Republic
| | - Alexander R. Lyon
- National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital London UK
| | - Ružica Maksimović
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Centre for Radiology and Magnetic Resonance Imaging, Clinical Centre of Serbia Belgrade Serbia
| | - Davor Miličić
- Department of Cardiovascular DiseasesUniversity Hospital Center Zagreb, University of Zagreb Zagreb Croatia
| | - Ivan Milinković
- Department of CardiologyClinical Center of Serbia Belgrade Serbia
| | - Michel Noutsias
- Mid‐German Heart Center, Department of Internal Medicine III, Division of CardiologyAngiology and Intensive Medical Care, University Hospital Halle, Martin‐Luther‐University Halle Halle Germany
| | - Ali Oto
- Department of CardiologyHacettepe University Faculty of Medicine Ankara Turkey
| | - Öztekin Oto
- Department of Cardiovascular SurgeryDokuz Eylül University Faculty of Medicine İzmir Turkey
| | - Siniša U. Pavlović
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Pacemaker Center, Clinical Center of Serbia Belgrade Serbia
| | | | - Arsen D. Ristić
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Department of CardiologyClinical Center of Serbia Belgrade Serbia
| | - Giuseppe M.C. Rosano
- Centre for Clinical and Basic Research, Department of Medical SciencesIRCCS San Raffaele Pisana Rome Italy
| | - Hubert Seggewiss
- Medizinische Klinik, Kardiologie & Internistische Intensivmedizin, Klinikum Würzburg‐Mitte Würzburg Germany
| | - Milika Ašanin
- University of Belgrade Faculty of Medicine Belgrade Serbia
- Department of CardiologyClinical Center of Serbia Belgrade Serbia
| | - Jelena P. Seferović
- Cardiovascular DivisionBrigham and Women's Hospital, Harvard Medical School Boston MA USA
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Center Serbia and Faculty of MedicineUniversity of Belgrade Belgrade Serbia
| | - Frank Ruschitzka
- Department of CardiologyUniversity Heart Center Zürich Switzerland
| | - Jelena Čelutkiene
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of MedicineVilnius University Vilnius Lithuania
- State Research Institute Centre for Innovative Medicine Vilnius Lithuania
| | - Tiny Jaarsma
- Department of Social and Welfare Studies, Faculty of Health ScienceLinköping University Linköping Sweden
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of CardiologyUniversity Hospital Basel, University of Basel Basel Switzerland
| | - Brenda Moura
- Cardiology DepartmentCentro Hospitalar São João Porto Portugal
| | - Loreena Hill
- School of Nursing and Midwifery, Queen's University Belfast Belfast UK
| | | | - Yuri Lopatin
- Volgograd State Medical University, Regional Cardiology Centre Volgograd Volgograd Russia
| | - Marco Metra
- Cardiology, Department of Medical and Surgical SpecialtiesRadiological Sciences, and Public Health, University of Brescia Brescia Italy
| | - Johannes Backs
- Department of Molecular Cardiology and EpigeneticsUniversity of Heidelberg Heidelberg Germany
- DZHK (German Centre for Cardiovascular Research) partner site Heidelberg/Mannheim Heidelberg Germany
| | - Wilfried Mullens
- BIOMED ‐ Biomedical Research Institute, Faculty of Medicine and Life SciencesHasselt University Diepenbeek Belgium
- Department of CardiologyZiekenhuis Oost‐Limburg Genk Belgium
| | - Ovidiu Chioncel
- University of Medicine Carol Davila Bucharest Romania
- Emergency Institute for Cardiovascular Diseases, ‘Prof. C. C. Iliescu’ Bucharest Romania
| | - Rudolf A. Boer
- Department of CardiologyUniversity Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Stefan Anker
- Division of Cardiology and Metabolism, Department of Cardiology (CVK)Charité Berlin Germany
- Berlin‐Brandenburg Center for Regenerative Therapies (BCRT) Berlin Germany
- DZHK (German Centre for Cardiovascular Research) partner site Berlin, Charité Berlin Germany
| | - Claudio Rapezzi
- Cardiology, Department of ExperimentalDiagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna Bologna Italy
| | - Andrew J.S. Coats
- Monash University, Australia, and University of Warwick Coventry UK
- Pharmacology, Centre of Clinical and Experimental Medicine, IRCCS San Raffaele Pisana, Rome, Italy, and St George's University of London London UK
| | - Carsten Tschöpe
- Berlin‐Brandenburg Center for Regenerative Therapies, Deutsches Zentrum für Herz‐Kreislauf‐Forschung (DZHK) Berlin, Department of CardiologyCampus Virchow Klinikum, Charite ‐ Universitaetsmedizin Berlin Berlin Germany
| |
Collapse
|
48
|
Maisch B. Cardio-Immunology of Myocarditis: Focus on Immune Mechanisms and Treatment Options. Front Cardiovasc Med 2019; 6:48. [PMID: 31032264 PMCID: PMC6473396 DOI: 10.3389/fcvm.2019.00048] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/27/2019] [Indexed: 12/13/2022] Open
Abstract
Myocarditis and inflammatory cardiomyopathy are syndromes, not aetiological disease entities. From animal models of cardiac inflammation we have detailed insight of the strain specific immune reactions based on the genetic background of the animal and the infectiosity of the virus. Innate and adaptive immunity also react in man. An aetiological diagnosis of a viral vs. a non-viral cause is possible by endomyocardial biopsy with histology, immunohistology and PCR for microbial agents. This review deals with the different etiologies of myocarditis and inflammatory cardiomyopathy on the basis of the genetic background and the predisposition for inflammation. It analyses the epidemiologic shift in cardiotropic viral agents in the last 30 years. Based on the understanding of the interaction between infection and the players of the innate and adaptive immune system it summarizes pathogenetic phases and clinical faces of myocarditis. It gives an up-to-date information on specific treatment options beyond symptomatic heart failure and antiarrhythmic therapy. Although inflammation can resolve spontaneously, specific treatment directed to the causative etiology is often required. For fulminant, acute, and chronic autoreactive myocarditis without viral persistence immunosuppressive treatment can be life-saving, for viral inflammatory cardiomyopathy ivIg treatment can resolve inflammation and often eradicate the virus.
Collapse
Affiliation(s)
- Bernhard Maisch
- Faculty of Medicine, and Heart and Vessel Center, Philipps-University, Marburg, Germany
| |
Collapse
|
49
|
Severe eosinophilic myocarditis associated with modafinil in a patient with normal peripheral eosinophil count. Clin Res Cardiol 2019; 108:963-966. [DOI: 10.1007/s00392-019-01434-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 02/05/2019] [Indexed: 12/28/2022]
|
50
|
Schultheiss HP, Fairweather D, Caforio ALP, Escher F, Hershberger RE, Lipshultz SE, Liu PP, Matsumori A, Mazzanti A, McMurray J, Priori SG. Dilated cardiomyopathy. Nat Rev Dis Primers 2019; 5:32. [PMID: 31073128 PMCID: PMC7096917 DOI: 10.1038/s41572-019-0084-1] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dilated cardiomyopathy (DCM) is a clinical diagnosis characterized by left ventricular or biventricular dilation and impaired contraction that is not explained by abnormal loading conditions (for example, hypertension and valvular heart disease) or coronary artery disease. Mutations in several genes can cause DCM, including genes encoding structural components of the sarcomere and desmosome. Nongenetic forms of DCM can result from different aetiologies, including inflammation of the myocardium due to an infection (mostly viral); exposure to drugs, toxins or allergens; and systemic endocrine or autoimmune diseases. The heterogeneous aetiology and clinical presentation of DCM make a correct and timely diagnosis challenging. Echocardiography and other imaging techniques are required to assess ventricular dysfunction and adverse myocardial remodelling, and immunological and histological analyses of an endomyocardial biopsy sample are indicated when inflammation or infection is suspected. As DCM eventually leads to impaired contractility, standard approaches to prevent or treat heart failure are the first-line treatment for patients with DCM. Cardiac resynchronization therapy and implantable cardioverter-defibrillators may be required to prevent life-threatening arrhythmias. In addition, identifying the probable cause of DCM helps tailor specific therapies to improve prognosis. An improved aetiology-driven personalized approach to clinical care will benefit patients with DCM, as will new diagnostic tools, such as serum biomarkers, that enable early diagnosis and treatment.
Collapse
Affiliation(s)
- Heinz-Peter Schultheiss
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany. .,Department of Cardiology, Charité-Universitaetsmedizin Berlin, Berlin, Germany.
| | - DeLisa Fairweather
- Mayo Clinic, Department of Cardiovascular Medicine, Jacksonville, FL, USA.
| | - Alida L. P. Caforio
- 0000 0004 1757 3470grid.5608.bDivision of Cardiology, Department of Cardiological Thoracic and Vascular Sciences and Public Health, University of Padua, Padova, Italy
| | - Felicitas Escher
- grid.486773.9Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany ,0000 0001 2218 4662grid.6363.0Department of Cardiology, Charité–Universitaetsmedizin Berlin, Berlin, Germany ,0000 0004 5937 5237grid.452396.fDZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Ray E. Hershberger
- 0000 0001 2285 7943grid.261331.4Divisions of Human Genetics and Cardiovascular Medicine in the Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| | - Steven E. Lipshultz
- 0000 0004 1936 9887grid.273335.3Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY USA ,0000 0000 9958 7286grid.413993.5Oishei Children’s Hospital, Buffalo, NY USA ,Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Peter P. Liu
- 0000 0001 2182 2255grid.28046.38University of Ottawa Heart Institute, Ottawa, Ontario Canada
| | - Akira Matsumori
- grid.410835.bClinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Andrea Mazzanti
- 0000 0004 1762 5736grid.8982.bDepartment of Molecular Medicine, University of Pavia, Pavia, Italy ,Department of Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
| | - John McMurray
- 0000 0001 2193 314Xgrid.8756.cBritish Heart Foundation (BHF) Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Silvia G. Priori
- 0000 0004 1762 5736grid.8982.bDepartment of Molecular Medicine, University of Pavia, Pavia, Italy ,Department of Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
| |
Collapse
|