1
|
Farrant JP, Schmitt M, Reid AB, Garratt CJ, Newman WG, Malhotra A, Beynon R, Mahmod M, Raman B, Cooper RM, Dawson D, Green T, Prasad SK, Singh A, Dodd S, Watkins H, Neubauer S, Miller CA. Considerations for drug trials in hypertrophic cardiomyopathy. ESC Heart Fail 2025; 12:1095-1112. [PMID: 39462184 PMCID: PMC11911595 DOI: 10.1002/ehf2.15138] [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: 09/12/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heterogeneous condition with potentially serious manifestations. Management has traditionally comprised therapies to palliate symptoms and implantable cardioverter-defibrillators to prevent sudden cardiac death. The need for disease-modifying therapies has been recognized for decades. More recently, an increasing number of novel and repurposed therapies hypothesized to target HCM disease pathways have been evaluated, culminating in the recent regulatory approval of mavacamten, a novel oral myosin inhibitor. HCM poses several unique challenges for clinical trials, which are important to recognize when designing trials and interpreting findings. This manuscript discusses the key considerations in the context of recent and ongoing randomized trials, including the roles of genotype, phenotype and symptom status in patient selection, the evidence base for clinical and mechanistic outcome measurements, trial duration and sample size.
Collapse
Affiliation(s)
- John P. Farrant
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
| | - Matthias Schmitt
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
| | - Anna B. Reid
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
| | - Clifford J. Garratt
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
| | - William G. Newman
- Manchester Centre for Genomic Medicine Manchester University NHS Foundation TrustOxford RoadManchesterM13 9WLUK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
| | - Aneil Malhotra
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
- Institute of SportManchester Metropolitan University99 Oxford RdManchesterM1 7ELUK
| | - Rhys Beynon
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordOX3 9DUUK
- NIHR Oxford Biomedical Research CentreOxford University Hospitals Foundation TrustOxfordOX3 9DUUK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordOX3 9DUUK
- NIHR Oxford Biomedical Research CentreOxford University Hospitals Foundation TrustOxfordOX3 9DUUK
| | - Robert M. Cooper
- Liverpool Heart and Chest HospitalThomas DrLiverpoolL14 3PEUK
- Liverpool John Moores University70 Mount PleasantMerseysideL3 5UXUK
| | - Dana Dawson
- School of MedicineUniversity of AberdeenAberdeenAB25 2ZDUK
- Cardiology DepartmentAberdeen Royal InfirmaryAberdeenAB25 2ZNUK
| | - Thomas Green
- Cardiology DepartmentNorthumbria Healthcare NHS TrustNorthumberlandUK
| | - Sanjay K. Prasad
- Royal Brompton and Harefield NHS Foundation TrustSydney StLondonSW3 6NPUK
- National Heart and Lung InstituteImperial College LondonLondon
| | - Anvesha Singh
- Department of Cardiovascular SciencesUniversity of Leicester and the NIHR Leicester Biomedical Research Centre, Glenfield HospitalGroby RoadLeicesterLE3 9QPUK
| | - Susanna Dodd
- Department of Health Data Sciences, Institute of Population Health, Faculty of Health and Life SciencesUniversity of LiverpoolBlock F, Waterhouse Boulevard, 1‐5 Brownlow StreetLiverpoolL69 3GLUK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordOX3 9DUUK
- NIHR Oxford Biomedical Research CentreOxford University Hospitals Foundation TrustOxfordOX3 9DUUK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordOX3 9DUUK
- NIHR Oxford Biomedical Research CentreOxford University Hospitals Foundation TrustOxfordOX3 9DUUK
| | - Christopher A. Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PLUK
- Manchester University NHS Foundation TrustSouthmoor Road, WythenshaweManchesterM23 9LTUK
- Wellcome Centre for Cell‐Matrix Research, Division of Cell‐Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science CentreUniversity of ManchesterOxford RoadManchesterM13 9PTUK
| |
Collapse
|
2
|
van de Bovenkamp AA, Nassiri S, Bakermans AJ, Burchell GL, de Man FS, van Loon RB, Handoko ML. Long-term hemodynamic responses and reverse remodeling after pharmacotherapy in HFpEF versus HFrEF: a systematic review and meta-analysis. Am J Physiol Heart Circ Physiol 2025; 328:H419-H432. [PMID: 39825764 DOI: 10.1152/ajpheart.00544.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/06/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025]
Abstract
The acute response to therapeutic afterload reduction differs between heart failure with preserved (HFpEF) versus reduced ejection fraction (HFrEF), with larger left ventricular (LV) stroke work augmentation in HFrEF compared with HFpEF. This may (partially) explain the neutral effect of HFrEF-medication in HFpEF. It is unclear whether such differences in hemodynamic response persist and/or differentially trigger reverse remodeling in the case of long-term afterload reduction. A systematic search was performed, identifying 21 clinical trials investigating renin-angiotensin-aldosterone system (RAAS) inhibitors, β-blockers, and sodium-glucose cotransport 2 inhibitors that report data on afterload reduction, stroke volume, and reverse remodeling in HFpEF and/or HFrEF. In both HFpEF and HFrEF, meta-analyses revealed limited long-term change in systolic/diastolic blood pressure (-5.6/-3.2 and -4.6/-1.4 mmHg, respectively) and LV afterload reduction (arterial elastance: -0.039 and -0.055 mmHg/mL, respectively). Long-term treatment did not result in an increase in stroke volume, with the exception of β-blockers in HFrEF. Indexed LV mass decreased slightly in both HFpEF and HFrEF (-2.8 and -2.3 g/m2, respectively). In HFrEF, treatment reduced LV end-diastolic and end-systolic volume (-8 and -6 mL, respectively), whereas in HFpEF there was no relevant change. Contrary to acute heart failure studies, long-term afterload reduction had little effect on blood pressure and stroke volume augmentation in both HFpEF and HFrEF. However, reverse remodeling was clearly present in HFrEF but was essentially absent in HFpEF.
Collapse
Affiliation(s)
- Arno A van de Bovenkamp
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - Soufiane Nassiri
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - Adrianus J Bakermans
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - George L Burchell
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ramon B van Loon
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
| | - M Louis Handoko
- Department of Cardiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Heart failure & Arrhythmias, Amsterdam, The Netherlands
- Department of Cardiology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
3
|
Calligaris M, Aleksova A, Fluca AL, Janjusevic M, Carpi G, Stefanizzi D, Carnevali S, Curcio F, Puca AA, Cattaneo M, Beltrami AP. Protective role of the longevity-associated BPIFB4 gene on cardiac microvascular cells and cardiac aging. Vascul Pharmacol 2025; 158:107470. [PMID: 39909151 DOI: 10.1016/j.vph.2025.107470] [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: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
In recent years, the role of the cardiac microvasculature in modulating the symptoms and disease progression of patients affected by cardiac pathology has been reconsidered. The term cardiac microvascular disease (CMD) describes the set of functional and/or structural alterations of the cardiac microvasculature that reduce the ability of the heart to adequately increase its coronary blood flow to keep up with increased metabolic demand. CMD is involved in the evolution of heart disease of both ischemic and non-ischemic origin as well as in cardiac aging. The primary actors involved in this process are the cells of the stromal compartment, whose nature and biology are now investigated to a new level of detail thanks to single-cell omics studies. Recent studies on the genetics of extreme longevity have identified a polymorphic haplotype variant of the BPIFB4 gene that confers prolonged life span and health span, atheroprotective advantages, and an improved immune response. The aim of this review was to focus on the beneficial effects of the longevity-associated variant (LAV) of BPIFB4 on cardiac microvascular cell biology, providing novel and exciting mechanisms of its action directed against the development or progression of many age-related cardiovascular diseases, thus emphasizing its translational therapeutic potential.
Collapse
Affiliation(s)
| | - Aneta Aleksova
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy; Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Alessandra Lucia Fluca
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy
| | - Milijana Janjusevic
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy
| | - Giada Carpi
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | | | | | - Francesco Curcio
- Department of Medicine (DMED), University of Udine, Udine, Italy; Laboratory Medicine Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Annibale Alessandro Puca
- IRCCS MultiMedica, Milan, Italy; Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Università degli Studi di Salerno, Salerno, Italy.
| | | | - Antonio Paolo Beltrami
- Department of Medicine (DMED), University of Udine, Udine, Italy; Laboratory Medicine Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy.
| |
Collapse
|
4
|
Mitsuboshi S, Morizumi M, Imai S, Hori S, Kotake K. Association between mineralocorticoid receptor antagonists and kidney harm: A systematic review and meta-analysis of randomized controlled trials. Pharmacotherapy 2025; 45:43-53. [PMID: 39578707 DOI: 10.1002/phar.4618] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 11/24/2024]
Abstract
Conflicting data have been reported on the association between mineralocorticoid receptor antagonists (MRAs) and acute kidney injury (AKI). This systematic review and meta-analysis aimed to evaluate whether MRAs affect the risk of AKI. MEDLINE via PubMed, the Cochrane Central Register of Controlled Trials, and the ClinicalTrials.gov website were comprehensively searched to extract all relevant studies. Randomized controlled trials (RCTs) were selected that compared MRA versus placebo or no treatment and had study populations consisting of patients with heart or kidney disease. The primary outcome was AKI. The secondary outcome was kidney injury, including AKI and non-AKI. Thirty-three studies were included in the meta-analysis. MRAs were not associated with an increased risk of AKI (risk ratio [RR] 1.13, 95% confidence interval [CI] 0.88-1.46, p = 0.29, I2 = 15%, 18,065 patients, 13 RCTs, moderate certainty). For the secondary outcome, MRAs were associated with an increased risk of kidney injury (RR 1.52, 95% CI 1.24-1.87, p < 0.01, I2 = 48%, 27,492 patients, 33 RCTs, low certainty). In particular, only canrenone (RR 5.39, 95% CI 2.17-13.37, p < 0.01) and spironolactone (RR 1.78, 95% CI 1.48-2.14, p < 0.01) were associated with an increased risk of kidney injury. However, eplerenone and finerenone seem not to increase the risk of kidney injury in patients with heart or kidney disease. The selection of MRAs might influence the risk of kidney-associated events. Further studies focusing on individual MRAs may be needed to clarify these differences.
Collapse
Affiliation(s)
| | - Makoto Morizumi
- Department of Pharmacy, Ohno Memorial Hospital, Osaka, Japan
| | - Shungo Imai
- Division of Drug Informatics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Satoko Hori
- Division of Drug Informatics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Kazumasa Kotake
- Department of Pharmacy, Zikei Hospital/Zikei Institute of Psychiatry, Okayama, Japan
| |
Collapse
|
5
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [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: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
6
|
Sato S, Ogawa Y, Wong CW, Le HL, Yee RW, Gombos DS, Negishi K, Hirayama M. Mineralocorticoid receptor expression and the effects of the mineralocorticoid receptor antagonist spironolactone in a murine model of graft-versus-host disease. Ocul Surf 2024; 34:477-488. [PMID: 39424225 DOI: 10.1016/j.jtos.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The topical administration of spironolactone, a mineralocorticoid receptor antagonist (MRA) improves dry eye symptoms in patients with ocular graft-versus-host disease (GVHD); however, the detailed mechanism remains unclear. This study aimed to investigate the effects of spironolactone eyedrops on the ocular surface using a chronic GVHD (cGVHD) mouse model and to determine the expression of the mineralocorticoid receptor (MR). METHODS A cGVHD mouse model was established by allogeneic bone marrow transplantation (BMT) from B10.D2 mice to BALB/c mice. Subsequently, cGVHD mice were treated with either 0.005 % spironolactone or vehicle eyedrops. The eyelids, cornea and conjunctiva of the recipients were analyzed at 4-week intervals post-BMT in both groups. RESULTS Signs of ocular GVHD, such as corneal epithelial damage, depletion of meibomian glands, and inflammatory cell infiltration onto the ocular surface, were significantly decreased in cGVHD mice treated with spironolactone eyedrops. The expression of the MR NR3C2 in the corneal and conjunctival epithelia was significantly increased in cGVHD mice. HSP47+NR3C2+ MR-expressing fibroblasts, CD45+NR3C2+ MR-expressing leukocytes, and CD4+NR3C2+ MR-expressing T cells infiltrated the ocular surface tissue of cGVHD mice significantly more than that of syngeneic controls. CONCLUSIONS MR expression is increased in epithelial cells, fibroblasts, and T cells in a murine cGVHD model, whereas MRA and spironolactone eyedrops could attenuate the severity of ocular GVHD. These findings suggest that MR signaling partially contributes to the development of ocular GVHD in this mouse model.
Collapse
Affiliation(s)
- Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.
| | - Calvin W Wong
- McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | | | - Richard W Yee
- MD PLLC, Bellaire, TX, United States; Department of Ophthalmology, University of Texas M D Anderson Cancer Center, Houston, TX, United States
| | - Dan S Gombos
- Department of Ophthalmology, University of Texas M D Anderson Cancer Center, Houston, TX, United States
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Rajagopalan S, Dobre M, Dazard JE, Vergara-Martel A, Connelly K, Farkouh ME, Gaztanaga J, Conger H, Dever A, Razavi-Nematollahi L, Fares A, Pereira G, Edwards-Glenn J, Cameron M, Cameron C, Al-Kindi S, Brook RD, Pitt B, Weir M. Mineralocorticoid Receptor Antagonism Prevents Aortic Plaque Progression and Reduces Left Ventricular Mass and Fibrosis in Patients With Type 2 Diabetes and Chronic Kidney Disease: The MAGMA Trial. Circulation 2024; 150:663-676. [PMID: 39129649 PMCID: PMC11503525 DOI: 10.1161/circulationaha.123.067620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/12/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Persistent mineralocorticoid receptor activation is a pathologic response in type 2 diabetes and chronic kidney disease. Whereas mineralocorticoid receptor antagonists are beneficial in reducing cardiovascular complications, direct mechanistic pathways for these effects in humans are lacking. METHODS The MAGMA trial (Mineralocorticoid Receptor Antagonism Clinical Evaluation in Atherosclerosis) was a randomized, double-blind, placebo-controlled trial in patients with high-risk type 2 diabetes with chronic kidney disease (not receiving dialysis) on maximum tolerated renin-angiotensin system blockade. The primary end point was change in thoracic aortic wall volume, expressed as absolute or percent value (ΔTWV or ΔPWV), using 3T magnetic resonance imaging at 12 months. Secondary end points were changes in left ventricle (LV) mass; LV fibrosis, measured as a change in myocardial native T1; and 24-hour ambulatory and central aortic blood pressures. Tertiary end points included plasma proteomic changes in 7596 plasma proteins using an aptamer-based assay. RESULTS A total of 79 patients were randomized to placebo (n=42) or 25 mg of spironolactone daily (n=37). After a modified intent-to-treat, including available baseline data of study end points, patients who completed the trial protocol were included in the final analyses. At the 12-month follow-up, the average change in PWV was 7.1±10.7% in the placebo group and 0.87±10.0% in the spironolactone group (P=0.028), and ΔTWV was 1.2±1.7 cm3 in the placebo group and 0.037±1.9 cm3 in the spironolactone group (P=0.022). Change in LV mass was 3.1±8.4 g in the placebo group and -5.8±8.4 g in the spironolactone group (P=0.001). Changes in LV T1 values were significantly different between the placebo and spironolactone groups (26.0±41.9 ms in the placebo group versus a decrease of -10.1±36.3 ms in the spironolactone group; P=6.33×10-4). Mediation analysis revealed that the spironolactone effect on thoracic aortic wall volume and myocardial mass remained significant after adjustment for ambulatory and central blood pressures. Proteomic analysis revealed a dominant effect of spironolactone on pathways involving oxidative stress, inflammation, and leukocyte activation. CONCLUSIONS Among patients with diabetes with moderate to severe chronic kidney disease at elevated cardiovascular risk, treatment with spironolactone prevented progression of aortic wall volume and resulted in regression of LV mass and favorable alterations in native T1, suggesting amelioration of left-ventricular fibrosis. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02169089.
Collapse
Affiliation(s)
- Sanjay Rajagopalan
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Mirela Dobre
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Jean-Eudes Dazard
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Armando Vergara-Martel
- University Hospitals, Cleveland, OH, USA
- Case Western Reserve University, Cleveland, OH, USA
| | - Kim Connelly
- St. Michael’s Hospital, University of Toronto, Toronto, CA
| | | | - Juan Gaztanaga
- New York University Langone Health School of Medicine, Winthrop, Mineola, NY
| | | | - Ann Dever
- University Hospitals, Cleveland, OH, USA
| | | | - Anas Fares
- University Hospitals, Cleveland, OH, USA
| | | | | | - Mark Cameron
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Sadeer Al-Kindi
- Debakey Heart and Vascular Center Houston Methodist Hospital, Houston TX
| | - Robert D. Brook
- University of Michigan Frankel Cardiovascular Center, Detroit, MI
| | | | - Matthew Weir
- Division of Nephrology, University of Maryland Medical Center, Baltimore, MD
| |
Collapse
|
8
|
Angeli E, Jordan M, Otto M, Stojanović SD, Karsdal M, Bauersachs J, Thum T, Fiedler J, Genovese F. The role of fibrosis in cardiomyopathies: An opportunity to develop novel biomarkers of disease activity. Matrix Biol 2024; 128:65-78. [PMID: 38423395 DOI: 10.1016/j.matbio.2024.02.008] [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: 10/16/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Cardiomyopathies encompass a spectrum of heart disorders with diverse causes and presentations. Fibrosis stands out as a shared hallmark among various cardiomyopathies, reflecting a common thread in their pathogenesis. This prevalent fibrotic response is intricately linked to the consequences of dysregulated extracellular matrix (ECM) remodeling, emphasizing its significance in the development and progression the disease. This review explores the ECM involvement in various cardiomyopathies and its impact on myocardial stiffness and fibrosis. Additionally, we discuss the potential of ECM fragments as early diagnosis, prognosis, and risk stratification. Biomarkers deriving from turnover of collagens and other ECM proteins hold promise in clinical applications. We outline current clinical management, future directions, and the potential for personalized ECM-targeted therapies with specific focus on microRNAs. In summary, this review examines the role of the fibrosis in cardiomyopathies, highlighting the potential of ECM-derived biomarkers in improving disease management with implications for precision medicine.
Collapse
Affiliation(s)
- Elisavet Angeli
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Nordic Bioscience A/S, Herlev, Denmark.
| | - Maria Jordan
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Mandy Otto
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Thomas Thum
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany; Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Federal Republic of Germany
| | - Jan Fiedler
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Federal Republic of Germany; Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Hanover, Federal Republic of Germany
| | - Federica Genovese
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
González A, López B, Ravassa S, San José G, Latasa I, Butler J, Díez J. Myocardial Interstitial Fibrosis in Hypertensive Heart Disease: From Mechanisms to Clinical Management. Hypertension 2024; 81:218-228. [PMID: 38084597 DOI: 10.1161/hypertensionaha.123.21708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Hypertensive heart disease (HHD) can no longer be considered as the beneficial adaptive result of the hypertrophy of cardiomyocytes in response to pressure overload leading to the development of left ventricular hypertrophy. The current evidence indicates that in patients with HHD, pathological lesions in the myocardium lead to maladaptive structural remodeling and subsequent alterations in cardiac function, electrical activity, and perfusion, all contributing to poor outcomes. Diffuse myocardial interstitial fibrosis is probably the most critically involved lesion in these disorders. Therefore, in this review, we will focus on the histological characteristics, the mechanisms, and the clinical consequences of myocardial interstitial fibrosis in patients with HHD. In addition, we will consider the most useful tools for the noninvasive diagnosis of myocardial interstitial fibrosis in patients with HHD, as well as the most effective available therapeutic strategies to prevent its development or facilitate its regression in this patient population. Finally, we will issue a call to action for the need for more fundamental and clinical research on myocardial interstitial fibrosis in HHD.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Department of Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona, Spain (A.G.)
| | - Begoña López
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Susana Ravassa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Gorka San José
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Iñigo Latasa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX (J.B.)
- Department of Medicine, University of Mississippi, Jackson (J.B.)
| | - Javier Díez
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| |
Collapse
|
10
|
Ravassa S, López B, Treibel TA, San José G, Losada-Fuentenebro B, Tapia L, Bayés-Genís A, Díez J, González A. Cardiac Fibrosis in heart failure: Focus on non-invasive diagnosis and emerging therapeutic strategies. Mol Aspects Med 2023; 93:101194. [PMID: 37384998 DOI: 10.1016/j.mam.2023.101194] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Heart failure is a leading cause of mortality and hospitalization worldwide. Cardiac fibrosis, resulting from the excessive deposition of collagen fibers, is a common feature across the spectrum of conditions converging in heart failure. Eventually, either reparative or reactive in nature, in the long-term cardiac fibrosis contributes to heart failure development and progression and is associated with poor clinical outcomes. Despite this, specific cardiac antifibrotic therapies are lacking, making cardiac fibrosis an urgent unmet medical need. In this context, a better patient phenotyping is needed to characterize the heterogenous features of cardiac fibrosis to advance toward its personalized management. In this review, we will describe the different phenotypes associated with cardiac fibrosis in heart failure and we will focus on the potential usefulness of imaging techniques and circulating biomarkers for the non-invasive characterization and phenotyping of this condition and for tracking its clinical impact. We will also recapitulate the cardiac antifibrotic effects of existing heart failure and non-heart failure drugs and we will discuss potential strategies under preclinical development targeting the activation of cardiac fibroblasts at different levels, as well as targeting additional extracardiac processes.
Collapse
Affiliation(s)
- Susana Ravassa
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Begoña López
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Thomas A Treibel
- Institute of Cardiovascular Science, University College London, UK; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Gorka San José
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Blanca Losada-Fuentenebro
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Leire Tapia
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain; Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain; ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Badalona, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| |
Collapse
|
11
|
Ridwan M, Dimiati H, Syukri M, Lesmana R. Potential molecular mechanism underlying cardiac fibrosis in diabetes mellitus: a narrative review. Egypt Heart J 2023; 75:46. [PMID: 37306727 PMCID: PMC10260731 DOI: 10.1186/s43044-023-00376-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/08/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is among the most common risk factors for cardiovascular disease in the world with prevalence of more than 500 million population in 2021. Cardiac fibrosis with its complex process has been hypothesized as one of the mechanisms explaining development of heart failure in diabetic patients. Recently, the biomolecular mechanism of cardiac fibrosis in the hyperglycemia setting has been focusing around transforming growth factor β-1 (TGFβ-1) as a major factor. However, there is interplay role of several factors including microRNAs (miRNAs) which acts as a potential regulator of cardiac fibrosis connected with TGFβ-1. In this review, we explored interplay role of several factors including microRNAs which acts as a potential regulator of cardiac fibrosis connected with TGFβ-1 in diabetes mellitus. This narrative review included articles from the PubMed and Science Direct databases published in the last 10 years (2012-2022). MAIN TEXT In diabetic patients, excessive activation of myofibroblasts occurs and triggers pro-collagen to convert into mature collagen to fill the cardiac interstitial space resulting in a pathological process of extracellular matrix remodeling. The balance between matrix metalloproteinase (MMP) and its inhibitor (tissue inhibitor of metalloproteinase, TIMP) is crucial in degradation of the extracellular matrix. Diabetes-related cardiac fibrosis is modulated by increasing level of TGF-β1 mediated by cellular components, including cardiomyocyte and non-cardiomyocyte cells involving fibroblasts, vascular pericytes smooth muscle cells, endothelial cells, mast cells, macrophages, and dendritic cells. Several miRNAs such as miR-21, miR-9, miR-29, miR-30d, miR-144, miR-34a, miR-150, miR-320, and miR-378 are upregulated in diabetic cardiomyopathy. TGF-β1, together with inflammatory cytokines, oxidative stress, combined sma and the mothers against decapentaplegic (smad) protein, mitogen-activated protein kinase (MAPK), and microRNAs, is interconnectedly involved in extracellular matrix production and fibrotic response. In this review, we explored interplay role of several factors including microRNAs which acts as a potential regulator of cardiac fibrosis connected with TGFβ-1 in diabetes mellitus. CONCLUSIONS Long-term hyperglycemia activates cardiac fibroblast via complex processes involving TGF-β1, miRNA, inflammatory chemokines, oxidative stress, smad, or MAPK pathways. There is increasing evidence of miRNA's roles lately in modulating cardiac fibrosis.
Collapse
Affiliation(s)
- Muhammad Ridwan
- Doctorate School of Medical Science, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23116, Indonesia
| | - Herlina Dimiati
- Department of Pediatrics, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia.
| | - Maimun Syukri
- Department of Internal Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Ronny Lesmana
- Physiology Division, Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
12
|
Masenga SK, Kirabo A. Hypertensive heart disease: risk factors, complications and mechanisms. Front Cardiovasc Med 2023; 10:1205475. [PMID: 37342440 PMCID: PMC10277698 DOI: 10.3389/fcvm.2023.1205475] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Hypertensive heart disease constitutes functional and structural dysfunction and pathogenesis occurring primarily in the left ventricle, the left atrium and the coronary arteries due to chronic uncontrolled hypertension. Hypertensive heart disease is underreported and the mechanisms underlying its correlates and complications are not well elaborated. In this review, we summarize the current understanding of hypertensive heart disease, we discuss in detail the mechanisms associated with development and complications of hypertensive heart disease especially left ventricular hypertrophy, atrial fibrillation, heart failure and coronary artery disease. We also briefly highlight the role of dietary salt, immunity and genetic predisposition in hypertensive heart disease pathogenesis.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Cam-Pus, Livingstone, Zambia
- School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| |
Collapse
|
13
|
Bertaud A, Joshkon A, Heim X, Bachelier R, Bardin N, Leroyer AS, Blot-Chabaud M. Signaling Pathways and Potential Therapeutic Strategies in Cardiac Fibrosis. Int J Mol Sci 2023; 24:ijms24021756. [PMID: 36675283 PMCID: PMC9866199 DOI: 10.3390/ijms24021756] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
Cardiac fibrosis constitutes irreversible necrosis of the heart muscle as a consequence of different acute (myocardial infarction) or chronic (diabetes, hypertension, …) diseases but also due to genetic alterations or aging. Currently, there is no curative treatment that is able to prevent or attenuate this phenomenon that leads to progressive cardiac dysfunction and life-threatening outcomes. This review summarizes the different targets identified and the new strategies proposed to fight cardiac fibrosis. Future directions, including the use of exosomes or nanoparticles, will also be discussed.
Collapse
|
14
|
Targeting Myocardial Fibrosis—A Magic Pill in Cardiovascular Medicine? Pharmaceutics 2022; 14:pharmaceutics14081599. [PMID: 36015225 PMCID: PMC9414721 DOI: 10.3390/pharmaceutics14081599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Fibrosis, characterized by an excessive accumulation of extracellular matrix, has long been seen as an adaptive process that contributes to tissue healing and regeneration. More recently, however, cardiac fibrosis has been shown to be a central element in many cardiovascular diseases (CVDs), contributing to the alteration of cardiac electrical and mechanical functions in a wide range of clinical settings. This paper aims to provide a comprehensive review of cardiac fibrosis, with a focus on the main pathophysiological pathways involved in its onset and progression, its role in various cardiovascular conditions, and on the potential of currently available and emerging therapeutic strategies to counteract the development and/or progression of fibrosis in CVDs. We also emphasize a number of questions that remain to be answered, and we identify hotspots for future research.
Collapse
|
15
|
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is an underdiagnosed, but potentially curable pulmonary vascular disease. The increased pulmonary vascular resistance in CTEPH is caused by unresolved proximal thrombus and secondary microvasculopathy in the pulmonary vasculature, leading to adaptive and maladaptive remodeling of the right ventricle (RV), eventual right heart failure, and death. Knowledge on the RV remodeling process in CTEPH is limited. The progression to RV failure in CTEPH is a markedly slower process. A detailed understanding of the pathophysiology and underlying mechanisms of RV remodeling may facilitate early diagnosis and the development of targeted therapy. While ultrasound, magnetic resonance imaging, right heart catheterization, and serum biomarkers have been used to assess cardiac function, the current treatment strategies reduce the afterload of the right heart, but are less effective in improving the maladaptive remodeling of the right heart. This review systematically summarizes the current knowledge on adaptive and maladaptive remodeling of the right heart in CTEPH from molecular mechanisms to clinical practice.
Collapse
|
16
|
Schimmel K, Ichimura K, Reddy S, Haddad F, Spiekerkoetter E. Cardiac Fibrosis in the Pressure Overloaded Left and Right Ventricle as a Therapeutic Target. Front Cardiovasc Med 2022; 9:886553. [PMID: 35600469 PMCID: PMC9120363 DOI: 10.3389/fcvm.2022.886553] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022] Open
Abstract
Myocardial fibrosis is a remodeling process of the extracellular matrix (ECM) following cardiac stress. "Replacement fibrosis" is a term used to describe wound healing in the acute phase of an injury, such as myocardial infarction. In striking contrast, ECM remodeling following chronic pressure overload insidiously develops over time as "reactive fibrosis" leading to diffuse interstitial and perivascular collagen deposition that continuously perturbs the function of the left (L) or the right ventricle (RV). Examples for pressure-overload conditions resulting in reactive fibrosis in the LV are systemic hypertension or aortic stenosis, whereas pulmonary arterial hypertension (PAH) or congenital heart disease with right sided obstructive lesions such as pulmonary stenosis result in RV reactive fibrosis. In-depth phenotyping of cardiac fibrosis has made it increasingly clear that both forms, replacement and reactive fibrosis co-exist in various etiologies of heart failure. While the role of fibrosis in the pathogenesis of RV heart failure needs further assessment, reactive fibrosis in the LV is a pathological hallmark of adverse cardiac remodeling that is correlated with or potentially might even drive both development and progression of heart failure (HF). Further, LV reactive fibrosis predicts adverse outcome in various myocardial diseases and contributes to arrhythmias. The ability to effectively block pathological ECM remodeling of the LV is therefore an important medical need. At a cellular level, the cardiac fibroblast takes center stage in reactive fibrotic remodeling of the heart. Activation and proliferation of endogenous fibroblast populations are the major source of synthesis, secretion, and deposition of collagens in response to a variety of stimuli. Enzymes residing in the ECM are responsible for collagen maturation and cross-linking. Highly cross-linked type I collagen stiffens the ventricles and predominates over more elastic type III collagen in pressure-overloaded conditions. Research has attempted to identify pro-fibrotic drivers causing fibrotic remodeling. Single key factors such as Transforming Growth Factor β (TGFβ) have been described and subsequently targeted to test their usefulness in inhibiting fibrosis in cultured fibroblasts of the ventricles, and in animal models of cardiac fibrosis. More recently, modulation of phenotypic behaviors like inhibition of proliferating fibroblasts has emerged as a strategy to reduce pathogenic cardiac fibroblast numbers in the heart. Some studies targeting LV reactive fibrosis as outlined above have successfully led to improvements of cardiac structure and function in relevant animal models. For the RV, fibrosis research is needed to better understand the evolution and roles of fibrosis in RV failure. RV fibrosis is seen as an integral part of RV remodeling and presents at varying degrees in patients with PAH and animal models replicating the disease of RV afterload. The extent to which ECM remodeling impacts RV function and thus patient survival is less clear. In this review, we describe differences as well as common characteristics and key players in ECM remodeling of the LV vs. the RV in response to pressure overload. We review pre-clinical studies assessing the effect of anti-fibrotic drug candidates on LV and RV function and their premise for clinical testing. Finally, we discuss the mode of action, safety and efficacy of anti-fibrotic drugs currently tested for the treatment of left HF in clinical trials, which might guide development of new approaches to target right heart failure. We touch upon important considerations and knowledge gaps to be addressed for future clinical testing of anti-fibrotic cardiac therapies.
Collapse
Affiliation(s)
- Katharina Schimmel
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Kenzo Ichimura
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sushma Reddy
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Pediatric Cardiology, Stanford University, Stanford, CA, United States
| | - Francois Haddad
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,Cardiovascular Medicine, Stanford University, Stanford, CA, United States
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA, United States,Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States,*Correspondence: Edda Spiekerkoetter,
| |
Collapse
|
17
|
Ke B, Tan X, Ren L, Fan Y, Zhang Y, Li F, Sun Q, Liu T, Jia L, Wang Y, Du J. Aldosterone dysregulation predicts the risk of mortality and rehospitalization in heart failure with a preserved ejection fraction. SCIENCE CHINA. LIFE SCIENCES 2022; 65:631-642. [PMID: 34258711 DOI: 10.1007/s11427-021-1945-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/14/2021] [Indexed: 10/20/2022]
Abstract
Serum aldosterone is associated with cardiac remodeling, which contributes to morbidity and mortality in heart failure (HF); however, the prognostic value of aldosterone in HF with a preserved ejection fraction (HFpEF) is unclear. We used liquid chromatography-tandem mass spectrometry to quantify serum aldosterone in 873 patients with HFpEF in a Registry Study of Biomarkers for HF. The retrospective study was conducted at Beijing Anzhen Hospital from May 2017 to October 2019. The primary endpoint was a composite of all-cause mortality and rehospitalization for HF. Aldosterone concentrations in patients with and without events were 124.22 pmol L-1 (interquartile range (IQR): 48.62-256.20) and 96.33 pmol L-1 (IQR: 37.33-215.76), respectively (P=0.023). Aldosterone independently predicted all-cause mortality (adjusted hazard ratio (aHR), 1.55; 95% confidence interval (95%CI), 1.06-2.27; P=0.024) and the primary endpoint (aHR, 1.43; 95%CI, 1.11-1.85; P=0.006). Patients with high aldosterone concentrations were at higher risk of concentric remodeling (adjusted odds ratio (aOR), 1.45; 95% CI, 1.03-2.04; P=0.034). Patients with high aldosterone and B-type natriuretic peptide concentrations were at a higher risk of the primary endpoint (hazard ratio (HR), 1.85; 95%CI, 1.29-2.66; P=0.001). We conclude that elevated aldosterone is associated with a risk of rehospitalization with HF and all-cause mortality in patients with HFpEF.
Collapse
Affiliation(s)
- Bingbing Ke
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Xin Tan
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lu Ren
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yangkai Fan
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yixin Zhang
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Fengjuan Li
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Qiqi Sun
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Tong Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Lixin Jia
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.,Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yuan Wang
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China. .,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| | - Jie Du
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, the Key Laboratory of Remodeling-Related Cardiovascular Disease, Ministry of Education, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China. .,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
18
|
Li Y, Sun G, Wang L. MiR-21 participates in LPS-induced myocardial injury by targeting Bcl-2 and CDK6. Inflamm Res 2022; 71:205-214. [DOI: 10.1007/s00011-021-01535-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 01/22/2023] Open
|
19
|
Potter E, Stephenson G, Harris J, Wright L, Marwick TH. Screening-Guided Spironolactone Treatment of Subclinical Left Ventricular Dysfunction for Heart Failure Prevention in At-risk Patients. Eur J Heart Fail 2022; 24:620-630. [PMID: 35014128 DOI: 10.1002/ejhf.2428] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/21/2021] [Accepted: 01/04/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Subclinical left ventricular dysfunction (LVD) is a prelude to symptomatic heart failure (HF). We hypothesised that screening-guided treatment with spironolactone would prevent incident HF in at-risk patients. METHODS We randomised asymptomatic, community-dwelling subjects aged ≥65 years old, with at least one non-ischaemic HF risk factor (hypertension, type 2 diabetes mellitus or obesity) to echocardiography-guided therapy or usual care. Spironolactone therapy was triggered by the detection of subclinical LVD (GLS ≤16% or diastolic abnormalities (at least one of E/e'>15, E/e' >10 with left atrial enlargement [LAE] or impaired relaxation [E/A <0.8, IR], LAE with IR), or borderline GLS (17%) with IR or borderline GLS with LAE. The primary outcome was incident HF at 24 months. Secondary outcomes were change in 6-minute walk test (6MWT) and change in LV function. RESULTS LVD was identified in 161 (46%) of 349 participants (age 70 years [68-73], 201 (58%) women). The trial was stopped because of a 55% rate of spironolactone discontinuation, due primarily to decline in renal function. Incident HF developed in 11 (3.5%) of 310 participants completing follow-up, with no difference between usual care and intervention (4 [2.5%] vs. 7 [4.7%], p=0.29), decline in 6MWT distance (p=0.28), persistent or new LVD (p=0.58), nor change in GLS with intervention (p=0.15). A per-protocol analysis of 131 patients with baseline LVD and a follow-up echocardiogram, showed resolution of LVD with spironolactone therapy (61% vs 33%, p=0.01). CONCLUSION The study was underpowered to determine whether screening-guided spironolactone therapy reduced incident HF because spironolactone was frequently discontinued due to renal function criteria. However, LVD resolved in more patients treated with spironolactone than in untreated patients. Future trials should use less conservative renal criteria for spironolactone discontinuation.
Collapse
Affiliation(s)
- Elizabeth Potter
- Baker Heart and Diabetes Institute.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria
| | | | | | | | - Thomas H Marwick
- Baker Heart and Diabetes Institute.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria
| |
Collapse
|
20
|
Zhou Q, Li P, Zhao H, Xu X, Li S, Zhao J, Xu D, Zeng Q. Heart Failure With Mid-range Ejection Fraction: A Distinctive Subtype or a Transitional Stage? Front Cardiovasc Med 2021; 8:678121. [PMID: 34113665 PMCID: PMC8185203 DOI: 10.3389/fcvm.2021.678121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure with mid-range ejection fraction (HFmrEF) was first proposed by Lam and Solomon in 2014, and was listed as a new subtype of heart failure (HF) in 2016 European Society of Cardiology guidelines. Since then, HFmrEF has attracted an increasing amount of attention, and the number of related studies on this topic has grown rapidly. The diagnostic criteria on the basis of left ventricular ejection fraction (LVEF) are straightforward; however, LVEF is not a static parameter, and it changes dynamically during the course of HF. Thus, HFmrEF may not be an independent disease with a uniform pathophysiological process, but rather a collection of patients with different characteristics. HFmrEF is often associated with various cardiovascular and non-cardiovascular diseases. Thus, the pathophysiological mechanisms of HFmrEF are particularly complex, and its clinical phenotypes are diverse. The complexity and heterogeneity of HFmrEF may be one reason for inconsistent results between clinical studies. In fact, whether HFmrEF is a distinctive subtype or a transitional stage between HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF) is controversial. In this review, we discuss the clinical characteristics, treatment and prognosis of patients with HFmrEF, as well as the differences among HFmrEF, HFrEF, and HFpEF.
Collapse
Affiliation(s)
- Qing Zhou
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Department of Cardiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Peixin Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hengli Zhao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University, Göttingen, Germany
| | - Shaoping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
21
|
Martin N, Manoharan K, Davies C, Lumbers RT. Beta-blockers and inhibitors of the renin-angiotensin aldosterone system for chronic heart failure with preserved ejection fraction. Cochrane Database Syst Rev 2021; 5:CD012721. [PMID: 34022072 PMCID: PMC8140651 DOI: 10.1002/14651858.cd012721.pub3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Beta-blockers and inhibitors of the renin-angiotensin-aldosterone system improve survival and reduce morbidity in people with heart failure with reduced left ventricular ejection fraction (LVEF); a review of the evidence is required to determine whether these treatments are beneficial for people with heart failure with preserved ejection fraction (HFpEF). OBJECTIVES To assess the effects of beta-blockers, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, angiotensin receptor neprilysin inhibitors, and mineralocorticoid receptor antagonists in people with HFpEF. SEARCH METHODS We updated searches of CENTRAL, MEDLINE, Embase, and one clinical trial register on 14 May 2020 to identify eligible studies, with no language or date restrictions. We checked references from trial reports and review articles for additional studies. SELECTION CRITERIA: We included randomised controlled trials with a parallel group design, enrolling adults with HFpEF, defined by LVEF greater than 40%. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 41 randomised controlled trials (231 reports), totalling 23,492 participants across all comparisons. The risk of bias was frequently unclear and only five studies had a low risk of bias in all domains. Beta-blockers (BBs) We included 10 studies (3087 participants) investigating BBs. Five studies used a placebo comparator and in five the comparator was usual care. The mean age of participants ranged from 30 years to 81 years. A possible reduction in cardiovascular mortality was observed (risk ratio (RR) 0.78, 95% confidence interval (CI) 0.62 to 0.99; number needed to treat for an additional benefit (NNTB) 25; 1046 participants; three studies), however, the certainty of evidence was low. There may be little to no effect on all-cause mortality (RR 0.82, 95% CI 0.67 to 1.00; 1105 participants; four studies; low-certainty evidence). The effects on heart failure hospitalisation, hyperkalaemia, and quality of life remain uncertain. Mineralocorticoid receptor antagonists (MRAs) We included 13 studies (4459 participants) investigating MRA. Eight studies used a placebo comparator and in five the comparator was usual care. The mean age of participants ranged from 54.5 to 80 years. Pooled analysis indicated that MRA treatment probably reduces heart failure hospitalisation (RR 0.82, 95% CI 0.69 to 0.98; NNTB = 41; 3714 participants; three studies; moderate-certainty evidence). However, MRA treatment probably has little or no effect on all-cause mortality (RR 0.91, 95% CI 0.78 to 1.06; 4207 participants; five studies; moderate-certainty evidence) and cardiovascular mortality (RR 0.90, 95% CI 0.74 to 1.11; 4070 participants; three studies; moderate-certainty evidence). MRA treatment may have little or no effect on quality of life measures (mean difference (MD) 0.84, 95% CI -2.30 to 3.98; 511 participants; three studies; low-certainty evidence). MRA treatment was associated with a higher risk of hyperkalaemia (RR 2.11, 95% CI 1.77 to 2.51; number needed to treat for an additional harmful outcome (NNTH) = 11; 4291 participants; six studies; high-certainty evidence). Angiotensin-converting enzyme inhibitors (ACEIs) We included eight studies (2061 participants) investigating ACEIs. Three studies used a placebo comparator and in five the comparator was usual care. The mean age of participants ranged from 70 to 82 years. Pooled analyses with moderate-certainty evidence suggest that ACEI treatment likely has little or no effect on cardiovascular mortality (RR 0.93, 95% CI 0.61 to 1.42; 945 participants; two studies), all-cause mortality (RR 1.04, 95% CI 0.75 to 1.45; 1187 participants; five studies) and heart failure hospitalisation (RR 0.86, 95% CI 0.64 to 1.15; 1019 participants; three studies), and may result in little or no effect on the quality of life (MD -0.09, 95% CI -3.66 to 3.48; 154 participants; two studies; low-certainty evidence). The effects on hyperkalaemia remain uncertain. Angiotensin receptor blockers (ARBs) Eight studies (8755 participants) investigating ARBs were included. Five studies used a placebo comparator and in three the comparator was usual care. The mean age of participants ranged from 61 to 75 years. Pooled analyses with high certainty of evidence suggest that ARB treatment has little or no effect on cardiovascular mortality (RR 1.02, 95% 0.90 to 1.14; 7254 participants; three studies), all-cause mortality (RR 1.01, 95% CI 0.92 to 1.11; 7964 participants; four studies), heart failure hospitalisation (RR 0.92, 95% CI 0.83 to 1.02; 7254 participants; three studies), and quality of life (MD 0.41, 95% CI -0.86 to 1.67; 3117 participants; three studies). ARB was associated with a higher risk of hyperkalaemia (RR 1.88, 95% CI 1.07 to 3.33; 7148 participants; two studies; high-certainty evidence). Angiotensin receptor neprilysin inhibitors (ARNIs) Three studies (7702 participants) investigating ARNIs were included. Two studies used ARBs as the comparator and one used standardised medical therapy, based on participants' established treatments at enrolment. The mean age of participants ranged from 71 to 73 years. Results suggest that ARNIs may have little or no effect on cardiovascular mortality (RR 0.96, 95% CI 0.79 to 1.15; 4796 participants; one study; moderate-certainty evidence), all-cause mortality (RR 0.97, 95% CI 0.84 to 1.11; 7663 participants; three studies; high-certainty evidence), or quality of life (high-certainty evidence). However, ARNI treatment may result in a slight reduction in heart failure hospitalisation, compared to usual care (RR 0.89, 95% CI 0.80 to 1.00; 7362 participants; two studies; moderate-certainty evidence). ARNI treatment was associated with a reduced risk of hyperkalaemia compared with valsartan (RR 0.88, 95% CI 0.77 to 1.01; 5054 participants; two studies; moderate-certainty evidence). AUTHORS' CONCLUSIONS There is evidence that MRA and ARNI treatment in HFpEF probably reduces heart failure hospitalisation but probably has little or no effect on cardiovascular mortality and quality of life. BB treatment may reduce the risk of cardiovascular mortality, however, further trials are needed. The current evidence for BBs, ACEIs, and ARBs is limited and does not support their use in HFpEF in the absence of an alternative indication. Although MRAs and ARNIs are probably effective at reducing the risk of heart failure hospitalisation, the treatment effect sizes are modest. There is a need for improved approaches to patient stratification to identify the subgroup of patients who are most likely to benefit from MRAs and ARNIs, as well as for an improved understanding of disease biology, and for new therapeutic approaches.
Collapse
Affiliation(s)
- Nicole Martin
- Institute of Health Informatics Research, University College London, London, UK
| | | | - Ceri Davies
- Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK
| |
Collapse
|
22
|
Kotfis K, Lechowicz K, Drożdżal S, Niedźwiedzka-Rystwej P, Wojdacz TK, Grywalska E, Biernawska J, Wiśniewska M, Parczewski M. COVID-19-The Potential Beneficial Therapeutic Effects of Spironolactone during SARS-CoV-2 Infection. Pharmaceuticals (Basel) 2021; 14:ph14010071. [PMID: 33477294 PMCID: PMC7830835 DOI: 10.3390/ph14010071] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
In March 2020, coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 was declared a global pandemic by the World Health Organization (WHO). The clinical course of the disease is unpredictable but may lead to severe acute respiratory infection (SARI) and pneumonia leading to acute respiratory distress syndrome (ARDS). It has been shown that pulmonary fibrosis may be one of the major long-term complications of COVID-19. In animal models, the use of spironolactone was proven to be an important drug in the prevention of pulmonary fibrosis. Through its dual action as a mineralocorticoid receptor (MR) antagonist and an androgenic inhibitor, spironolactone can provide significant benefits concerning COVID-19 infection. The primary effect of spironolactone in reducing pulmonary edema may also be beneficial in COVID-19 ARDS. Spironolactone is a well-known, widely used and safe anti-hypertensive and antiandrogenic medication. It has potassium-sparing diuretic action by antagonizing mineralocorticoid receptors (MRs). Spironolactone and potassium canrenoate, exerting combined pleiotropic action, may provide a therapeutic benefit to patients with COVID-19 pneumonia through antiandrogen, MR blocking, antifibrotic and anti-hyperinflammatory action. It has been proposed that spironolactone may prevent acute lung injury in COVID-19 infection due to its pleiotropic effects with favorable renin–angiotensin–aldosterone system (RAAS) and ACE2 expression, reduction in transmembrane serine protease 2 (TMPRSS2) activity and antiandrogenic action, and therefore it may prove to act as additional protection for patients at highest risk of severe pneumonia. Future prospective clinical trials are warranted to evaluate its therapeutic potential.
Collapse
Affiliation(s)
- Katarzyna Kotfis
- Department of Anesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
- Correspondence: ; Tel.: +48-91-466-11-44
| | - Kacper Lechowicz
- Department of Anesthesiology, Intensive Therapy and Acute Intoxications, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Sylwester Drożdżal
- Department of Pharmacokinetics and Monitored Therapy, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | | | - Tomasz K. Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Jowita Biernawska
- Department of Anesthesiology and Intensive Therapy, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland;
| | - Magda Wiśniewska
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, 71-455 Szczecin, Poland;
| |
Collapse
|
23
|
The effect of ginsenoside Rg5, isolated from black ginseng, on heart failure in zebrafish based on untargeted metabolomics. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
24
|
Sweeney M, Corden B, Cook SA. Targeting cardiac fibrosis in heart failure with preserved ejection fraction: mirage or miracle? EMBO Mol Med 2020; 12:e10865. [PMID: 32955172 PMCID: PMC7539225 DOI: 10.15252/emmm.201910865] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is central to the pathology of heart failure, particularly heart failure with preserved ejection fraction (HFpEF). Irrespective of the underlying profibrotic condition (e.g. ageing, diabetes, hypertension), maladaptive cardiac fibrosis is defined by the transformation of resident fibroblasts to matrix-secreting myofibroblasts. Numerous profibrotic factors have been identified at the molecular level (e.g. TGFβ, IL11, AngII), which activate gene expression programs for myofibroblast activation. A number of existing HF therapies indirectly target fibrotic pathways; however, despite multiple clinical trials in HFpEF, a specific clinically effective antifibrotic therapy remains elusive. Therapeutic inhibition of TGFβ, the master-regulator of fibrosis, has unfortunately proven toxic and ineffective in clinical trials to date, and new approaches are needed. In this review, we discuss the pathophysiology and clinical implications of interstitial fibrosis in HFpEF. We provide an overview of trials targeting fibrosis in HFpEF to date and discuss the promise of potential new therapeutic approaches and targets in the context of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Mark Sweeney
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- Wellcome Trust 4i/NIHR Clinical Research FellowImperial CollegeLondonUK
| | - Ben Corden
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Stuart A Cook
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
25
|
McDiarmid AK, Swoboda PP, Erhayiem B, Bounford KA, Bijsterveld P, Tyndall K, Fent GJ, Garg P, Dobson LE, Musa TA, Foley JRJ, Witte KK, Kearney MT, Greenwood JP, Plein S. Myocardial Effects of Aldosterone Antagonism in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2020; 9:e011521. [PMID: 31852424 PMCID: PMC6988171 DOI: 10.1161/jaha.118.011521] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Background Spironolactone may have prognostic benefit in selected patients with heart failure with preserved ejection fraction. This study assessed the myocardial tissue effects of spironolactone in heart failure with preserved ejection fraction. Methods and Results A 1:1 randomized controlled study of 6 months of spironolactone versus control in heart failure with preserved ejection fraction. The primary outcome was change in myocardial extracellular volume fraction by cardiovascular magnetic resonance as a surrogate of diffuse fibrosis. Of 55 randomized patients, 40 (20 women; age, 75.2±5.9 years) completed follow-up (19 treatment, 21 control). A significant change in extracellular volume over the study period was not seen (treatment, 28.7±3.7% versus 27.7±3.4% [P=0.14]; controls, 27.6±3.4% versus 28.3±4.4% [P=0.14]); however, the rate of extracellular volume expansion was decreased by spironolactone (-1.0±2.4% versus 0.8±2.2%). Indexed left ventricular mass decreased with treatment (104.4±26.6 versus 94.0±20.6 g/m2; P=0.001) but not in controls (101.4±29.4 versus 104.0±32.8 g/m2; P=0.111). Extracellular mass decreased by 13.8% (15.1±4.8 versus 13.0±3.4 g/m2; P=0.003), and cellular mass decreased by 8.3% (37.6±10.0 versus 34.3±7.9 g/m2; P=0.001) with spironolactone, but was static in controls. Conclusions Spironolactone did not lead to significant change in extracellular volume. However, spironolactone did decrease rate of extracellular expansion, with a decrease in the mass of both cellular and extracellular myocardial compartments. These data point to the mechanism of action of spironolactone in heart failure with preserved ejection fraction, including a direct tissue effect with a reduction in rate of myocardial fibrosis.
Collapse
Affiliation(s)
- Adam K. McDiarmid
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
- Department of CardiologyFreeman HospitalNewcastle‐upon‐TyneUnited Kingdom
| | - Peter P. Swoboda
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Bara Erhayiem
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | | | - Petra Bijsterveld
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Keith Tyndall
- Leeds Teaching Hospitals NHS TrustLeedsUnited Kingdom
| | - Graham J. Fent
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Pankaj Garg
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Laura E. Dobson
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Tarique A. Musa
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - James R. J. Foley
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Klaus K. Witte
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Mark T. Kearney
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - John P. Greenwood
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre and Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsUnited Kingdom
| |
Collapse
|