1
|
Wang Q, Tang TM, Youlton M, Weldy CS, Kenney AM, Ronen O, Hughes JW, Chin ET, Sutton SC, Agarwal A, Li X, Behr M, Kumbier K, Moravec CS, Tang WHW, Margulies KB, Cappola TP, Butte AJ, Arnaout R, Brown JB, Priest JR, Parikh VN, Yu B, Ashley EA. Epistasis regulates genetic control of cardiac hypertrophy. NATURE CARDIOVASCULAR RESEARCH 2025; 4:740-760. [PMID: 40473955 DOI: 10.1038/s44161-025-00656-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/14/2025] [Indexed: 06/11/2025]
Abstract
Although genetic variant effects often interact nonadditively, strategies to uncover epistasis remain in their infancy. Here we develop low-signal signed iterative random forests to elucidate the complex genetic architecture of cardiac hypertrophy, using deep learning-derived left ventricular mass estimates from 29,661 UK Biobank cardiac magnetic resonance images. We report epistatic variants near CCDC141, IGF1R, TTN and TNKS, identifying loci deemed insignificant in genome-wide association studies. Functional genomic and integrative enrichment analyses reveal that genes mapped from these loci share biological process gene ontologies and myogenic regulatory factors. Transcriptomic network analyses using 313 human hearts demonstrate strong co-expression correlations among these genes in healthy hearts, with significantly reduced connectivity in failing hearts. To assess causality, RNA silencing in human induced pluripotent stem cell-derived cardiomyocytes, combined with novel microfluidic single-cell morphology analysis, confirms that cardiomyocyte hypertrophy is nonadditively modifiable by interactions between CCDC141, TTN and IGF1R. Our results expand the scope of cardiac genetic regulation to epistasis.
Collapse
MESH Headings
- Humans
- Epistasis, Genetic
- Connectin/genetics
- Connectin/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Male
- Genetic Predisposition to Disease
- Deep Learning
- Phenotype
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/diagnostic imaging
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/metabolism
- Female
- Middle Aged
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/diagnostic imaging
- Heart Failure/physiopathology
- Gene Regulatory Networks
- Genome-Wide Association Study
- Induced Pluripotent Stem Cells/metabolism
- Ventricular Remodeling/genetics
- Magnetic Resonance Imaging
- Aged
Collapse
Grants
- 23POST1023278 American Heart Association (American Heart Association, Inc.)
- 23CDA1042900 American Heart Association (American Heart Association, Inc.)
- DGE-2146752 National Science Foundation (NSF)
- IIS 1741340 National Science Foundation (NSF)
- DMS-1613002 National Science Foundation (NSF)
- F32HL160067 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- L30HL159413 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08HL143185 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1R01HL144843 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08HL167699 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01HL105993 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01GM152718 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
Collapse
Affiliation(s)
- Qianru Wang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tiffany M Tang
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Michelle Youlton
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Molecular Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Chad S Weldy
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Ana M Kenney
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
- Department of Statistics, University of California Irvine, Irvine, CA, USA
| | - Omer Ronen
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - J Weston Hughes
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Elizabeth T Chin
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA
| | - Shirley C Sutton
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Abhineet Agarwal
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Xiao Li
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Merle Behr
- Faculty of Informatics and Data Science, University of Regensburg, Regensburg, Germany
| | - Karl Kumbier
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Christine S Moravec
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kenneth B Margulies
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA
| | - Rima Arnaout
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA
| | - James B Brown
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA
- Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - James R Priest
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA
- Tenaya Therapeutics, San Francisco, CA, USA
| | - Victoria N Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Bin Yu
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA.
| | - Euan A Ashley
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Pugliese L, Luciano A, Chiocchi M. The Role of Cardiac Magnetic Resonance Imaging in the Management of Hypertrophic Cardiomyopathy. J Cardiovasc Dev Dis 2025; 12:189. [PMID: 40422960 DOI: 10.3390/jcdd12050189] [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: 04/09/2025] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiomyopathy, caused by either sarcomere protein or other gene mutations. It is a complex and highly heterogeneous disorder, with phenotypes ranging from asymptomatic to severe disease, characterized by asymmetric left ventricular (LV) hypertrophy unexplained by loading conditions, which is also associated with myocardial fiber disarray, and preserved or increased ejection fraction without LV dilation. Comprehensive personal and family history, physical examination, and ECG testing raise suspicion of HCM, and echocardiogram represents the first-line imaging modality for confirming a diagnosis. Moreover, contrast-enhanced cardiac magnetic resonance (CMR) imaging has increasingly emerged as a fundamental diagnostic and prognostic tool in HCM management. This article reviews the role of CMR in HCM identification and differentiation from phenotypic mimics, characterization of HCM phenotypes, monitoring of disease progression, evaluation of pre- and post-septal reduction treatments, and selection of candidates for implantable cardioverter-defibrillator. By providing information on cardiac morphology and function and tissue characterization, CMR is particularly helpful in the quantification of myocardial wall thickness, the detection of hypertrophy in areas blind to echocardiogram, subtle morphologic features in the absence of LV hypertrophy, myocardial fibrosis, and apical aneurysm, the evaluation of LV outflow tract obstruction, and the assessment of LV function in end-stage dilated HCM.
Collapse
Affiliation(s)
- Luca Pugliese
- Department of Medical Surgical Sciences and Translational Medicine, University of Rome La Sapienza, Radiology Unit, Sant'Andrea University Hospital, 00189 Rome, Italy
| | - Alessandra Luciano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Unit of Diagnostic Imaging, Policlinico Tor Vergata, 00133 Rome, Italy
| | - Marcello Chiocchi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Unit of Diagnostic Imaging, Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
3
|
Wenzel JP, Albrecht JN, Toprak B, Petersen E, Nikorowitsch J, Cavus E, Jahnke C, Riedl KA, Adam G, Twerenbold R, Blankenberg S, Kirchhof P, Lund G, Tahir E, Müllerleile K, Radunski UK. Head-to-head comparison of cardiac magnetic resonance imaging and transthoracic echocardiography in the general population (MATCH). Clin Res Cardiol 2025:10.1007/s00392-025-02660-1. [PMID: 40353872 DOI: 10.1007/s00392-025-02660-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 04/21/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Comparing transthoracic echocardiography (TTE) and cardiac magnetic resonance imaging (CMR) is crucial for cardiac assessment. This study aims to clarify their comparability in a large population sample. METHODS CMR and two- (2D) and three-dimensional (3D) TTE were used to quantify left and right heart dimensions in participants of the Hamburg City Health Study. Intertechnique agreement was evaluated using Bland-Altman analyses, Pearson correlation coefficients, and Cohen's Kappa. RESULTS Data from 2126 participants (median age 67 [IQR: 59-72] years, 897 (42.2%) female) were analyzed. Left ventricular (LV) diastolic volumes were similar (CMR: 117.0 [96.2, 138.0] ml, 2D-TTE: 111.8 [93.6, 134.3] ml, r = 0.7, p < 0.001), while systolic volumes were lower with CMR (CMR: 36.0 [26.9, 46.0] ml, 2D-TTE: 46.7 [37.9, 57.5] ml, r = 0.67, p < 0.001). CMR LV ejection fraction (LVEF) was 10% higher than 2D-TTE (CMR = 69.0 [64.0, 74.0]%, 2D-TTE = 58.3 [55.5, 61.7]%, p < 0.001; r = 0.40, p < 0.001). Left atrial volumes correlated moderately with low bias (CMR: 53.0 [40.0, 68.0] ml, 2D-TTE: 51.6 [41.5, 64.0] ml, r = 0.63, p < 0.001). LV mass showed good correlation but was higher using 2D-TTE (r = 0.74, p < 0.001). Right ventricular (RV) volumes showed the largest differences, with CMR demonstrating lower interobserver variability (ICC = 0.97 vs. 0.61 for 2D-TTE) and markedly larger volumes (RVEDV mean bias = 74.8 ml, r = 0.50, p < 0.001). CONCLUSION In a large general population, CMR quantifies cardiac function and dimensions more reliably than TTE. Both modalities provide significantly different absolute values, limiting intertechnique transferability. TRIAL REGISTRATION Retrospectively registered at ClinicalTrial.gov, registration number: NCT03934957, registration date: 04/01/2019, https://clinicaltrials.gov/study/NCT03934957 .
Collapse
Affiliation(s)
- Jan-Per Wenzel
- Clinic for Rhythmology, University Hospital Schleswig-Holstein (UKSH), Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Jan-Niklas Albrecht
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Betül Toprak
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Elina Petersen
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
- Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Julius Nikorowitsch
- Clinic for Rhythmology, University Hospital Schleswig-Holstein (UKSH), Lübeck, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Ersin Cavus
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Charlotte Jahnke
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Katharina Alina Riedl
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Raphael Twerenbold
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
- Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Stefan Blankenberg
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Paulus Kirchhof
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Gunnar Lund
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Enver Tahir
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Müllerleile
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ulf K Radunski
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Clinic Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
4
|
Pan N, Li Z, Xu C, Gao J, Hu H. Hybrid method for automatic initialization and segmentation of ventricular on large-scale cardiovascular magnetic resonance images. BMC Med Imaging 2025; 25:155. [PMID: 40335966 PMCID: PMC12057224 DOI: 10.1186/s12880-025-01683-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Cardiovascular diseases are the number one cause of death globally, making cardiac magnetic resonance image segmentation a popular research topic. Existing schemas relying on manual user interaction or semi-automatic segmentation are infeasible when dealing thousands of cardiac MRI studies. Thus, we proposed a full automatic and robust algorithm for large-scale cardiac MRI segmentation by combining the advantages of deep learning localization and 3D-ASM restriction. MATERIAL AND METHODS The proposed method comprises several key techniques: 1) a hybrid network integrating CNNs and Transformer as a encoder with the EFG (Edge feature guidance) module (named as CTr-HNs) to localize the target regions of the cardiac on MRI images, 2) initial shape acquisition by alignment of coarse segmentation contours to the initial surface model of 3D-ASM, 3) refinement of the initial shape to cover all slices of MRI in the short axis by complex transformation. The datasets used are from the UK BioBank and the CAP (Cardiac Atlas Project). In cardiac coarse segmentation experiments on MR images, Dice coefficients (Dice), mean contour distances (MCD), and mean Hausdorff distances (HD95) are used to evaluate segmentation performance. In SPASM experiments, Point-to-surface (P2S) distances, Dice score are compared between automatic results and ground truth. RESULTS The CTr-HNs from our proposed method achieves Dice coefficients (Dice), mean contour distances (MCD), and mean Hausdorff distances (HD95) of 0.95, 0.10 and 1.54 for the LV segmentation respectively, 0.88, 0.13 and 1.94 for the LV myocardium segmentation, and 0.91, 0.24 and 3.25 for the RV segmentation. The overall P2S errors from our proposed schema is 1.45 mm. For endocardium and epicardium, the Dice scores are 0.87 and 0.91 respectively. CONCLUSIONS Our experimental results show that the proposed schema can automatically analyze large-scale quantification from population cardiac images with robustness and accuracy.
Collapse
Affiliation(s)
- Ning Pan
- College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430074, China
- Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Wuhan, 430074, China
- Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, Wuhan, 430074, China
| | - Zhi Li
- College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430074, China
- Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Wuhan, 430074, China
- Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, Wuhan, 430074, China
| | - Cailu Xu
- College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430074, China
- Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Wuhan, 430074, China
- Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, Wuhan, 430074, China
| | - Junfeng Gao
- College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430074, China
- Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Wuhan, 430074, China
- Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, Wuhan, 430074, China
| | - Huaifei Hu
- College of Biomedical Engineering, South-Central Minzu University, Wuhan, 430074, China.
- Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Wuhan, 430074, China.
- Key Laboratory of Cognitive Science, State Ethnic Affairs Commission, Wuhan, 430074, China.
| |
Collapse
|
5
|
Olesen ASO, Miger K, Sajadieh A, Abild-Nielsen AG, Pedersen L, Schultz HHL, Grand J, Thune JJ, Nielsen OW. Remote dielectric sensing to detect pulmonary congestion in acute dyspnoeic patients: Reproducibility and the effect of pulmonary comorbidities. Int J Cardiol 2025; 425:133068. [PMID: 39956459 DOI: 10.1016/j.ijcard.2025.133068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Remote Dielectric Sensing (ReDS) is a fast and non-invasive method that estimates lung fluid. We previously described moderate accuracies for ReDS to detect acute heart failure in consecutive patients. We hypothesise that unprecise ReDS values may stem from concomitant pulmonary diseases. PURPOSE To examine the ReDS reproducibility and the effect of pulmonary comorbidities on ReDS values in acute dyspnoeic patients. METHODS This prospective observational study included 97 consecutive patients ≥50 years with acute dyspnoea. Upon admission, patients underwent low-dose chest computed tomography (CT), echocardiography and ReDS examination. ReDS is by default performed on the right hemithorax in sitting position. For reproducibility comparisons, we conducted additional ReDS measurements two centimetres above and below the default placement, and in sitting and supine position. Two blinded radiologists evaluated the CT scans for pulmonary congestion and pulmonary diseases. RESULTS Comparing three ReDS measurements on the right hemithorax revealed coefficients of variations of 9.6 %, 8.2 %, and 8.3 %. For sitting versus supine comparison, the coefficient of variation was 9.5 % for the default ReDS placement. Patients with CT-verified pulmonary congestion had a coefficient of variation of 5.9 % in sitting versus supine comparison, while those without had 10.3 %. In multivariable regression, lower ReDS values were observed in patients with pneumonia (-1.81, p = 0.215, N = 51), emphysema (-5.44, p = 0.001, N = 26), and higher in fibrosis (5.58, p = 0.032, N = 8) and congestion (5.79, p = 0.002, N = 17), compared to those without. CONCLUSION ReDS values of lung fluid content and reproducibility were affected by pulmonary diseases. ReDS showed consistent reproducibility for patients with CT-verified pulmonary congestion.
Collapse
Affiliation(s)
- Anne Sophie Overgaard Olesen
- Dept of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Dept of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| | - Kristina Miger
- Dept of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Dept of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Ahmad Sajadieh
- Dept of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Dept of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | | | - Lars Pedersen
- Dept of Respiratory Medicine and Infectious Diseases, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Hans Henrik Lawaetz Schultz
- Dept of Cardiology, Unit of Lung Transplantation, Copenhagen University Hospital - Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Johannes Grand
- Dept of Cardiology, Copenhagen University Hospital - Amager and Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Jens Jakob Thune
- Dept of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Dept of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Olav Wendelboe Nielsen
- Dept of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
6
|
Cheong D, Alloah Q, Fishbein JS, Rajagopal HG. Comparison and Agreement between Cardiovascular Computed Tomography-Derived Mid-Diastolic and End-Diastolic Ventricular Volume in Patients with Congenital Heart Disease. Pediatr Cardiol 2025; 46:844-852. [PMID: 38689021 DOI: 10.1007/s00246-024-03504-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Prospective electrocardiogram (ECG)-triggered cardiovascular computed tomography (CCT) is primarily utilized for anatomical information in congenital heart disease (CHD) and has not been utilized for calculation of the end-diastolic volume (EDV); however, the mid-diastolic volume (MDV) may be measured. The objective of this study was to evaluate the feasibility and agreement between ventricular EDV and MDV. 31 retrospectively ECG-gated CCT were analyzed for the study of the 450 consecutive CCT. CCT images were processed using syngo.via with automatic contouring followed by manual adjustment of the endocardial borders of the left ventricles (LV) and right ventricles (RV) at end-diastolic and mid-diastolic phase (measured at 70% of cardiac cycle). The correlation and agreements between EDV and MDV were demonstrated using Spearman rank coefficient and intraclass correlation coefficient (ICC), respectively. Mean age ± SD was 28.8 ± 12.5 years, 19 were male (61.3%) and tetralogy of Fallot (TOF) was the most common diagnosis (58.1%), 35% (11/31) patients with a pacemaker, ICD or other such contraindication for a CMRI, 23% (7/31) with claustrophobia, and 6.5% (2/31) with developmental delay with refusal for sedation did not have a previous CMRI. The mean ± SD indexed LV EDV and LV MDV were 91.1 ± 24.5 and 84.8 ± 22.3 ml/m2, respectively. The mean ± SD indexed RV EDV and RV MDV were 136.8 ± 41 and 130.2 ± 41.5 ml/m2, respectively. EDV and MDV had a strong positive correlation and good agreement (ICC 0.92 for LV and 0.95 for RV). This agreement was preserved in a subset of patients (21) with dilated RV (indexed RV EDV z-score > 2). Intra-observer reliability (0.97 and 0.98 for LV and RV MDV, respectively) and inter-observer reliability (0.96 and 0.90 for LV and RV MDV, respectively) were excellent. In a select group of patients with CHD, measuring MDV by CCT is feasible and these values have good agreements with EDV. This may be used to derive functional data from prospectively ECG-triggered CCT studies. Further large-scale analysis is needed to determine accuracy and clinical correlation.
Collapse
Affiliation(s)
- Daniel Cheong
- Pediatric Cardiology, Cohen Children's Medical Center, Northwell Health, 2000 Marcus Ave, Suite 300, New Hyde Park, NY, 11042-1069, USA.
| | - Qais Alloah
- Pediatric Cardiology, Cohen Children's Medical Center, Northwell Health, 2000 Marcus Ave, Suite 300, New Hyde Park, NY, 11042-1069, USA
| | - Joanna S Fishbein
- Biostatistics Unit, Office of Academic Affairs, Northwell Health, New Hyde Park, USA
| | - Hari G Rajagopal
- Pediatric Cardiology, Cohen Children's Medical Center, Northwell Health, 2000 Marcus Ave, Suite 300, New Hyde Park, NY, 11042-1069, USA
| |
Collapse
|
7
|
El Mathari S, Bhoera RA, Hopman LHGA, Heidendael J, Malekzadeh A, Nederveen A, van Ooij P, Götte MJW, Kluin J. Disparities in quantification of mitral valve regurgitation between cardiovascular magnetic resonance imaging and trans-thoracic echocardiography: a systematic review. Int J Cardiovasc Imaging 2025; 41:647-658. [PMID: 39499451 PMCID: PMC11982156 DOI: 10.1007/s10554-024-03280-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/27/2024] [Indexed: 11/07/2024]
Abstract
Primary mitral regurgitation (MR) is a prevalent valvular heart disease. Therapy stratification for MR depends on accurate assessment of MR severity and left ventricular (LV) dimensions. While trans-thoracic echocardiography (TTE) has been the standard/preferred assessment method, cardiovascular magnetic resonance imaging (CMR) has gained recognition for its superior assessment of LV dimensions and MR severity. Both imaging modalities have their own advantages and limitation for therapy guidance. However, the differences between the two modalities for assessing/grade severity and clinical impact of MR remains unclear. This systematic review aims to evaluate the differences between TTE and CMR in quantifying MR severity and LV dimensions, providing insights for optimal clinical management. A literature search was performed from inception up to March 21st 2023. This resulted in 2,728 articles. After screening, 22 articles were deemed eligible for inclusion in the meta-analysis. The included study variables were, mitral valve regurgitation volume (MRVOL), regurgitation fraction (MRFRAC), LV end-diastolic volume (LVEDV), LV end-systolic volume (LVESV), LV stroke volume (LVSV), and LV ejection fraction (LVEF). TTE showed a significant higher MRVOL (10.4 ml, I2 = 88%, p = 0.002) and MRFRAC (6.3%, I2 = 51%, p = 0.05) compared to CMR, while CMR demonstrated a higher LVEDV (21.9 ml, I2 = 66%, p = < 0.001) and LVESV (16.8 ml, I2 = 0%, p = < 0.001) compared to TTE. Our findings demonstrate substantial disparities in TTE and CMR derived measurements for parameters that play a pivotal role in the clinical stratification guidelines. This discrepancy prompts a critical question regarding the prognostic value of both imaging modalities, which warrants future research.
Collapse
Affiliation(s)
- Sulayman El Mathari
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| | - Rahul A Bhoera
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Luuk H G A Hopman
- Department of Cardiology, Amsterdam University Medical Center, Room D3-221, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Josephine Heidendael
- Department of Cardiology, Amsterdam University Medical Center, Room D3-221, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arjan Malekzadeh
- Medical Library, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Pim van Ooij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Marco J W Götte
- Department of Cardiology, Amsterdam University Medical Center, Room D3-221, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jolanda Kluin
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Losurdo A, Panico C, Catalano C, Serio S, Giordano L, Monti L, Catapano F, Figliozzi S, D'Andrea C, Dipasquale A, Persico P, Di Muzio A, Cremonesi M, Marchese A, Tronconi MC, Perrino M, Finocchiaro G, Lugli E, Francone M, Santoro A, Condorelli G, Simonelli M, Kallikourdis M. Cardiac MRI study of adverse events in patients treated with immune checkpoint inhibitors: a prospective cohort study of cardiac adverse events. J Immunother Cancer 2025; 13:e010568. [PMID: 40107671 PMCID: PMC11927457 DOI: 10.1136/jitc-2024-010568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) revolutionized cancer therapy, yet require management of immune-related adverse events (irAEs). Fulminant myocarditis is a rare irAE, but lower-severity cardiac events are being reported more frequently, leading to an unmet need for irAE prevention, early diagnosis, and treatment, especially for long-life-expectancy patients. We recruited 57 patients, stratified according to therapy regime (monotherapy (30%) or combination (33%) cohort) or history of cardiac disease or presence of at least two cardiovascular risk factors other than prior or active smoking (cardiovascular cohort (37%)). We performed a complete cardiological assessment with clinical visit, 12-lead ECG, multiparametric cardiac MRI as well as peripheral blood mononuclear cell immunophenotyping, prior to ICI initiation and around 2 months later. ICI treatment was associated with a significant left ventricular ejection function (LVEF) reduction pre-ICI versus post-ICI treatment (60.1±8% to 58.1±8%, p=0.002, paired t-test) and more than 3% LVEF loss in a substantial proportion of patients (18; 32%). These patients also showed significantly higher T2 values (p=0.037, unpaired t-test), putative sign of cardiac edema. The loss of cardiac function did not differ among patients with different tumor types, therapy regimes or history of cardiac disease. Immunophenotyping analyses showed a reduction of programmed cell death protein 1 staining on both CD4+ and CD8+ T cells, and an upregulation of HLA-DR on CD8+ T cells. Using a very sensitive and comprehensive approach in patients unselected for cardiac history, we found a subclinical but significant LVEF decrease. These findings may inform ongoing discussions on optimal management of cardiac irAEs in patients undergoing ICI treatment and warrant further evaluation.
Collapse
Affiliation(s)
- Agnese Losurdo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Cristina Panico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Cardiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Chiara Catalano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Simone Serio
- Cardiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
- Institute of Genetic and Biomedical Research (IRGB), National Research Council of Italy, Milan, Italy
| | - Laura Giordano
- Biostatistics Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Lorenzo Monti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Radiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Federica Catapano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Radiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Stefano Figliozzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Radiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Carla D'Andrea
- Cardiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Angelo Dipasquale
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Pasquale Persico
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Antonio Di Muzio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
| | - Marco Cremonesi
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | | | - Maria Chiara Tronconi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Matteo Perrino
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Giovanna Finocchiaro
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Radiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Cardiology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Adaptive Immunity Laboratory, IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| |
Collapse
|
9
|
Yang X, Liu H, Wu X. High-altitude pulmonary hypertension: a comprehensive review of mechanisms and management. Clin Exp Med 2025; 25:79. [PMID: 40063280 PMCID: PMC11893705 DOI: 10.1007/s10238-025-01577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/26/2025] [Indexed: 03/14/2025]
Abstract
High-altitude pulmonary hypertension (HAPH) is characterized by an increase in pulmonary artery pressure due to prolonged exposure to hypoxic environment at high altitudes. The development of HAPH involves various factors such as pressure changes, inflammation, oxidative stress, gene regulation, and signal transduction. The pathophysiological mechanisms of this condition operate at molecular, cellular, and genetic levels. Diagnosis of HAPH often relies on echocardiography, cardiac catheterization, and other methods to assess pulmonary artery pressure and its impact on cardiac function. Treatment options for HAPH encompass both nondrug and drug therapies. While advancements have been made in understanding the pathological mechanisms through research on animal models and clinical trials, there are still limitations to be addressed. Future research should focus on exploring molecular targets, personalized medicine, long-term management strategies, and interdisciplinary approaches. By leveraging advanced technologies like systems biology, omics technology, big data, and artificial intelligence, a comprehensive analysis of HAPH pathogenesis can lead to the identification of new treatment targets and strategies, ultimately enhancing patient quality of life and prognosis. Furthermore, research on health monitoring and preventive measures for populations living at high altitudes should be intensified to reduce the incidence and mortality of HAPH.
Collapse
Affiliation(s)
- Xitong Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Hong Liu
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Xinhua Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China.
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China.
| |
Collapse
|
10
|
Gentile F, Chianca M, Bazan L, Sciarrone P, Chubuchny V, Taddei C, Poggianti E, Passino C, Emdin M, Giannoni A. Incremental Prognostic Value of Echocardiography Measures of Right Ventricular Systolic Function in Patients With Chronic Heart Failure. J Am Heart Assoc 2025; 14:e038616. [PMID: 39968776 DOI: 10.1161/jaha.124.038616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Tricuspid annular plane systolic excursion (TAPSE), Doppler tissue imaging-derived tricuspid lateral annular systolic wave velocity (S'), and right ventricular fractional area change (RV-FAC) are the most widely used echocardiographic measures of right ventricular systolic function. This study aimed to compare the prognostic value of TAPSE, S', and RV-FAC in a large cohort of patients with chronic heart failure. METHODS Consecutive outpatients with heart failure and left ventricular ejection fraction <50% on guideline-recommended therapies undergoing echocardiography were followed up for the end point of cardiac and all-cause death. RESULTS Among 1590 patients (71±12 years, 77% men, left ventricular ejection fraction 34%±9%), 202 (13%) died from cardiac causes during a median follow-up of 28 (interquartile range, 14-40) months. According to the recommended cut points for TAPSE (<17 mm), S' (<9.5 cm/s), or RV-FAC (<35%), right ventricular systolic dysfunction was found in 37%, 40%, and 35% of patients, respectively, with 21%, 31%, and 33% of discordant cases comparing TAPSE versus S', TAPSE versus RV-FAC, and S' versus RV-FAC. Both TAPSE <17 mm and RV-FAC <35% were more accurate than S' <9.5 cm/s in predicting the risk of cardiac death (P<0.001), and their combination showed incremental prognostic power (P<0.001). Adding S' to the combination of TAPSE and RV-FAC did not provide further incremental value (P=0.145). Similar findings were obtained when all-cause death was considered as the end point. CONCLUSIONS In patients with chronic heart failure and left ventricular ejection fraction <50%, TAPSE, and RV-FAC are more accurate than S' in predicting the risk of cardiac and all-cause death. Considering both RV-FAC and TAPSE provides incremental prognostic value.
Collapse
Affiliation(s)
- Francesco Gentile
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
- Fondazione Toscana Gabriele Monasterio Pisa Italy
| | - Michela Chianca
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
| | - Lorenzo Bazan
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
| | - Paolo Sciarrone
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
- Fondazione Toscana Gabriele Monasterio Pisa Italy
| | | | | | | | - Claudio Passino
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
- Fondazione Toscana Gabriele Monasterio Pisa Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
- Fondazione Toscana Gabriele Monasterio Pisa Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center Scuola Superiore Sant'Anna Pisa Italy
- Fondazione Toscana Gabriele Monasterio Pisa Italy
| |
Collapse
|
11
|
Rowe SJ, Bekhuis Y, Mitchell A, Janssens K, D'Ambrosio P, Spencer LW, Paratz ED, Claessen G, Fatkin D, La Gerche A. Genetics, Fitness, and Left Ventricular Remodelling: The Current State of Play. Can J Cardiol 2025; 41:364-374. [PMID: 39681159 DOI: 10.1016/j.cjca.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiorespiratory fitness (CRF) exists on a spectrum and is driven by a constellation of factors, including genetic and environmental differences. This results in wide interindividual variation in baseline CRF and the ability to improve CRF with regular endurance exercise training. As opposed to monogenic conditions, CRF is described as a complex genetic trait as it is believed to be influenced by multiple common genetic variants in addition to exogenous factors. Importantly, CRF is an independent predictor of morbidity and mortality, and so understanding the impact of genetic variation on CRF may provide insights into both human athletic performance and personalized risk assessment and prevention. Despite rapidly advancing technology, progress in this field has been restricted by small sample sizes and the limited number of genetic studies using the "gold standard" objective measure of peak oxygen consumption (VO2peak) for CRF assessment. In recent years, there has been increasing interest in the heritability of numerous parameters of cardiac structure and function and how this may relate to both normal cardiac physiology and disease pathology. Regular endurance training can result in exercise-induced cardiac remodelling, which manifests as balanced dilation of cardiac chambers and is associated with superior CRF. This results in a complex relationship between CRF, cardiac size, and exercise, and whether shared genetic pathways may influence this remains unknown. In this review we highlight recent and relevant studies into the genomic predictors of CRF with a unique emphasis on how this may relate to cardiac remodelling and human adaptation to endurance exercise.
Collapse
Affiliation(s)
- Stephanie J Rowe
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia. https://twitter.com/_sjrowe
| | - Youri Bekhuis
- Department of Cardiology and Jessa & Science, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences/LCRC, UHasselt, Diepenbeek, Belgium; Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium. https://twitter.com/YouriBekhuis
| | - Amy Mitchell
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Kristel Janssens
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Exercise and Nutrition Research Program, The Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Paolo D'Ambrosio
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia; Cardiology Department, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Luke W Spencer
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth D Paratz
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia. https://twitter.com/pretzeldr
| | - Guido Claessen
- Department of Cardiology and Jessa & Science, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences/LCRC, UHasselt, Diepenbeek, Belgium; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium. https://twitter.com/KJanssensAU
| | - Diane Fatkin
- Cardiology Department, St Vincent's Hospital, Darlinghurst, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Kensington, New South Wales, Australia; Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Andre La Gerche
- Heart, Exercise and Research Trials, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria, Australia; Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
12
|
Kramer CM, Borlaug BA, Zile MR, Ruff D, DiMaria JM, Menon V, Ou Y, Zarante AM, Hurt KC, Murakami M, Packer M. Tirzepatide Reduces LV Mass and Paracardiac Adipose Tissue in Obesity-Related Heart Failure: SUMMIT CMR Substudy. J Am Coll Cardiol 2025; 85:699-706. [PMID: 39566869 DOI: 10.1016/j.jacc.2024.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Obesity is a known risk factor for heart failure with preserved ejection fraction (HFpEF) and is considered a distinct phenotype with more concentric remodeling. Epicardial adipose tissue (EAT) is also increased in obesity-related HFpEF and is associated with adverse events. OBJECTIVES The cardiac magnetic resonance (CMR) substudy of the SUMMIT trial aimed to examine the effects of tirzepatide on cardiac structure and function with the underlying hypothesis that it would reduce left ventricular (LV) mass and EAT in obesity-related HFpEF. METHODS A total of 175 patients with obesity-related HFpEF from the parent study of tirzepatide (2.5 mg subcutaneously weekly, increasing to a maximum of 15 mg weekly) or matching placebo underwent CMR at baseline, which consisted of multiplanar cine imaging. A total of 106 patients completed the CMR and had adequate image quality for analysis of LV and left atrial structure and function and paracardiac (epicardial plus pericardial) adipose tissue at both baseline and 52 weeks. The prespecified primary endpoint of this substudy was between-group changes in LV mass. RESULTS LV mass decreased by 11 g (95% CI: -19 to -4 g) in the treated group (n = 50) when corrected for placebo (n = 56) (P = 0.004). Paracardiac adipose tissue decreased in the treated group by 45 mL (95% CI: -69 to -22 mL) when corrected for placebo (P < 0.001). The change in LV mass in the treated group correlated with changes in body weight (P < 0.02) and tended to correlate with changes in waist circumference and blood pressure (P = 0.06 for both). The LV mass change also correlated with changes in LV end-diastolic volume and left atrial end-diastolic and end-systolic volumes (P < 0.03 for all). CONCLUSIONS The CMR substudy of the SUMMIT trial demonstrated that tirzepatide therapy in obesity-related HFpEF led to reduced LV mass and paracardiac adipose tissue as compared with placebo, and the change in LV mass paralleled weight loss. These physiologic changes may contribute to the reduction in heart failure events seen in the main SUMMIT trial. (A Study of Tirzepatide [LY3298176] in Participants With Heart Failure With Preserved Ejection Fraction [HFpEF] and Obesity: The SUMMIT Trial; NCT04847557).
Collapse
Affiliation(s)
- Christopher M Kramer
- Cardiovascular Division, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA.
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael R Zile
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Dustin Ruff
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Joseph M DiMaria
- Cardiovascular Division, Department of Medicine, University of Virginia Health, Charlottesville, Virginia, USA
| | - Venu Menon
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yang Ou
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Karla C Hurt
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, Texas, USA; Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Green PG, Watson WD, Bussmann BM, De Maria GL, Neubauer S, Lewis AJM, Rider OJ, Herring N. Metabolic flexibility and reverse remodelling of the failing human heart. Eur Heart J 2025:ehaf033. [PMID: 39998427 DOI: 10.1093/eurheartj/ehaf033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/12/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND AND AIMS Cardiac resynchronization therapy (CRT) produces long-term reverse remodelling which requires greater adenosine triphosphate delivery to the contractile machinery. Whilst the heart retains some metabolic flexibility in non-ischaemic cardiomyopathy, whether this correlates with reverse remodelling is unknown. This study investigated whether CRT acutely changes cardiac substrate uptake, and whether this translates to favourable reverse remodelling. METHODS The effect of CRT on cardiac substrate uptake was assessed via direct coronary flow and arteriovenous measurements, with metabolomic/lipidomic analysis on infusions of insulin/glucose and intralipid. Cardiac function was assessed with left ventricular pressure-volume loops during implantation, and cardiac magnetic resonance before and 6 months following CRT, with and without biventricular pacing. RESULTS Regardless of substrate infusion, CRT acutely improved stroke work without increasing O2 uptake on both insulin/glucose (by 34%, P = .05) and intralipid (by 36%, P = .03). This was followed by increased fatty acid (FA) uptake on insulin/glucose (R = 0.89, P = .03) and increased β-hydroxybutyrate uptake (R = 0.81, P = .05) during intralipid infusion. After 6 months, there was a 48% (P < .001) reduction in left ventricular end diastolic volume, beyond that achievable by acutely shortening or lengthening QRS duration. Reverse remodelling significantly correlated with increased FA uptake with CRT on insulin/glucose (R = 0.71, P = .05) driven by long and medium chain uptake, and increased ketone uptake with CRT on intralipid (R = 0.79, P = .05). CONCLUSIONS CRT acutely alters the metabolic phenotype of non-ischaemic cardiomyopathy towards a more physiological picture of FA uptake which correlates with reverse remodelling. Retained metabolic flexibility may therefore be critical for subsequent reverse remodelling.
Collapse
Affiliation(s)
- Peregrine G Green
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - William D Watson
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Benjamin M Bussmann
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Giovanni Luigi De Maria
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Andrew J M Lewis
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
14
|
Ma J, Zhu X, Kaushik S, Ali E, Li L, Manickam K, Li K, Janich MA. Qualitative and Quantitative Evaluation of a Deep Learning-Based Reconstruction for Accelerated Cardiac Cine Imaging. Bioengineering (Basel) 2025; 12:231. [PMID: 40150695 PMCID: PMC11939508 DOI: 10.3390/bioengineering12030231] [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: 12/31/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Two-dimensional (2D) cine imaging is essential in routine clinical cardiac MR (CMR) exams for assessing cardiac structure and function. Traditional cine imaging requires patients to hold their breath for extended periods and maintain consistent heartbeats for optimal image quality, which can be challenging for those with impaired breath-holding capacity or irregular heart rhythms. This study aims to systematically assess the performance of a deep learning-based reconstruction (Sonic DL Cine, GE HealthCare, Waukesha, WI, USA) for accelerated cardiac cine acquisition. Multiple retrospective experiments were designed and conducted to comprehensively evaluate the technique using data from an MR-dedicated extended cardiac torso anatomical phantom (digital phantom) and healthy volunteers on different cardiac planes. Image quality, spatiotemporal sharpness, and biventricular cardiac function were qualitatively and quantitatively compared between Sonic DL Cine-reconstructed images with various accelerations (4-fold to 12-fold) and fully sampled reference images. Both digital phantom and in vivo experiments demonstrate that Sonic DL Cine can accelerate cine acquisitions by up to 12-fold while preserving comparable SNR, contrast, and spatiotemporal sharpness to fully sampled reference images. Measurements of cardiac function metrics indicate that function measurements from Sonic DL Cine-reconstructed images align well with those from fully sampled reference images. In conclusion, this study demonstrates that Sonic DL Cine is able to reconstruct highly under-sampled (up to 12-fold acceleration) cine datasets while preserving SNR, contrast, spatiotemporal sharpness, and quantification accuracy for cardiac function measurements. It also provides a feasible approach for thoroughly evaluating the deep learning-based method.
Collapse
Affiliation(s)
- Junjie Ma
- GE HealthCare, Jersey City, NJ 07302, USA
| | | | | | - Eman Ali
- GE HealthCare, 80807 Munich, Germany
| | | | | | - Ke Li
- GE HealthCare, Waukesha, WI 53188, USA
| | | |
Collapse
|
15
|
Labib D, Haykowsky M, Sonnex E, Mackey JR, Thompson RB, Paterson DI, Pituskin E. Long-term cardiac MRI follow up of MANTICORE (Multidisciplinary Approach to Novel Therapies in Cardio-Oncology REsearch). CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:13. [PMID: 39923094 PMCID: PMC11806551 DOI: 10.1186/s40959-025-00313-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND This study investigates the long-term cardiac effects of trastuzumab-based chemotherapy in early breast cancer (EBC) survivors. We extend the original MANTICORE trial which showed that angiotensin-converting enzyme inhibitors (ACEI) and beta-blockers (BB) could mitigate the decline in left ventricular (LV) ejection fraction (EF) during the first year of trastuzumab treatment. OBJECTIVES We hypothesized that, over time, cardiac function would decline further and adverse changes in cardiac geometry would occur due to the aging of the population and prior treatment. METHODS The study enrolled 52 participants from the original MANTICORE trial cohort, with cardiac magnetic resonance (CMR) imaging conducted at a median of 6.5 years post randomization to treatment. RESULTS We found that, contrary to the hypothesis, participants maintained LV EF over the follow-up period. Specifically, the placebo group exhibited a recovery in LV EF to levels comparable with the treatment groups, suggesting no long-term differential impact on cardiac function. However, a significant reduction in LV mass was observed across all groups, the clinical implications of which remain unclear. CONCLUSIONS The findings suggest that in a selected population receiving trastuzumab-based chemotherapy, extended cardiac imaging surveillance beyond one-year post-treatment may be unnecessary. We posit that the presence of HER2 overexpressing breast cancer influenced hypertrophic changes to cardiac geometry observed at baseline and one year, which resolved after completing HER2-blocking treatment. The study also highlights the need for further research to understand the significance of changes in cardiac geometry during and after breast cancer treatment.
Collapse
Affiliation(s)
- Dina Labib
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Calgary, Canada
- Department of Cardiovascular Medicine, Cairo University, Cairo, Egypt
| | - Mark Haykowsky
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada
| | | | | | - Richard B Thompson
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - D Ian Paterson
- University of Ottawa Heart Institute, Ottawa Ontario, Canada
| | - Edith Pituskin
- Faculty of Nursing, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
16
|
Robles-Mezcua A, Januzzi JL, Pavón-Morón FJ, Rodríguez-Capitán J, López-Garrido MA, Cruzado-Álvarez C, Robles-Mezcua M, Gutiérrez-Bedmar M, Couto-Mallón D, Rueda-Calle EC, Barreiro-Pérez M, Sánchez PL, Gómez-Doblas JJ, Jiménez-Navarro MF, García-Pinilla JM. Effects of sacubitril/valsartan on cardiac remodeling in heart failure with reduced ejection fraction: An integrated study of molecular biomarkers and imaging techniques. Biomed Pharmacother 2025; 183:117874. [PMID: 39889435 DOI: 10.1016/j.biopha.2025.117874] [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: 09/05/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Treatment of heart failure and reduced ejection fraction (HFrEF) using angiotensin receptor-neprilysin inhibitor demonstrates beneficial effects on cardiac remodeling (CR). We assessed the impact of sacubitril/valsartan on the concentrations of HF biomarkers in relation to parameters of CR using imaging techniques in patients with HFrEF. In a prospective single-center open-label study, 68 patients with symptomatic HFrEF were treated with sacubitril/valsartan and followed-up every three months for 12 months. Soluble suppression of tumorigenicity 2 (sST2), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and high-sensitivity cardiac troponin I (hs-cTnI) were measured in blood samples. Additionally, echocardiography and cardiac magnetic resonance imaging (cMRI) were performed to assess heart structural and functional changes. Following treatment initiation, follow-up visits revealed an improved NYHA functional class in these patients, alongside significant decreases in all circulating biomarkers, increases in left ventricular ejection fraction (LVEF), and reductions in volume- and diameter-related LV parameters. Sustained gradual decreases in sST2 concentrations over time correlated with NT-proBNP concentrations (rho=+0.26, P < 0.001). Both biomarkers were inversely correlated with LVEF, and positively correlated with volume- and diameter-related LV parameters from echocardiography and cMRI. However, NT-proBNP concentrations exhibited stronger correlations with these LV parameters and were associated with the number of LV segments showing fibrosis, unlike sST2. Sacubitril/valsartan treatment in HFrEF leads to reduced sST2 and NT-proBNP concentrations with distinct decreasing curves, which are linked to reverse CR through LV-related parameters. In contrast to sST2, NT-proBNP is also associated with fibrosis, suggesting that both biomarkers unveil distinct mechanisms during CR in patients treated with sacubitril/valsartan.
Collapse
Affiliation(s)
- Ainhoa Robles-Mezcua
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Unidad de Insuficiencia Cardíaca y Cardiopatías Familiares, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina y Dermatología, Facultad de Medicina, Universidad de Málaga, Spain
| | - James L Januzzi
- Massachusetts General Hospital, Harvard Medical School and Baim Institute for Clinical Research, Boston, MA, USA
| | - Francisco Javier Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| | - Jorge Rodríguez-Capitán
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Concepción Cruzado-Álvarez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Unidad de Insuficiencia Cardíaca y Cardiopatías Familiares, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - María Robles-Mezcua
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Unidad de Insuficiencia Cardíaca y Cardiopatías Familiares, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Mario Gutiérrez-Bedmar
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina Preventiva y Salud Pública, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - David Couto-Mallón
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Cardiología, Complejo Hospitalario Universitario A Coruña (CHUAC), Instituto Investigación Biomédica A Coruña (INIBIC), A Coruña, Spain
| | - Eloy C Rueda-Calle
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Manuel Barreiro-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Cardiología, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Pedro L Sánchez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Cardiología, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Facultad de Medicina, Universidad de Salamanca (USAL), Salamanca, Spain
| | - Juan José Gómez-Doblas
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina y Dermatología, Facultad de Medicina, Universidad de Málaga, Spain
| | - Manuel F Jiménez-Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina y Dermatología, Facultad de Medicina, Universidad de Málaga, Spain.
| | - José M García-Pinilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Unidad de Insuficiencia Cardíaca y Cardiopatías Familiares, Hospital Universitario Virgen de la Victoria, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Medicina y Dermatología, Facultad de Medicina, Universidad de Málaga, Spain
| |
Collapse
|
17
|
Rowe SJ, Xiang R, Paratz ED, Takeuchi F, La Gerche A. Left ventricular size and heart failure: A cardiac MRI assessment of 38,129 individuals from the UK Biobank. Int J Cardiol 2025; 419:132687. [PMID: 39490584 DOI: 10.1016/j.ijcard.2024.132687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Previous studies suggest that prevalent heart failure (HF) differs based on left ventricular ejection fraction (LVEF) and left ventricular (LV) chamber size. Furthermore, the prevalence of HF with preserved ejection fraction (HFpEF) is often considered approaching, or exceeding that of HF with reduced ejection fraction in the community. AIM The aim of this study was to evaluate prevalent and incident HF based on LVEF and CMR-determined LV size within a large community-dwelling cohort. METHODS Individuals from the United Kingdom Biobank (UKB) who underwent CMR and had available health record linkage to allow ascertainment of HF diagnosis were included. The cohort was analysed according to LVEF, LV end-diastolic volume (LVEDV) quartiles and LVEDV indexed to body surface area (LVEDVi). RESULTS 38,129 individuals were included, comprising those with reduced LVEF (LVEF<50 %, n = 5096) and preserved LVEF (LVEF 50-60 %, n = 22,907, LVEF≥60 %, n = 10,126). Prevalent HF was highest in males with LVEF<50 %, and participants with reduced LVEF had higher rates of incident HF (p < 0.001) during the follow-up period (median = 2.46 years from CMR). Mean LVEDV and LVEDVi were largest in individuals with EF < 50 % (146.9 ± 36.2 ml and 76.8 ± 16.4 ml/m2 respectively). Compared to the smallest quartiles, the largest quartiles for LVEDV were associated with increased odds of HF (odds ratio 2.14 [95 % confidence interval 1.47-3.12], p < 0.001). CONCLUSIONS Over 50 % of HF cases occur in individuals with LVEF ≥50 %, however HF prevalence is highest in those with reduced LVEF, particularly in males. Larger LV size is associated with increased HF across the LVEF spectrum.
Collapse
Affiliation(s)
- Stephanie J Rowe
- Heart, Exercise and Research Trials (HEART) Lab, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Cardiology, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| | - Ruidong Xiang
- The University of Melbourne, Parkville, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Elizabeth D Paratz
- Heart, Exercise and Research Trials (HEART) Lab, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Cardiology, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia
| | - Fumihiko Takeuchi
- Department of Cardiology, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Andre La Gerche
- Heart, Exercise and Research Trials (HEART) Lab, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Cardiology, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia; Victor Chang Cardiovascular Research Institute, Darlinghurst, Australia
| |
Collapse
|
18
|
Kelle S, Nolden AC, Müller ML, Beyer RE, Steen H, Remppis BA, Wieditz J, Kentenich H, Tuit A, Cvetkovic M, Witt UE, André F, Schmidt S, Huppertz A, Simic D, Müller D, Shukri A, Issing M, Glardon A, Reber KC, Landmesser U, Frey N, Pieske B, Stock S, Falk V, Friede T, Thiede G. Rationale and design of the HERZCHECK trial: Detection of early heart failure using telemedicine and cardiovascular magnetic resonance in structurally weak regions (NCT05122793). J Cardiovasc Magn Reson 2025; 27:101841. [PMID: 39824461 PMCID: PMC11870244 DOI: 10.1016/j.jocmr.2025.101841] [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: 10/15/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND AND AIMS Heart failure (HF) is an imminent global health problem. Yet established screening algorithms for asymptomatic pre-HF, allowing for early and effective preventive interventions, are largely lacking. The HERZCHECK trial, conducted in structurally underserved rural regions of North-Eastern Germany, aims to close this gap by evaluating the feasibility, diagnostic efficacy, and cost-effectiveness of a fully mobile, telemedically-supervised screening approach, combining cardiovascular magnetic resonance (CMR) imaging and laboratory testing as central elements. STUDY DESIGN AND METHODOLOGY The HERZCHECK trial is a prospective, randomized controlled trial employing a prospective randomized open blinded endpoint design. The study targets asymptomatic adults aged 40-69 years without a history of HF, but with at least one of the following cardiovascular risk factors: hypertension, hypercholesterolemia, obesity, smoking/tobacco consumption, chronic diabetes mellitus, or chronic kidney disease. Participants undergo a comprehensive screening examination including a questionnaire-based medical history, laboratory testing, and CMR at baseline. Based on CMR-derived global longitudinal strain (GLS), participants are classified as stratum A (GLS < -15%), B (GLS ≥ -15% to < -11%), or C (GLS ≥ -11%), with strata B and C being defined as asymptomatic pre-HF. Ten percent of participants in stratum A and all of stratum B and C are subsequently randomized into two groups, receiving either conventional or innovative medical reports, the latter including information on GLS, guideline-based recommendations, and access to a lifestyle intervention app for cardiovascular prevention. Additionally, treating physicians of participants in the innovative group are granted access to an expert center for telemedical inquiries. Follow-up assessments are performed over 12 months to evaluate changes in GLS, as well as adverse cardiac events and quality of life. CONCLUSION HERZCHECK aims to provide a blueprint for a comprehensive, contemporary screening approach tailored to the needs of the targeted structurally underserved population. By implementing this approach in a representative at-risk cohort, HERZCHECK will provide important new information about (a) the prevalence of asymptomatic pre-HF in at-risk patients and (b) the feasibility, added diagnostic value and health economic aspects of CMR exams as part of future screening mechanisms for HF in clinical routine care (NCT05122793).
Collapse
Affiliation(s)
- Sebastian Kelle
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| | - Anna Clara Nolden
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Maximilian Leo Müller
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Rebecca Elisabeth Beyer
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Henning Steen
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | | | - Johannes Wieditz
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Hannah Kentenich
- Institute for Health Economics and Clinical Epidemiology (IGKE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alex Tuit
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mina Cvetkovic
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Undine Ella Witt
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany; Herzinstitut Berlin, Kardiologische Gemeinschaftspraxis, Berlin, Germany
| | - Florian André
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sein Schmidt
- Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Neurology, Charité- Universitätsmedizin Berlin, Berlin, Germany; Center for Stroke Research (CSB), Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Huppertz
- University of Potsdam, University Outpatient Clinic, Sports Medicine and Sports Orthopaedics, Potsdam, Germany
| | - Dusan Simic
- Institute for Health Economics and Clinical Epidemiology (IGKE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Dirk Müller
- Institute for Health Economics and Clinical Epidemiology (IGKE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Arim Shukri
- Institute for Health Economics and Clinical Epidemiology (IGKE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | - Ulf Landmesser
- Department of Cardiology, Angiology, and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Burkert Pieske
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Division of Cardiology, University Medicine Rostock, Rostock, Germany
| | - Stephanie Stock
- Institute for Health Economics and Clinical Epidemiology (IGKE), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Volkmar Falk
- Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Lower Saxony, Göttingen, Germany
| | - Gisela Thiede
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Berlin, Germany; Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
19
|
Iakovlev N, Schiffers FA, Tapia SL, Shen D, Hong K, Markl M, Lee DC, Katsaggelos AK, Kim D. Computationally Efficient Implicit Training Strategy for Unrolled Networks (IMUNNE): A Preliminary Analysis Using Accelerated Real-Time Cardiac Cine MRI. IEEE Trans Biomed Eng 2025; 72:187-197. [PMID: 39141476 PMCID: PMC11825888 DOI: 10.1109/tbme.2024.3443635] [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] [Indexed: 08/16/2024]
Abstract
OBJECTIVE Highly-undersampled, dynamic MRI reconstruction, particularly in multi-coil scenarios, is a challenging inverse problem. Unrolled networks achieve state-of-the-art performance in MRI reconstruction but suffer from long training times and extensive GPU memory cost. METHODS In this work, we propose a novel training strategy for IMplicit UNrolled NEtworks (IMUNNE) for highly-undersampled, multi-coil dynamic MRI reconstruction. It formulates the MRI reconstruction problem as an implicit fixed-point equation and leverages gradient approximation for backpropagation, enabling training of deep architectures with fixed memory cost. This study represents the first application of implicit network theory in the context of real-time cine MRI. The proposed method is evaluated using a prospectively undersampled, real-time cine dataset using radial k-space sampling, comprising balanced steady-state free precession (b-SSFP) readouts. Experiments include a hyperparameter search, head-to-head comparisons with a complex U-Net (CU-Net) and an alternating unrolled network (Alt-UN), and an analysis of robustness under noise perturbations; peak signal-to-noise ratio, structural similarity index, normalized root mean-square error, spatio-temporal entropic difference, and a blur metric were used. RESULTS IMUNNE produced significantly and slightly better image quality compared to CU-Net and Alt-UN, respectively. Compared with Alt-UN, IMUNNE significantly reduced training and inference times, making it a promising approach for highly-accelerated, multi-coil real-time cine MRI reconstruction. CONCLUSION IMUNNE strategy successfully applies unrolled networks to image reconstruction of highly-accelerated, real-time radial cine MRI. SIGNIFICANCE Implicit training enables rapid, high-quality, and cost-effective CMR exams by reducing training and inference times and lowering memory cost associated with advanced reconstruction methods.
Collapse
|
20
|
Tang XL, Wysoczynski M, Gumpert AM, Solanki M, Li Y, Wu WJ, Zheng S, Ruble H, Li H, Stowers H, Zheng S, Ou Q, Tanveer N, Slezak J, Kalra DK, Bolli R. Intravenous infusions of mesenchymal stromal cells have cumulative beneficial effects in a porcine model of chronic ischaemic cardiomyopathy. Cardiovasc Res 2024; 120:1939-1952. [PMID: 39163570 PMCID: PMC11630033 DOI: 10.1093/cvr/cvae173] [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: 01/26/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
AIMS The development of cell therapy as a widely available clinical option for ischaemic cardiomyopathy is hindered by the invasive nature of current cell delivery methods. Furthermore, the rapid disappearance of cells after transplantation provides a cogent rationale for using repeated cell doses, which, however, has not been done thus far in clinical trials because it is not feasible with invasive approaches. The goal of this translational study was to test the therapeutic utility of the intravenous route for cell delivery. METHODS AND RESULTS Pigs with chronic ischaemic cardiomyopathy induced by myocardial infarction received one or three intravenous doses of allogeneic bone marrow mesenchymal stromal cells (MSCs) or placebo 35 days apart. Rigour guidelines, including blinding and randomization, were strictly followed. A comprehensive assessment of left ventricular (LV) function was conducted with three independent methods (echocardiography, magnetic resonance imaging, and haemodynamic studies). The results demonstrate that three doses of MSCs improved both load-dependent and independent indices of LV function and reduced myocardial hypertrophy and fibrosis; in contrast, one dose failed to produce most of these benefits. CONCLUSIONS To our knowledge, this is the first study to show that intravenous infusion of a cell product improves LV function and structure in a large animal model of chronic ischaemic cardiomyopathy and that repeated infusions are necessary to produce robust effects. This study, conducted in a clinically relevant model, supports a new therapeutic strategy based on repeated intravenous infusions of allogeneic MSCs and provides a foundation for a first-in-human trial testing this strategy in patients with chronic ischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Anna M Gumpert
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Mitesh Solanki
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Yan Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shirong Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Halina Ruble
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Hong Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Heather Stowers
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shengnan Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Nida Tanveer
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Bratislava, Slovakia
| | - Dinesh K Kalra
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| |
Collapse
|
21
|
Zheng A, Adam R, Peebles C, Harden S, Shambrook J, Abbas A, Vedwan K, Adam G, Haydock P, Cowburn P, Young C, Long J, Walkden M, Smith S, Greenwood E, Olden P, Flett A. Effect of optimisation to contemporary HFrEF medical therapy with sacubitril/valsartan (Entresto) and dapaglifloziN on left Ventricular reverse remodelling as demonstrated by cardiac magnetic resonance (CMR) Imaging: the ENVI study. Open Heart 2024; 11:e002933. [PMID: 39622578 PMCID: PMC11624772 DOI: 10.1136/openhrt-2024-002933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024] Open
Abstract
INTRODUCTION Heart failure with reduced ejection fraction (HFrEF) guidelines recommend 'four pillars' of medical therapy and device therapy if left ventricular ejection fraction (LVEF) remains ≤35% after 3 months optimum medical therapy.We conducted the first study to examine the effects of optimisation to contemporary medical therapy on cardiac reverse remodelling, as demonstrated by cardiac magnetic resonance imaging (CMR).We hypothesised a proportion of patients would undergo beneficial remodelling and LVEF improvement above the threshold for complex device prescription after 6 months. METHODS HFrEF patients with symptomatic LVEF≤35% despite ACE inhibitor/beta blocker/mineralocorticoid receptor antagonist therapy, and qualified for sacubitril/valsartan switchover were recruited to this single centre prospective study.CMR was performed at baseline and at follow-up. Clinical, volumetric and outcome data were collected and compared. RESULTS Between June 2021 and August 2022, 49 patients were recruited. The majority (80%) were male, mean age 63±14 years. 35 (71%) had non-ischaemic cardiomyopathy. 2 (4%) patients died and 47 were followed up for a median of 7.4 months. There were no heart failure hospitalisations.Significant reductions were seen in median indexed left atrial volume: 54 mL/m2 (41-72) to 39 mL/m2 (30-60) (p<0.001); indexed left ventricular end-diastolic volume: 109 mL/m2 (74-125) to 76 mL/m2 (58-102) (p<0.001); indexed left ventricular end-systolic volume: 74mL/m2 (50-92) to 43 mL/m2 (27-58) (p<0.001) and mean indexed left ventricular mass: 72±13 g/m2 to 62±13 g/m2 (p<0.001).Median LVEF increased by 12 points from 31% to 43% (p<0.001). 29 (59%) patients improved to LVEF>35%. 13 (27%) patients improved to LVEF≥50%.Median N-terminal pro B type natriuretic peptide (NTproBNP) reduced from 883 ng/L (293-2043) to 429 ng/L (171-1421) (p<0.001). CONCLUSIONS Optimisation to contemporary HFrEF medical therapy results in beneficial cardiac reverse remodelling and significant improvements in LVEF and NTproBNP at 6 months as demonstrated by CMR. 59% of our cohort no longer met complex device indications. Guidelines suggest re-assessment of LVEF at 3 months, but our data suggests a longer period is required. TRIAL REGISTRATION NUMBER NCT05348226.
Collapse
Affiliation(s)
- Alice Zheng
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Robert Adam
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Charles Peebles
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Stephen Harden
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James Shambrook
- Radiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ausami Abbas
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Katharine Vedwan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Georgina Adam
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Paul Haydock
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Peter Cowburn
- Cardiology, University Hospital Southampton, Southampton, UK
| | - Christopher Young
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jane Long
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Michelle Walkden
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Simon Smith
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Paula Olden
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Flett
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
22
|
Vinco G, Porto MD, Demattè C, Giovanelli C, Caruso F, Marinetti A, Quattrocchi CC, Greco MD, D'Onofrio M. Role of Cardiovascular Magnetic Resonance in the Assessment of Native Aortic Regurgitation With Insights on Mixed and Multiple Valvular Heart Disease: A Narrative Review. Echocardiography 2024; 41:e70045. [PMID: 39655361 DOI: 10.1111/echo.70045] [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: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiovascular magnetic resonance imaging (CMR) has received extensive validation for the assessment of valvular heart disease (VHD) and offers an accurate and direct method for the quantification of aortic regurgitation (AR). According to the current guidelines, CMR represents a useful second-line investigation in patients with poor acoustic windows or when echocardiography is inconclusive, for example, in cases of multiple or eccentric aortic jets. Without ionizing radiation exposure, CMR provides in-depth information not only on the severity degree of AR, providing a precise quantification of regurgitant volume and fraction, but also on cardiac structure and function, being recognized as the gold standard for the assessment of heart chamber size and systolic function. CMR allows a free choice of cardiac imaging planes and provides further information on the myocardium, thanks to the tissue characterization ability offered by several sequences, such as the late gadolinium enhancement technique. The possibilities offered by CMR become even more interesting in the context of mixed and multiple VHD, where the echocardiographic assessments often encounter difficulties in the quantification of each single valve lesion. The current scientific data support a greater expansion of CMR in this field, thanks to its additional advantages for the diagnosis, risk stratification, and to guide treatment. This review investigates the current CMR techniques and protocols in AR, with special insights into the evaluation of mixed aortic valve disease and multiple VHD including AR.
Collapse
Affiliation(s)
- Giulia Vinco
- Department of Radiology, G.B. Rossi University Hospital, University of Verona, Verona, Italy
| | | | - Cristina Demattè
- Department of Cardiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
| | - Cristiana Giovanelli
- Department of Cardiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
| | - Fabio Caruso
- Department of Radiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
| | - Alessandro Marinetti
- Department of Radiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
| | - Carlo Cosimo Quattrocchi
- Department of Radiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
- Centre for Medical Sciences - CISMed, University of Trento, Trento, Italy
| | - Maurizio Del Greco
- Department of Cardiology, Santa Maria del Carmine Hospital, APSS, Rovereto, Italy
| | - Mirko D'Onofrio
- Department of Radiology, G.B. Rossi University Hospital, University of Verona, Verona, Italy
| |
Collapse
|
23
|
Qin JJ, Gok M, Gholipour A, LoPilato J, Kirkby M, Poole C, Smith P, Grover R, Grieve SM. Four-Dimensional Flow MRI for Cardiovascular Evaluation (4DCarE): A Prospective Non-Inferiority Study of a Rapid Cardiac MRI Exam: Study Protocol and Pilot Analysis. Diagnostics (Basel) 2024; 14:2590. [PMID: 39594256 PMCID: PMC11593203 DOI: 10.3390/diagnostics14222590] [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: 10/10/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Accurate measurements of flow and ventricular volume and function are critical for clinical decision-making in cardiovascular medicine. Cardiac magnetic resonance (CMR) is the current gold standard for ventricular functional evaluation but is relatively expensive and time-consuming, thus limiting the scale of clinical applications. New volumetric acquisition techniques, such as four-dimensional flow (4D-flow) and three-dimensional volumetric cine (3D-cine) MRI, could potentially reduce acquisition time without loss in accuracy; however, this has not been formally tested on a large scale. METHODS 4DCarE (4D-flow MRI for cardiovascular evaluation) is a prospective, multi-centre study designed to test the non-inferiority of a compressed 20 min exam based on volumetric CMR compared with a conventional CMR exam (45-60 min). The compressed exam utilises 4D-flow together with a single breath-hold 3D-cine to provide a rapid, accurate quantitative assessment of the whole heart function. Outcome measures are (i) flow and chamber volume measurements and (ii) overall functional evaluation. Secondary analyses will explore clinical applications of 4D-flow-derived parameters, including wall shear stress, flow kinetic energy quantification, and vortex analysis in large-scale cohorts. A target of 1200 participants will enter the study across three sites. The analysis will be performed at a single core laboratory site. Pilot Results: We present a pilot analysis of 196 participants comparing flow measurements obtained by 4D-flow and conventional 2D phase contrast, which demonstrated moderate-good consistency in ascending aorta and main pulmonary artery flow measurements between the two techniques. Four-dimensional flow underestimated the flow compared with 2D-PC, by approximately 3 mL/beat in both vessels. CONCLUSIONS We present the study protocol of a prospective non-inferiority study of a rapid cardiac MRI exam compared with conventional CMR. The pilot analysis supports the continuation of the study. STUDY REGISTRATION This study is registered with the Australia and New Zealand Clinical Trials Registry (Registry number ACTRN12622000047796, Universal Trial Number: U1111-1270-6509, registered 17 January 2022-Retrospectively registered).
Collapse
Affiliation(s)
- Jiaxing Jason Qin
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Mustafa Gok
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Radiology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin 09010, Turkey
| | - Alireza Gholipour
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Jordan LoPilato
- ANU Medical School, Australian National University, Canberra, ACT 2601, Australia
| | - Max Kirkby
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Christopher Poole
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- iCoreLab, North Sydney, NSW 2060, Australia
| | - Paul Smith
- Epworth Radiology, Waurn Ponds, VIC 3216, Australia
| | - Rominder Grover
- Macquarie University Hospital, Macquarie Park, NSW 2109, Australia
| | - Stuart M. Grieve
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (J.J.Q.); (M.G.)
- School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Lumus Imaging, St George Private Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
24
|
Marwick TH, Dewar E, Nolan M, Shirazi M, Dias P, Wright L, Fitzgerald B, Kearney L, Srivastava P, Atherton J, Negishi K, Sverdlov AL, Wahi S, Otton J, Selvanayagam J, Thomas L, Thavendiranathan P. Strain surveillance during chemotherapy to improve cardiovascular outcomes: the SUCCOUR-MRI trial. Eur Heart J 2024; 45:4414-4424. [PMID: 39217601 PMCID: PMC11542702 DOI: 10.1093/eurheartj/ehae574] [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: 06/18/2024] [Revised: 07/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The detection of cancer therapy-related cardiac dysfunction (CTRCD) by reduction of left ventricular ejection fraction (LVEF) during chemotherapy usually triggers the initiation of cardioprotective therapy. This study addressed whether the same approach should be applied to patients with worsening of global longitudinal strain (GLS) without attaining thresholds of LVEF. METHODS Strain surveillance during chemotherapy for improving cardiovascular outcomes (SUCCOUR-MRI) was a prospective multicentre randomized controlled trial involving 14 sites. Of 355 patients receiving anthracyclines with normal baseline LVEF, 333 patients (age 59 ± 13 years, 79% women) with at least one other CTRCD risk factor, able to undergo magnetic resonance imaging (MRI), GLS, and three-dimensional echocardiography were tracked over 12 months. A total of 105 patients (age 59 ± 13 years, 75% women, 69% breast cancer) developing GLS-CTRCD (>12% relative reduction of GLS without a change in LVEF) were randomized to cardioprotection with neurohormonal antagonists vs. usual care. The primary endpoint was 12-month change in MRI-LVEF; the secondary endpoint was MRI-LVEF-defined CTRCD. RESULTS During follow-up, two patients died, and two developed heart failure. Most patients were randomized at 3 months (62%). Median doses of angiotensin inhibition/blockade and beta-blockade were 75% and 50% of respective targets; 21 (43%) had side-effects attributed to cardioprotection. Due to a smaller LVEF change from baseline with cardioprotection than usual care (-2.5 ± 5.4% vs. -5.6 ± 5.9%, P = .009), follow-up LVEF was higher after cardioprotection (59 ± 5% vs. 55 ± 6%, P < .0001). After adjustment for baseline LVEF, the mean (95% confidence interval) difference in the change in LVEF between the two groups was -3.6% (-1.8% to -5.5%, P < .001). After cardioprotection, 1/49 patients developed 12-month LVEF-CTRCD, compared to 6/56 in usual care (P = .075). Global longitudinal strain improved at 3 months post-randomization in the cardioprotection group, with little change with usual care. CONCLUSIONS In patients with isolated GLS reduction after anthracyclines, cardioprotection is associated with better preservation of 12-month MRI-LVEF compared with usual care.
Collapse
Affiliation(s)
- Thomas H Marwick
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardiovascular Imaging, Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Elizabeth Dewar
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Nolan
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardio-Oncology Section, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mitra Shirazi
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Peter Dias
- Advara Heart Care, Murdoch, WA, Australia
| | - Leah Wright
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | | | - John Atherton
- University of Queensland Faculty of Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
| | - Kazuaki Negishi
- Cardiology Department, Nepean Hospital, Kingswood, NSW, Australia
- Nepean Clinical School, University of Sydney, Kingswood, NSW, Australia
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, The University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
| | - Sudhir Wahi
- Princess Alexandra Hospital, Brisbane, Australia
| | - James Otton
- Liverpool Hospital, Liverpool, NSW, Australia
| | | | - Liza Thomas
- Westmead Clinical School, University of Sydney and University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
25
|
Weir-McCall JR, Fitton CA, Gandy SJ, Lambert M, Littleford R, Houston JG, Belch JJF. Sex-specific Associations between Left Ventricular Remodeling at MRI and Long-term Cardiovascular Risk. Radiology 2024; 313:e232997. [PMID: 39499172 DOI: 10.1148/radiol.232997] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Background Left ventricular mass (LVM) is an established marker of cardiovascular risk; however, long-term follow-up studies in individuals with low to intermediate risk are lacking. Purpose To assess the sex-specific association of LVM measured with cardiac MRI with cardiovascular outcomes in those with a less than 20% 10-year risk of cardiovascular disease (CVD). Materials and Methods A total of 1528 volunteers older than 40 years of age with no history of CVD, a 10-year risk of CVD of less than 20%, and a B-type natriuretic peptide level greater than their sex-specific median underwent cardiac MRI between June 2008 and February 2013 as part of the Tayside Screening for Cardiac Events, or TASCFORCE, prospective study. LVM was indexed to body surface area, and the LVM-to-volume ratio was calculated. Follow-up for cardiovascular events was performed using national electronic health records. Cox proportional hazard models and Kaplan-Meier curves were applied to assess the impact of LVM. Results A total of 1495 participants (mean age, 54.5 years ± 8.3 [SD]; 925 female, 570 male) completed cardiac MRI, with a median follow-up of 10 years (IQR, 3 years). In female participants, LVM was associated with age, blood pressure, smoking status, and cholesterol level, while in male participants, LVM was associated with age and blood pressure. In female participants, the LVM-to-volume ratio was associated with cardiovascular events (hazard ratio [HR], 2.3 [95% CI: 1.1, 4.9] for the highest quartile vs the lowest quartile), while the LVM was not. In male participants, the LVM was associated with cardiovascular events (HR, 3.2 [95% CI: 1.5,7.0] for the highest quartile vs the lowest quartile), while the LVM-to-volume ratio was not. Conclusion In those with low to intermediate risk without established CVD, different remodeling patterns predict cardiovascular events, with increased LVM predictive in male participants, while LVM-to-volume ratio is predictive in female participants. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Garot and Duhamel in this issue.
Collapse
Affiliation(s)
- Jonathan R Weir-McCall
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - Catherine A Fitton
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - Stephen J Gandy
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - Matthew Lambert
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - Roberta Littleford
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - J Graeme Houston
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| | - Jill J F Belch
- From the School of Clinical Medicine, University of Cambridge, Cambridge, UK (J.R.W.M.); Department of Radiology, Royal Papworth Hospital, Cambridge, UK (J.R.W.M.); Division of Cardiovascular Research, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK (C.A.F., J.G.H., J.J.F.B.); NHS Tayside Medical Physics, Ninewells Hospital, Dundee, UK (S.J.G.); Stroke Department, NHS Tayside, Dundee, UK (M.L.); and The University of Queensland, Brisbane, Australia (R.L.)
| |
Collapse
|
26
|
El Mathari S, Hopman LHGA, Bouchnaf C, Heidendael JF, Nederveen AJ, van Ooij P, Selder JL, van Loon RB, Götte MJW, Kluin J. Clinical implications of different methods to assess left atrial remodeling: A comparative study between echocardiography and cardiac magnetic resonance imaging for left atrial volume index quantification. Int J Cardiol 2024; 414:132443. [PMID: 39128567 DOI: 10.1016/j.ijcard.2024.132443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/14/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Left atrial volume index (LAVI) serves as a crucial marker for assessing left atrial (LA) remodeling, particularly in patients with mitral valve regurgitation (MR). Recent guidelines recommend a LAVI exceeding 60 mL/m2 as Class IIa recommendation for mitral valve repair surgery in asymptomatic MR patients with preserved left ventricular function. Traditionally, echocardiography is the standard for assessing LAVI in MR patients. However, cardiac magnetic resonance imaging (CMR) is increasingly recognized for its more precise measurements of cardiac dimensions and volumes. But still, literature remains scarce on comparing the efficacy of both modalities in assessing LAVI measurements. METHODS This retrospective study included 168 MR patients undergoing both echocardiography and CMR assessments within a six-month period. LAVI measurements were compared using Pearson correlation and Bland-Altman plots. Patients were stratified based on MR grades, and clinical implications were assessed. RESULTS Mean LAVI differed significantly between echocardiography and CMR (47.1 ± 20.8 mL/m2 versus 70 ± 20.3 mL/m2, p < 0.001, respectively). CMR consistently yielded higher LAVI measurements compared to echocardiography, with a mean difference of approximately 20 mL/m2. CMR measurements resulted in an increased incidence of patients meeting the class IIa LAVI criterion (LAVI >60 mL/m2) by 37%. Variations in LAVI did not differ across MR grades. CONCLUSION Echocardiography systematically underestimates LAVI compared to CMR in MR patients. While current guidelines rely on echocardiography, CMR's precision suggests the need for CMR-specific LAVI cutoff values to guide clinical management effectively. Establishing such values could refine patient stratification and timing of surgery, potentially improving clinical outcomes for MR patients.
Collapse
Affiliation(s)
- Sulayman El Mathari
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| | - Luuk H G A Hopman
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Chaimae Bouchnaf
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Josephine F Heidendael
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, the Netherlands
| | - Pim van Ooij
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, the Netherlands
| | - Jasper L Selder
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ramon B van Loon
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Marco J W Götte
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jolanda Kluin
- Department of Cardiothoracic Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
27
|
Gissler MC, Antiochos P, Ge Y, Heydari B, Gräni C, Kwong RY. Cardiac Magnetic Resonance Evaluation of LV Remodeling Post-Myocardial Infarction: Prognosis, Monitoring and Trial Endpoints. JACC Cardiovasc Imaging 2024; 17:1366-1380. [PMID: 38819335 DOI: 10.1016/j.jcmg.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 06/01/2024]
Abstract
Adverse left ventricular remodeling (ALVR) and subsequent heart failure after myocardial infarction (MI) remain a major cause of patient morbidity and mortality worldwide. Overt inflammation has been identified as the common pathway underlying myocardial fibrosis and development of ALVR post-MI. With its ability to simultaneously provide information about cardiac structure, function, perfusion, and tissue characteristics, cardiac magnetic resonance (CMR) is well poised to inform prognosis and guide early surveillance and therapeutics in high-risk cohorts. Further, established and evolving CMR-derived biomarkers may serve as clinical endpoints in prospective trials evaluating the efficacy of novel anti-inflammatory and antifibrotic therapies. This review provides an overview of post-MI ALVR and illustrates how CMR may help clinical adoption of novel therapies via mechanistic or prognostic imaging markers.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Panagiotis Antiochos
- Cardiology and Cardiac MR Centre, University Hospital Lausanne, Lausanne, Switzerland
| | - Yin Ge
- Division of Cardiology, St Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Bobak Heydari
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christoph Gräni
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raymond Y Kwong
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
28
|
Edin C, Ekstedt M, Karlsson M, Wegmann B, Warntjes M, Swahn E, Östgren CJ, Ebbers T, Lundberg P, Carlhäll CJ. Liver fibrosis is associated with left ventricular remodeling: insight into the liver-heart axis. Eur Radiol 2024; 34:7492-7502. [PMID: 38795131 PMCID: PMC11519090 DOI: 10.1007/s00330-024-10798-1] [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: 11/01/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/27/2024]
Abstract
OBJECTIVE In nonalcoholic fatty liver disease (NAFLD), liver fibrosis is the strongest predictor of adverse outcomes. We sought to investigate the relationship between liver fibrosis and cardiac remodeling in participants from the general population using magnetic resonance imaging (MRI), as well as explore potential mechanistic pathways by analyzing circulating cardiovascular biomarkers. METHODS In this cross-sectional study, we prospectively included participants with type 2 diabetes and individually matched controls from the SCAPIS (Swedish CArdioPulmonary bioImage Study) cohort in Linköping, Sweden. Between November 2017 and July 2018, participants underwent MRI at 1.5 Tesla for quantification of liver proton density fat fraction (spectroscopy), liver fibrosis (stiffness from elastography), left ventricular (LV) structure and function, as well as myocardial native T1 mapping. We analyzed 278 circulating cardiovascular biomarkers using a Bayesian statistical approach. RESULTS In total, 92 participants were enrolled (mean age 59.5 ± 4.6 years, 32 women). The mean liver stiffness was 2.1 ± 0.4 kPa. 53 participants displayed hepatic steatosis. LV concentricity increased across quartiles of liver stiffness. Neither liver fat nor liver stiffness displayed any relationships to myocardial tissue characteristics (native T1). In a regression analysis, liver stiffness was related to increased LV concentricity. This association was independent of diabetes and liver fat (Beta = 0.26, p = 0.0053), but was attenuated (Beta = 0.17, p = 0.077) when also adjusting for circulating levels of interleukin-1 receptor type 2. CONCLUSION MRI reveals that liver fibrosis is associated to structural LV remodeling, in terms of increased concentricity, in participants from the general population. This relationship could involve the interleukin-1 signaling. CLINICAL RELEVANCE STATEMENT Liver fibrosis may be considered a cardiovascular risk factor in patients without cirrhosis. Further research on the mechanisms that link liver fibrosis to left ventricular concentricity may reveal potential therapeutic targets in patients with non-alcoholic fatty liver disease (NAFLD). KEY POINTS Previously, studies on liver fibrosis and cardiac remodeling have focused on advanced stages of liver fibrosis. Liver fibrosis is associated with left ventricular (LV) concentricity and may relate to interleukin-1 receptor type 2. Interleukin-1 signaling is a potential mechanistic interlink between early liver fibrosis and LV remodeling.
Collapse
Affiliation(s)
- Carl Edin
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Clinical Physiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Markus Karlsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Bertil Wegmann
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Marcel Warntjes
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Eva Swahn
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Cardiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Carl Johan Östgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Division of Prevention, Rehabilitation and Community Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Tino Ebbers
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Carl-Johan Carlhäll
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.
- Department of Clinical Physiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
29
|
Ipek R, Holland J, Cramer M, Rider O. CMR to characterize myocardial structure and function in heart failure with preserved left ventricular ejection fraction. Eur Heart J Cardiovasc Imaging 2024; 25:1491-1504. [PMID: 39205602 PMCID: PMC11522877 DOI: 10.1093/ehjci/jeae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Despite remarkable progress in therapeutic drugs, morbidity, and mortality for heart failure (HF) remains high in developed countries. HF with preserved ejection fraction (HFpEF) now accounts for around half of all HF cases. It is a heterogeneous disease, with multiple aetiologies, and as such poses a significant diagnostic challenge. Cardiac magnetic resonance (CMR) has become a valuable non-invasive modality to assess cardiac morphology and function, but beyond that, the multi-parametric nature of CMR allows novel approaches to characterize haemodynamics and with magnetic resonance spectroscopy (MRS), the study of metabolism. Furthermore, exercise CMR, when combined with lung water imaging provides an in-depth understanding of the underlying pathophysiological and mechanistic processes in HFpEF. Thus, CMR provides a comprehensive phenotyping tool for HFpEF, which points towards a targeted and personalized therapy with improved diagnostics and prevention.
Collapse
Affiliation(s)
- Rojda Ipek
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
- Divison of Cardiology, Pulmonary Disease and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jennifer Holland
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
| | - Mareike Cramer
- Divison of Cardiology, Pulmonary Disease and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Oliver Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research (OCMR), John Radcliffe Hospital, Level 0, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
30
|
Puntmann VO, Beitzke D, Kammerlander A, Voges I, Gabbert DD, Doerr M, Chamling B, Bozkurt B, Kaski JC, Spatz E, Herrmann E, Rohde G, DeLeuw P, Taylor L, Windemuth-Kieselbach C, Harz C, Santiuste M, Schoeckel L, Hirayama J, Taylor PC, Berry C, Nagel E. Design and rationale of MYOFLAME-19 randomised controlled trial: MYOcardial protection to reduce post-COVID inFLAMmatory heart disease using cardiovascular magnetic resonance Endpoints. J Cardiovasc Magn Reson 2024; 27:101121. [PMID: 39481808 PMCID: PMC11697771 DOI: 10.1016/j.jocmr.2024.101121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Cardiac symptoms due to postacute inflammatory cardiac involvement affect a broad segment of previously well people with only mild acute coronavirus disease 2019 (COVID-19) illness and without overt structural heart disease. Cardiovascular magnetic resonance (CMR) imaging can identify the underlying subclinical disease process, which is associated with chronic cardiac symptoms. Specific therapy directed at reducing postacute cardiac inflammatory involvement before development of myocardial injury and impairment is missing. METHODS Prospective multicenter randomized placebo-controlled study of myocardial protection therapy (combined immunosuppressive/antiremodeling) of low-dose prednisolone and losartan. Consecutive symptomatic individuals with a prior COVID-19 infection, no pre-existing significant comorbidities or structural heart disease, undergo standardized assessments with questionnaires, CMR imaging, and cardiopulmonary exercise testing (CPET). Eligible participants fulfilling the criteria of subclinical post-COVID inflammatory heart involvement on baseline CMR examination are randomized to treatment with either verum or placebo for a total of 16 weeks (W16). Participants and investigators remain blinded to the group allocation throughout the study duration. The primary efficacy endpoint is the absolute change of left ventricular ejection fraction to baseline at W16, measured by CMR, between the verum treatment and placebo group by absolute difference, using unpaired t-test confirmatively at a significance level of 0.05 significance level. Secondary endpoints include assessment of changes of symptoms, CMR parameters, and CPET after W16, and frequency of major adverse cardiac events after 1 year. Safety data will be analyzed for frequency, severity, and types of adverse events (AEs) for all treatment groups. The proportion of AEs related to the contrast agent gadobutrol will also be analyzed. A calculated sample size is a total of 280 participants (accounting for 22 subjects (8%) drop out), randomized in 1:1 fashion to 140 in the verum and 140 placebo groups. CONCLUSION Myoflame-19 study will examine the efficacy of a myocardial protection therapy in symptomatic participants with post-COVID inflammatory cardiac involvement determined by CMR. The aim of the intervention is to reduce the symptoms and inflammatory myocardial injury, improve exercise tolerance, and preclude the development of cardiac impairment.
Collapse
Affiliation(s)
- Valentina O Puntmann
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, Frankfurt am Main, Germany; German Centre for Cardiovascular Research - Partner Site Rhein-Main, Rhein-Main, Germany.
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Radiology and Nuclear Medicine, University Hospital Vienna, Vienna, Austria
| | | | - Inga Voges
- Department of Cardiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; German Centre for Cardiovascular Research - Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Dominik D Gabbert
- Department of Cardiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Marcus Doerr
- Department of Cardiology, Angiology, and Pulmonology, Internal Intensive Care Unit, University Hospital Greifswald, Greifswald, Germany; German Centre for Cardiovascular Research - Partner Site Greifswald, Greifswald, Germany
| | - Bishwas Chamling
- Department of Cardiology, Angiology, and Pulmonology, Internal Intensive Care Unit, University Hospital Greifswald, Greifswald, Germany; German Centre for Cardiovascular Research - Partner Site Greifswald, Greifswald, Germany
| | - Biykem Bozkurt
- Winters Center for Heart Failure Research, Houston, Texas, USA; Cardiovascular Research Institute, Baylor College of Medicine, DeBakey VA Medical Center, Houston, Texas, USA
| | - Juan Carlos Kaski
- Cardiovascular Sciences, Molecular and Clinical Sciences, St George's, University of London, London, UK
| | - Erica Spatz
- Cardiovascular Medicine, Yale Center for Outcomes Research and Evaluation, Yale School of Medicine, 800 Howard Avenue, New Haven, Connecticut 06519, USA
| | - Eva Herrmann
- Institute of Biostatistics and Mathematical Modeling, Center for Health Sciences, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Gernot Rohde
- Department of Respiratory Medicine, Goethe University Frankfurt, Medical Clinic I, University Hospital, Frankfurt am Main, Germany
| | | | - Lenka Taylor
- Pharmacy of the Clinical Trial Unit, Medical School, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Headington, Oxford OX3 7LD, UK
| | - Colin Berry
- University of Glasgow, School of Cardiovascular & Metabolic Health, BHF Glasgow Cardiovascular Research Centre (GCRC), Glasgow, UK
| | - Eike Nagel
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, Frankfurt am Main, Germany; German Centre for Cardiovascular Research - Partner Site Rhein-Main, Rhein-Main, Germany
| |
Collapse
|
31
|
Lim S, Bae JH, Oh H, Hwang IC, Yoon YE, Cho GY. Effect of ertugliflozin on left ventricular function in type 2 diabetes and pre-heart failure: the Ertu-GLS randomized clinical trial. Cardiovasc Diabetol 2024; 23:373. [PMID: 39438942 PMCID: PMC11515769 DOI: 10.1186/s12933-024-02463-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The therapeutic effects of ertugliflozin, a sodium-glucose cotransporter 2 inhibitor, on cardiovascular outcome are not fully understood. This study aimed to evaluate the efficacy and safety of ertugliflozin on cardiac function in people with type 2 diabetes and pre-heart failure. METHODS We conducted a 24-week randomized, double-blind, placebo-controlled trial involving individuals with type 2 diabetes inadequately controlled with antidiabetic medications. Participants with left ventricular hypertrophy, E/e' >15, or impaired left ventricular global longitudinal strain (LVGLS) were randomized 1:1 to receive either ertugliflozin (5 mg once daily) or a placebo. The primary outcome was the change in LVGLS. Secondary outcomes included changes in left ventricular mass index (LVMI) and left ventricular ejection fraction (LVEF). Prespecified exploratory outcomes, including angiotensin-converting enzyme 2 (ACE2) and angiotensin (1-7) levels, were also assessed. RESULTS A total of 102 individuals (mean age, 63.9 ± 9.2 years; 38% women) were included. The ertugliflozin group showed a significant improvement in LVGLS (- 15.5 ± 3.1% to - 16.6 ± 2.8%, P = 0.004) compared to the placebo group (- 16.7 ± 2.7% to - 16.4 ± 2.6%, P = 0.509), with a significant between-group difference (P = 0.013). Improvements in LVMI and LVEF were also observed. Additionally, significant reductions in HbA1c, systolic blood pressure, whole-body and visceral fat, uric acid, proteinuria, N-terminal pro-B-type natriuretic peptide, and lipoprotein(a) were noted. ACE2 and angiotensin (1-7) levels significantly increased in the ertugliflozin group compared to the placebo group and correlated with changes in LVGLS [r = 0.456, P < 0.001 for ACE2; r = 0.541, P < 0.001 for angiotensin (1-7)]. Adverse events were similar between the two groups. CONCLUSIONS This study demonstrated that ertugliflozin has beneficial effects on left ventricular function in individuals with type 2 diabetes and pre-heart failure, and it provided insights into potential underlying mechanisms. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03717194.
Collapse
MESH Headings
- Humans
- Male
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/complications
- Female
- Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
- Sodium-Glucose Transporter 2 Inhibitors/adverse effects
- Middle Aged
- Aged
- Double-Blind Method
- Ventricular Function, Left/drug effects
- Treatment Outcome
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/adverse effects
- Stroke Volume/drug effects
- Time Factors
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/diagnosis
- Biomarkers/blood
- Recovery of Function
- Angiotensin-Converting Enzyme 2/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/drug therapy
- Hypertrophy, Left Ventricular/diagnostic imaging
- Heart Failure/drug therapy
- Heart Failure/physiopathology
- Heart Failure/diagnosis
- Blood Glucose/drug effects
- Blood Glucose/metabolism
Collapse
Affiliation(s)
- Soo Lim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| | - Jae Hyun Bae
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Heran Oh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - In-Chang Hwang
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yeonyee E Yoon
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Goo-Yeong Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
32
|
Arzt M, Fox H, Stadler S, Hetzenecker A, Oldenburg O, Hamer OW, Poschenrieder F, Wiest C, Tanacli R, Kelle S, Bruch L, Seidel M, Koller M, Zeman F, Buchner S. Treatment of sleep apnoea early after myocardial infarction with adaptive servo-ventilation: a proof-of-concept randomised controlled trial. Eur Respir J 2024; 64:2302338. [PMID: 38991707 DOI: 10.1183/13993003.02338-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 06/14/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Sleep disordered breathing (SDB) has been associated with less myocardial salvage and smaller infarct size reduction after acute myocardial infarction (AMI). The Treatment of sleep apnoea Early After Myocardial infarction with Adaptive Servo-Ventilation (TEAM-ASV I) trial investigated the effects of adding adaptive servo-ventilation (ASV) for SDB to standard therapy on the myocardial salvage index (MSI) and change in infarct size within 12 weeks after AMI. METHODS In this multicentre, randomised, open-label trial, patients with AMI and successful percutaneous coronary intervention within 24 h after symptom onset plus SDB (apnoea-hypopnoea index ≥15 events·h-1) were randomised to standard medical therapy alone (control) or plus ASV (starting 3.6±1.4 days post-AMI). The primary outcome was the MSI at 12 weeks post-AMI. Cardiac magnetic resonance (CMR) imaging was performed at ≤5 days and 12 weeks after AMI. RESULTS 76 individuals were enrolled from February 2014 to August 2020; 39 had complete CMR data for analysis of the primary end-point. The MSI was significantly higher in the ASV versus control group (difference 14.6% (95% CI 0.14-29.1%); p=0.048). At 12 weeks, absolute (6.6 (95% CI 4.8-8.5) versus 2.8 (95% CI 0.9-4.8) % of left ventricular mass; p=0.003) and relative (44 (95% CI 30-57) versus 21 (95% CI 6-35) % of baseline; p=0.013) reductions in infarct size were greater in the ASV versus control group. No serious treatment-related adverse events occurred. CONCLUSIONS Early treatment of SDB with ASV improved the MSI and decreased infarct size at 12 weeks after AMI. Larger randomised trials are required to confirm these findings.
Collapse
Affiliation(s)
- Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Henrik Fox
- Clinic for General and Interventional Cardiology/Angiology Heart and Diabetes Center, NRW Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Stefan Stadler
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Andrea Hetzenecker
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Center for Pneumology, Donaustauf Hospital, Donaustauf, Germany
| | - Olaf Oldenburg
- Center for Cardiology, Ludgerus-Kliniken, Münster, Germany
| | - Okka W Hamer
- Center for Pneumology, Donaustauf Hospital, Donaustauf, Germany
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Poschenrieder
- Center for Pneumology, Donaustauf Hospital, Donaustauf, Germany
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | - Clemens Wiest
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Radu Tanacli
- Department of Internal Medicine/Cardiology, German Heart Center Berlin, Berlin, Germany
- Department of Internal Medicine/Cardiology, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Sebastian Kelle
- Department of Internal Medicine/Cardiology, German Heart Center Berlin, Berlin, Germany
- Department of Internal Medicine/Cardiology, Charité Campus Virchow Klinikum, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Leonhard Bruch
- Department of Internal Medicine, Unfallkrankenhaus Berlin, Berlin, Germany
| | - Mirko Seidel
- Department of Internal Medicine, Unfallkrankenhaus Berlin, Berlin, Germany
| | - Michael Koller
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Buchner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
- Internal Medicine II - Cardiology, Sana Clinics of the District of Cham, Cham, Germany
| |
Collapse
|
33
|
Chadalavada S, Fung K, Rauseo E, Lee AM, Khanji MY, Amir-Khalili A, Paiva J, Naderi H, Banik S, Chirvasa M, Jensen MT, Aung N, Petersen SE. Myocardial Strain Measured by Cardiac Magnetic Resonance Predicts Cardiovascular Morbidity and Death. J Am Coll Cardiol 2024; 84:648-659. [PMID: 39111972 PMCID: PMC11320766 DOI: 10.1016/j.jacc.2024.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Myocardial strain using cardiac magnetic resonance (CMR) is a sensitive marker for predicting adverse outcomes in many cardiac disease states, but the prognostic value in the general population has not been studied conclusively. OBJECTIVES The goal of this study was to assess the independent prognostic value of CMR feature tracking (FT)-derived LV global longitudinal (GLS), circumferential (GCS), and radial strain (GRS) metrics in predicting adverse outcomes (heart failure, myocardial infarction, stroke, and death). METHODS Participants from the UK Biobank population imaging study were included. Univariable and multivariable Cox models were used for each outcome and each strain marker (GLS, GCS, GRS) separately. The multivariable models were tested with adjustment for prognostically important clinical features and conventional global LV imaging markers relevant for each outcome. RESULTS Overall, 45,700 participants were included in the study (average age 65 ± 8 years), with a median follow-up period of 3 years. All univariable and multivariable models demonstrated that lower absolute GLS, GCS, and GRS were associated with increased incidence of heart failure, myocardial infarction, stroke, and death. All strain markers were independent predictors (incrementally above some respective conventional LV imaging markers) for the morbidity outcomes, but only GLS predicted death independently: (HR: 1.18; 95% CI: 1.07-1.30). CONCLUSIONS In the general population, LV strain metrics derived using CMR-FT in radial, circumferential, and longitudinal directions are strongly and independently predictive of heart failure, myocardial infarction, and stroke, but only GLS is independently predictive of death in an adult population cohort.
Collapse
Affiliation(s)
- Sucharitha Chadalavada
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Kenneth Fung
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Elisa Rauseo
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
| | - Aaron M Lee
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
| | - Mohammed Y Khanji
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | | | - Jose Paiva
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
| | - Hafiz Naderi
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Shantanu Banik
- Circle Cardiovascular Imaging Inc, Calgary, Alberta, Canada
| | | | | | - Nay Aung
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom; Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom; Health Data Research UK, London, United Kingdom; Alan Turing Institute, The British Library, John Dodson House, London, United Kingdom.
| |
Collapse
|
34
|
De Alcubierre D, Feola T, Cozzolino A, Pofi R, Galea N, Catalano C, Auriemma RS, Pirchio R, Pivonello R, Isidori AM, Giannetta E. The spectrum of cardiac abnormalities in patients with acromegaly: results from a case-control cardiac magnetic resonance study. Pituitary 2024; 27:416-427. [PMID: 38847918 PMCID: PMC11289141 DOI: 10.1007/s11102-024-01403-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE Cardiac abnormalities are common in patients with acromegaly, contributing to the increased morbidity and mortality. Cardiac magnetic resonance (CMR) is the gold standard for measuring cardiac morpho-functional changes. This study aims to detect cardiac alterations in acromegaly through CMR, even when the disease is adequately controlled. METHODS In this, multicentre, case-control study, we compared consecutive patients with acromegaly, cured after surgery or requiring medical treatment, with matched controls recruited among patients harbouring non-functioning adrenal incidentalomas. RESULTS We included 20 patients with acromegaly (7 females, mean age 50 years) and 17 controls. Indexed left ventricular-end-diastolic volume (LV-EDVi) and LV-end-systolic volume (LV-ESVi) were higher in patients than in controls (p < 0.001), as were left ventricular mass (LVMi) (p = 0.001) and LV-stroke volume (LV-SVi) (p = 0.028). Right ventricle (RV) EDVi and ESVi were higher, whereas RV-ejection fraction (RV-EF) was lower (p = 0.002) in patients than in controls (p < 0.001). No significant differences were observed in the prevalence of cardiometabolic comorbidities, including hypertension, glucose and lipid metabolism impairment, obstructive sleep apnoea syndrome, and obesity. IGF1 x upper limit of normal significantly predicted LVMi (b = 0.575; p = 0.008). Subgroup analysis showed higher LVMi (p = 0.025) and interventricular septum thickness (p = 0.003) in male than female patients, even after adjusting cardiac parameters for confounding factors. CONCLUSIONS The CMR analysis reveals a cluster of biventricular structural and functional impairment in acromegaly, even when the biochemical control if achieved. These findings appear specifically triggered by the exposure to GH-IGF1 excess and show sex-related differences advocating a possible interaction with sex hormones in cardiac disease progression.
Collapse
Affiliation(s)
- Dario De Alcubierre
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - Alessia Cozzolino
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Riccardo Pofi
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill hospital, Oxford University Hospitals, NHS Trust, Oxford, UK
| | - Nicola Galea
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Carlo Catalano
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Renata Simona Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Naples, Italy
| | - Rosa Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università Federico II di Napoli, Naples, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome, 00161, Italy.
- Centre for Rare Diseases (ENDO-ERN accredited), Policlinico Umberto I, Rome, Italy.
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome, 00161, Italy.
| |
Collapse
|
35
|
Eyre K, Rafiee MJ, Leo M, Ma J, Hillier E, Amini N, Pressacco J, Janich MA, Zhu X, Friedrich MG, Chetrit M. Clinical utility of a rapid two-dimensional balanced steady-state free precession sequence with deep learning reconstruction. J Cardiovasc Magn Reson 2024; 26:101069. [PMID: 39079600 PMCID: PMC11367510 DOI: 10.1016/j.jocmr.2024.101069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/14/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Cardiovascular magnetic resonance (CMR) cine imaging is still limited by long acquisition times. This study evaluated the clinical utility of an accelerated two-dimensional (2D) cine sequence with deep learning reconstruction (Sonic DL) to decrease acquisition time without compromising quantitative volumetry or image quality. METHODS A sub-study using 16 participants was performed using Sonic DL at two different acceleration factors (8× and 12×). Quantitative left-ventricular volumetry, function, and mass measurements were compared between the two acceleration factors against a standard cine method. Following this sub-study, 108 participants were prospectively recruited and imaged using a standard cine method and the Sonic DL method with the acceleration factor that more closely matched the reference method. Two experienced clinical readers rated images based on their diagnostic utility and performed all image contouring. Quantitative contrast difference and endocardial border sharpness were also assessed. Left- and right-ventricular volumetry, left-ventricular mass, and myocardial strain measurements were compared between cine methods using Bland-Altman plots, Pearson's correlation, and paired t-tests. Comparative analysis of image quality was measured using Wilcoxon-signed-rank tests and visualized using bar graphs. RESULTS Sonic DL at an acceleration factor of 8 more closely matched the reference cine method. There were no significant differences found across left ventricular volumetry, function, or mass measurements. In contrast, an acceleration factor of 12 resulted in a 6% (5.51/90.16) reduction of measured ejection fraction when compared to the standard cine method and a 4% (4.32/88.98) reduction of measured ejection fraction when compared to Sonic DL at an acceleration factor of 8. Thus, Sonic DL at an acceleration factor of 8 was chosen for downstream analysis. In the larger cohort, this accelerated cine sequence was successfully performed in all participants and significantly reduced the acquisition time of cine images compared to the standard 2D method (reduction of 37% (5.98/16) p < 0.0001). Diagnostic image quality ratings and quantitative image quality evaluations were statistically not different between the two methods (p > 0.05). Left- and right-ventricular volumetry and circumferential and radial strain were also similar between methods (p > 0.05) but left-ventricular mass and longitudinal strain were over-estimated using the proposed accelerated cine method (mass over-estimated by 3.36 g/m2, p < 0.0001; longitudinal strain over-estimated by 1.97%, p = 0.001). CONCLUSION This study found that an accelerated 2D cine method with DL reconstruction at an acceleration factor of 8 can reduce CMR cine acquisition time by 37% (5.98/16) without significantly affecting volumetry or image quality. Given the increase of scan time efficiency, this undersampled acquisition method using deep learning reconstruction should be considered for routine clinical CMR.
Collapse
Affiliation(s)
- Katerina Eyre
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada.
| | | | - Margherita Leo
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Junjie Ma
- GE HealthCare, Milwaukee, Wisconsin, USA
| | - Elizabeth Hillier
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Negin Amini
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Josephine Pressacco
- Department of Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | | | | | - Matthias G Friedrich
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada; Area19 Medical Inc., Montreal, Canada; Division of Cardiology, McGill University, Montreal, Quebec, Canada
| | - Michael Chetrit
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada; Division of Cardiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
36
|
Cheung HC, Vimalesvaran K, Zaman S, Michaelides M, Shun-Shin MJ, Francis DP, Cole GD, Howard JP. Automating quality control in cardiac magnetic resonance: Artificial intelligence for discriminative assessment of planning and motion artifacts and real-time reacquisition guidance. J Cardiovasc Magn Reson 2024; 26:101067. [PMID: 39079601 PMCID: PMC11416635 DOI: 10.1016/j.jocmr.2024.101067] [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: 05/12/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Accurate measurements from cardiovascular magnetic resonance (CMR) images require precise positioning of scan planes and elimination of motion artifacts from arrhythmia or breathing. Unidentified or incorrectly managed artifacts degrade image quality, invalidate clinical measurements, and decrease diagnostic confidence. Currently, radiographers must manually inspect each acquired image to confirm diagnostic quality and decide whether reacquisition or a change in sequences is warranted. We aimed to develop artificial intelligence (AI) to provide continuous quality scores across different quality domains, and from these, determine whether cines are clinically adequate, require replanning, or warrant a change in protocol. METHODS A three-dimensional convolutional neural network was trained to predict cine quality graded on a continuous scale by a level 3 CMR expert, focusing separately on planning and motion artifacts. It incorporated four distinct output heads for the assessment of image quality in terms of (a, b, c) 2-, 3- and 4-chamber misplanning, and (d) long- and short-axis arrhythmia/breathing artifact. Backpropagation was selectively performed across these heads based on the labels present for each cine. Each image in the testing set was reported by four level 3 CMR experts, providing a consensus on clinical adequacy. The AI's assessment of image quality and ability to identify images requiring replanning or sequence changes were evaluated with Spearman's rho and the area under receiver operating characteristic curve (AUROC), respectively. RESULTS A total of 1940 cines across 1387 studies were included. On the test set of 383 cines, AI-judged image quality correlated strongly with expert judgment, with Spearman's rho of 0.84, 0.84, 0.81, and 0.81 for 2-, 3- and 4-chamber planning quality and the extent of arrhythmia or breathing artifacts, respectively. The AI also showed high efficacy in flagging clinically inadequate cines (AUROC 0.88, 0.93, and 0.93 for identifying misplanning of 2-, 3- and 4-chamber cines, and 0.90 for identifying movement artifacts). CONCLUSION AI can assess distinct domains of CMR cine quality and provide continuous quality scores that correlate closely with a consensus of experts. These ratings could be used to identify cases where reacquisition is warranted and guide corrective actions to optimize image quality, including replanning, prospective gating, or real-time imaging.
Collapse
Affiliation(s)
- Hoi C Cheung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kavitha Vimalesvaran
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sameer Zaman
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michalis Michaelides
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthew J Shun-Shin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Darrel P Francis
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Graham D Cole
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - James P Howard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
37
|
Maurizi N, Nowak A, Gruner C, Namdar M, Schmied C, Porretta AP, Barbey G, Monzambani V, Monney P, Barbey F. Fabry disease: development and progression of left ventricular hypertrophy despite long-term enzyme replacement therapy. Heart 2024; 110:997-1004. [PMID: 38749654 DOI: 10.1136/heartjnl-2024-323975] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Enzyme replacement therapy (ERT) may halt or attenuate disease progression in patients with Anderson-Fabry disease (AFD). However, whether left ventricular hypertrophy (LVH) can be prevented by early therapy or may still progress despite ERT over a long-term follow-up is still unclear. METHODS Consecutive patients with AFD from the Independent Swiss-Fabry Cohort receiving ERT who were at least followed up for 5 years were included. Cardiac progression was defined as an increase of >10 g/m2 in left ventricular mass index (LVMI) between the first and the last available follow-up transthoracic echocardiography. RESULTS 60 patients (35 (23-48) years, 39 (65%) men) were followed up for 10.5 (7.2-12.2) years. 22 had LVH at ERT start (LVMI of 150±38 g/m2). During follow-up, 22 (36%, 34±15 years) had LVMI progression of 12.1 (7-17.6) g/m2 per 100 patient-years, of these 7 (11%, 29±13 years) with no LVH at baseline. Three of them progressed to LVH. LVMI progression occurred mostly in men (17 of 39 (43%) vs 5 of 21 (24%), p<0.01) and after the age of 30 years (17 of 22 (77%)). LVH at ERT start was associated with LVMI progression (OR 1.3, 95% CI 1.1 to 2.6; p=0.02). A total of 19 (31%) patients experienced a major AFD-related event. They were predominantly men (17 of 19, 89%), older (45±11 vs 32±9 years) with baseline LVH (12 of 19, 63%), and 10 of 19 (52%) presented with LVMI progression. CONCLUSIONS Over a median follow-up of >10 years under ERT, 36% of the patients still had LVMI cardiac progression, and 32%, predominantly older men, experienced major AFD-related events. LVH at treatment initiation was a strong predictor of LVMI progression and adverse events on ERT.
Collapse
Affiliation(s)
- Niccolo Maurizi
- Department of Cardiology, University Hospital of Lausannne, Lausanne, Switzerland
| | - Albina Nowak
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | - Christiane Gruner
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Mehdi Namdar
- Department of Cardiology, Hopitaux Universitaires de Genève, Geneva, Switzerland
| | - Christian Schmied
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Guillaume Barbey
- Department of Cardiology, University Hospital of Lausannne, Lausanne, Switzerland
| | - Veronique Monzambani
- Department of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - Pierre Monney
- Department of Cardiology, University Hospital of Lausannne, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Frédéric Barbey
- Department of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
38
|
Larsen BS, Biering-Sørensen T, Olsen FJ. Ischemic stroke and the emerging role of left atrial function. Expert Rev Cardiovasc Ther 2024; 22:289-300. [PMID: 38943632 DOI: 10.1080/14779072.2024.2370814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/18/2024] [Indexed: 07/01/2024]
Abstract
INTRODUCTION Ischemic stroke is a leading cause of morbidity and mortality worldwide. Emerging evidence suggests that left atrial (LA) dysfunction could play a role in the pathophysiology of ischemic stroke, as a possible contributor and as a predictive biomarker. AREAS COVERED This narrative review details the intricate relationship between LA function, atrial fibrillation (AF), and ischemic stroke. We discuss imaging techniques used to assess LA function, the mechanisms by which impaired LA function may contribute to stroke, and its potential as a prognostic marker of stroke. EXPERT OPINION There is a lack of evidence-based treatments of LA dysfunction in both primary and secondary stroke prevention. This is partly due to the lack of a practical clinical definition and unanswered questions concerning the clinical implications of LA dysfunction in patients without AF. Until such questions are resolved, addressing well-known cardiovascular risk factors, like hypertension and obesity, should be prioritized for preventing AF and ischemic stroke. These risk factors are closely tied to atrial remodeling, emphasizing the importance of targeting primary modifiable factors for preventing future morbidity and mortality.
Collapse
Affiliation(s)
- Bjørn Strøier Larsen
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tor Biering-Sørensen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Flemming Javier Olsen
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| |
Collapse
|
39
|
Miller RJH, Shanbhag A, Killekar A, Lemley M, Bednarski B, Kavanagh PB, Feher A, Miller EJ, Bateman T, Builoff V, Liang JX, Newby DE, Dey D, Berman DS, Slomka PJ. AI-Defined Cardiac Anatomy Improves Risk Stratification of Hybrid Perfusion Imaging. JACC Cardiovasc Imaging 2024; 17:780-791. [PMID: 38456877 PMCID: PMC11222053 DOI: 10.1016/j.jcmg.2024.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Computed tomography attenuation correction (CTAC) improves perfusion quantification of hybrid myocardial perfusion imaging by correcting for attenuation artifacts. Artificial intelligence (AI) can automatically measure coronary artery calcium (CAC) from CTAC to improve risk prediction but could potentially derive additional anatomic features. OBJECTIVES The authors evaluated AI-based derivation of cardiac anatomy from CTAC and assessed its added prognostic utility. METHODS The authors considered consecutive patients without known coronary artery disease who underwent single-photon emission computed tomography/computed tomography (CT) myocardial perfusion imaging at 3 separate centers. Previously validated AI models were used to segment CAC and cardiac structures (left atrium, left ventricle, right atrium, right ventricular volume, and left ventricular [LV] mass) from CTAC. They evaluated associations with major adverse cardiovascular events (MACEs), which included death, myocardial infarction, unstable angina, or revascularization. RESULTS In total, 7,613 patients were included with a median age of 64 years. During a median follow-up of 2.4 years (IQR: 1.3-3.4 years), MACEs occurred in 1,045 (13.7%) patients. Fully automated AI processing took an average of 6.2 ± 0.2 seconds for CAC and 15.8 ± 3.2 seconds for cardiac volumes and LV mass. Patients in the highest quartile of LV mass and left atrium, LV, right atrium, and right ventricular volume were at significantly increased risk of MACEs compared to patients in the lowest quartile, with HR ranging from 1.46 to 3.31. The addition of all CT-based volumes and CT-based LV mass improved the continuous net reclassification index by 23.1%. CONCLUSIONS AI can automatically derive LV mass and cardiac chamber volumes from CT attenuation imaging, significantly improving cardiovascular risk assessment for hybrid perfusion imaging.
Collapse
Affiliation(s)
- Robert J H Miller
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Cardiac Sciences, University of Calgary, Calgary Alberta, Canada
| | - Aakash Shanbhag
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA; Signal and Image Processing Institute, Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, California, USA
| | - Aditya Killekar
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mark Lemley
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Bryan Bednarski
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paul B Kavanagh
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Attila Feher
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| | - Timothy Bateman
- Cardiovascular Imaging Technologies LLC, Kansas City, Missouri, USA
| | - Valerie Builoff
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joanna X Liang
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David E Newby
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Damini Dey
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniel S Berman
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Piotr J Slomka
- Department of Medicine (Division of Artificial Intelligence in Medicine), Imaging, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
40
|
Gorecka M, Craven TP, Jex N, Chew PG, Dobson LE, Brown LAE, Higgins DM, Thirunavukarasu S, Sharrack N, Javed W, Kotha S, Giannoudi M, Procter H, Parent M, Schlosshan D, Swoboda PP, Plein S, Levelt E, Greenwood JP. Mitral regurgitation assessment by cardiovascular magnetic resonance imaging during continuous in-scanner exercise: a feasibility study. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2024; 40:1543-1553. [PMID: 38780711 DOI: 10.1007/s10554-024-03141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Exercise imaging using current modalities can be challenging. This was patient focused study to establish the feasibility and reproducibility of exercise-cardiovascular magnetic resonance imaging (EX-CMR) acquired during continuous in-scanner exercise in asymptomatic patients with primary mitral regurgitation (MR). METHODS This was a prospective, feasibility study. Biventricular volumes/function, aortic flow volume, MR volume (MR-Rvol) and regurgitant fraction (MR-RF) were assessed at rest and during low- (Low-EX) and moderate-intensity exercise (Mod-EX) in asymptomatic patients with primary MR. RESULTS Twenty-five patients completed EX-CMR without complications. Whilst there were no significant changes in the left ventricular (LV) volumes, there was a significant increase in the LVEF (rest 63 ± 5% vs. Mod-EX 68 ± 6%;p = 0.01). There was a significant reduction in the right ventricular (RV) end-systolic volume (rest 68 ml(60-75) vs. Mod-EX 46 ml(39-59);p < 0.001) and a significant increase in the RV ejection fraction (rest 55 ± 5% vs. Mod-EX 65 ± 8%;p < 0.001). Whilst overall, there were no significant group changes in the MR-Rvol and MR-RF, individual responses were variable, with MR-Rvol increasing by ≥ 15 ml in 4(16%) patients and decreasing by ≥ 15 ml in 9(36%) of patients. The intra- and inter-observer reproducibility of LV volumes and aortic flow measurements were excellent, including at Mod-EX. CONCLUSION EX-CMR is feasible and reproducible in patients with primary MR. During exercise, there is an increase in the LV and RV ejection fraction, reduction in the RV end-systolic volume and a variable response of MR-Rvol and MR-RF. Understanding the individual variability in MR-Rvol and MR-RF during physiological exercise may be clinically important.
Collapse
Affiliation(s)
- Miroslawa Gorecka
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Thomas P Craven
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Nick Jex
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Pei G Chew
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Laura E Dobson
- Department of Cardiology, Wythenshawe Hospital, Manchester University NHS Trust, Manchester, UK
| | - Louise A E Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | | | - Sharmaine Thirunavukarasu
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Noor Sharrack
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Wasim Javed
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Sindhoora Kotha
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Marilena Giannoudi
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Henry Procter
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Martine Parent
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Dominik Schlosshan
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Peter P Swoboda
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Sven Plein
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - Eylem Levelt
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK
| | - John P Greenwood
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds Teaching Hospitals NHS Trust, Leeds, LS2 9JT, UK.
- Baker Heart and Diabetes Institute & Monash University, Melbourne, Australia.
| |
Collapse
|
41
|
Deshmukh T, Selvakumar D, Thavapalachandran S, Archer O, Figtree GA, Feneley M, Grieve SM, Thomas L, Pathan F, Chong JJH. Correlation of Noninvasive Cardiac MRI Measures of Left Ventricular Myocardial Function and Invasive Pressure-Volume Parameters in a Porcine Ischemia-Reperfusion Model. Radiol Cardiothorac Imaging 2024; 6:e230252. [PMID: 38842454 PMCID: PMC11211950 DOI: 10.1148/ryct.230252] [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: 08/22/2023] [Revised: 03/24/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
Purpose To assess the correlation between noninvasive cardiac MRI-derived parameters with pressure-volume (PV) loop data and evaluate changes in left ventricular function after myocardial infarction (MI). Materials and Methods Sixteen adult female swine were induced with MI, with six swine used as controls and 10 receiving platelet-derived growth factor-AB (PDGF-AB). Load-independent measures of cardiac function, including slopes of end-systolic pressure-volume relationship (ESPVR) and preload recruitable stroke work (PRSW), were obtained on day 28 after MI. Cardiac MRI was performed on day 2 and day 28 after infarct. Global longitudinal strain (GLS) and global circumferential strain (GCS) were measured. Ventriculo-arterial coupling (VAC) was derived from PV loop and cardiac MRI data. Pearson correlation analysis was performed. Results GCS (r = 0.60, P = .01), left ventricular ejection fraction (LVEF) (r = 0.60, P = .01), and cardiac MRI-derived VAC (r = 0.61, P = .01) had a significant linear relationship with ESPVR. GCS (r = 0.75, P < .001) had the strongest significant linear relationship with PRSW, followed by LVEF (r = 0.67, P = .005) and cardiac MRI-derived VAC (r = 0.60, P = .01). GLS was not significantly correlated with ESPVR or PRSW. There was a linear correlation (r = 0.82, P < .001) between VAC derived from cardiac MRI and from PV loop data. GCS (-3.5% ± 2.3 vs 0.5% ± 1.4, P = .007) and cardiac MRI-derived VAC (-0.6 ± 0.6 vs 0.3 ± 0.3, P = .001) significantly improved in the animals treated with PDGF-AB 28 days after MI compared with controls. Conclusion Cardiac MRI-derived parameters of MI correlated with invasive PV measures, with GCS showing the strongest correlation. Cardiac MRI-derived measures also demonstrated utility in assessing therapeutic benefit using PDGF-AB. Keywords: Cardiac MRI, Myocardial Infarction, Pressure Volume Loop, Strain Imaging, Ventriculo-arterial Coupling Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
- Tejas Deshmukh
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Dinesh Selvakumar
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Sujitha Thavapalachandran
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Oliver Archer
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Gemma A. Figtree
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Michael Feneley
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Stuart M. Grieve
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | - Liza Thomas
- From the Centre for Heart Research, Westmead Institute for Medical
Research, 176 Hawkesbury Rd, Westmead, Sydney, NSW 2145, Australia (T.D., D.S.,
S.T., J.J.H.C.); Department of Cardiology, Westmead Hospital, Westmead,
Australia (T.D., D.S., S.T., O.A., L.T., J.J.H.C.); Sydney School of Health
Sciences, Faculty of Medicine and Health, University of Sydney, Sydney,
Australia (T.D., D.S., S.T., L.T., J.J.H.C.); Cardiovascular Discovery Group,
Kolling Institute, University of Sydney and Royal North Shore Hospital, St
Leonards, Sydney, Australia (G.A.F.); Department of Cardiology, St
Vincent’s Hospital, Darlinghurst, Australia (M.F.); Cardiac Mechanics
Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
(M.F.); Imaging and Phenotyping Laboratory, Faculty of Medicine and Health,
Charles Perkins Centre, University of Sydney, Sydney, Australia (S.M.G.);
Department of Radiology, Royal Prince Alfred Hospital, Camperdown, Australia
(S.M.G.); Nepean Clinical School of Medicine, Charles Perkin Centre Nepean,
University of Sydney, Kingswood, Australia (F.P.); and Department of Cardiology,
Nepean Hospital, Kingswood, Australia (F.P.)
| | | | | |
Collapse
|
42
|
Wang Q, Tang TM, Youlton N, Weldy CS, Kenney AM, Ronen O, Weston Hughes J, Chin ET, Sutton SC, Agarwal A, Li X, Behr M, Kumbier K, Moravec CS, Wilson Tang WH, Margulies KB, Cappola TP, Butte AJ, Arnaout R, Brown JB, Priest JR, Parikh VN, Yu B, Ashley EA. Epistasis regulates genetic control of cardiac hypertrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.11.06.23297858. [PMID: 37987017 PMCID: PMC10659487 DOI: 10.1101/2023.11.06.23297858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The combinatorial effect of genetic variants is often assumed to be additive. Although genetic variation can clearly interact non-additively, methods to uncover epistatic relationships remain in their infancy. We develop low-signal signed iterative random forests to elucidate the complex genetic architecture of cardiac hypertrophy. We derive deep learning-based estimates of left ventricular mass from the cardiac MRI scans of 29,661 individuals enrolled in the UK Biobank. We report epistatic genetic variation including variants close to CCDC141 , IGF1R , TTN , and TNKS. Several loci where variants were deemed insignificant in univariate genome-wide association analyses are identified. Functional genomic and integrative enrichment analyses reveal a complex gene regulatory network in which genes mapped from these loci share biological processes and myogenic regulatory factors. Through a network analysis of transcriptomic data from 313 explanted human hearts, we found strong gene co-expression correlations between these statistical epistasis contributors in healthy hearts and a significant connectivity decrease in failing hearts. We assess causality of epistatic effects via RNA silencing of gene-gene interactions in human induced pluripotent stem cell-derived cardiomyocytes. Finally, single-cell morphology analysis using a novel high-throughput microfluidic system shows that cardiomyocyte hypertrophy is non-additively modifiable by specific pairwise interactions between CCDC141 and both TTN and IGF1R . Our results expand the scope of genetic regulation of cardiac structure to epistasis.
Collapse
|
43
|
Barros A, Udine M, Spurney C, Olivieri L, Loke YH. Discordance interpretation of left ventricular size between echocardiography and cardiac magnetic resonance in pediatric patients with aortic/mitral regurgitation. Int J Cardiovasc Imaging 2024; 40:1049-1057. [PMID: 38519822 PMCID: PMC11147931 DOI: 10.1007/s10554-024-03073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/22/2024] [Indexed: 03/25/2024]
Abstract
PURPOSE This study investigated discordance between echocardiography (echo) and cardiac magnetic resonance (CMR) measurements of the left ventricle (LV) in pediatric patients with aortic and/or mitral regurgitation (AR/MR). METHODS Retrospective cohort study of pediatric patients. The cohorts were comprised of patients with AR/MR vs. non-AR/MR. Left ventricular end diastolic volume (LVEDV) by CMR and left ventricular internal diameter diastolic (LVIDd) by echo were obtained from clinical reports then echo images were reviewed to remeasure LVEDV by bullet method. Left ventricular internal diameter systolic (LVIDs) and left ventricular ejection fraction (LVEF) measurements by echo and LVEF by CMR were obtained from clinical reports. Fractional shortening (FS%) was recalculated. Z-scores were calculated using normative data. Correlation between echo and CMR LV measurements was assessed using correlation coefficients. Bland-Altman plots assessed bias between imaging modalities. Receiver operator characteristic (ROC) analysis was performed for detection of LV enlargement and LV dysfunction. RESULTS AR/MR patients had greater discrepancy in LV size interpretation by Z-score compared to non-AR/MR patients. This discrepancy persisted when the bullet method short axis measurements were incorporated. There was negative bias in echo-based measurements compared to CMR. The diagnostic performance of echo in identifying moderate LV enlargement was worse for AR/MR pediatrics patients. CONCLUSION The discordant interpretation of LV size by echo compared to CMR is worse in pediatric patients with AR/MR when compared to patients without AR/MR even when short axis measurements are incorporated. This finding suggests non-uniform geometrical changes in the LV as it enlarges due to AR/MR.
Collapse
Affiliation(s)
| | | | | | - Laura Olivieri
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | - Yue-Hin Loke
- Children's National Hospital, Washington, DC, USA
| |
Collapse
|
44
|
Broncano J, Hanneman K, Ghoshhajra B, Rajiah PS. Cardiac Computed Tomography of Native Cardiac Valves. Radiol Clin North Am 2024; 62:399-417. [PMID: 38553177 DOI: 10.1016/j.rcl.2023.12.004] [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] [Indexed: 04/02/2024]
Abstract
Valvular heart disease (VHD) is a significant clinical problem associated with high morbidity and mortality. Although not being the primary imaging modality in VHD, cardiac computed tomography (CCT) provides relevant information about its morphology, function, severity grading, and adverse cardiac remodeling assessment. Aortic valve calcification quantification is necessary for grading severity in cases of low-flow/low-gradient aortic stenosis. Moreover, CCT details significant information necessary for adequate percutaneous treatment planning. CCT may help to detail the etiology of VHD as well as to depict other less frequent causes of valvular disease, such as infective endocarditis, valvular neoplasms, or other cardiac pseudomasses.
Collapse
Affiliation(s)
- Jordi Broncano
- Cardiothoracic Imaging Unit, Radiology Department, Hospital San Juan de Dios, HT Medica, Avenida El Brillante Nº 36, Córdoba 14012, Spain.
| | - Kate Hanneman
- Department of Medical Imaging, Toronto General Hospital, Peter Munk Cardiac Center, University Health Network (UHN), University of Toronto, 1 PMB-298, 585 University Avenue, Toronto, Ontario M5G2N2, Canada
| | - Brian Ghoshhajra
- Cardiovascular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charles River Plaza East, 165 Cambridge Street, Boston, MA 02114, USA
| | | |
Collapse
|
45
|
Christersson M, Gustafsson S, Lampa E, Almstedt M, Cars T, Bodegård J, Arefalk G, Sundström J. Usefulness of Heart Failure Categories Based on Left Ventricular Ejection Fraction. J Am Heart Assoc 2024; 13:e032257. [PMID: 38591322 PMCID: PMC11262517 DOI: 10.1161/jaha.123.032257] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/03/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Heart failure guidelines have recently introduced a narrow category with mildly reduced left ventricular ejection fraction (LVEF) (heart failure with mildly reduced ejection fraction; LVEF 41%-49%) between the previous categories of reduced (heart failure with reduced ejection fraction; LVEF ≤40%) and preserved (heart failure with preserved ejection fraction; LVEF ≥50%) ejection fraction. Grouping of continuous measurements into narrow categories can be questioned if their variability is high. METHODS AND RESULTS We constructed a cohort of all 9716 new cases of chronic heart failure with an available LVEF in Stockholm, Sweden, from January 1, 2015, until December 31, 2020. All values of LVEF were collected over time, and patients were followed up until death, moving out of Stockholm, or end of study. Mixed models were used to quantify within-person variance in LVEF, and multistate Markov models, with death as an absorbing state, to quantify the stability of LVEF categories. LVEF values followed a normal distribution. The SD of the within-person variance in LVEF over time was 7.4%. The mean time spent in any LVEF category before transition to another category was on average <1 year for heart failure with mildly reduced ejection fraction. Probabilities of transitioning between categories during the first year were substantial; patients with heart failure with mildly reduced ejection fraction had a probability of <25% of remaining in that category 1 year later. CONCLUSIONS LVEF follows a normal distribution and has considerable variability over time, which may impose a risk for underuse of efficient treatment. The heart failure with mildly reduced ejection fraction category is especially inconstant. Assumptions of a patient's current LVEF should take this variability and the normal distribution of LVEF into account.
Collapse
Affiliation(s)
| | | | - Erik Lampa
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | | | | | - Johan Bodegård
- Cardiovascular, Renal and Metabolism, Medical DepartmentBioPharmaceuticals, AstraZenecaOsloNorway
| | - Gabriel Arefalk
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Johan Sundström
- Department of Medical SciencesUppsala UniversityUppsalaSweden
- The George Institute for Global Health, University of New South WalesSydneyAustralia
| |
Collapse
|
46
|
Liu J, Li J, Xia C, He W, Li X, Shen S, Zhou X, Tong N, Peng L. The effect of hyperlipidemia and body fat distribution on subclinical left ventricular function in obesity: a cardiovascular magnetic resonance study. Cardiovasc Diabetol 2024; 23:120. [PMID: 38566090 PMCID: PMC10985902 DOI: 10.1186/s12933-024-02208-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Obesity is often associated with multiple comorbidities. However, whether obese subjects with hyperlipidemia in the absence of other complications have worse cardiac indices than metabolically healthy obese subjects is unclear. Therefore, we aimed to determine the effect of hyperlipidemia on subclinical left ventricular (LV) function in obesity and to evaluate the association of cardiac parameters with body fat distribution. MATERIALS AND METHODS Ninety-two adults were recruited and divided into 3 groups: obesity with hyperlipidemia (n = 24, 14 males), obesity without hyperlipidemia (n = 25, 13 males), and c ntrols (n = 43, 25 males). LV strain parameters (peak strain (PS), peak diastolic strain rate (PDSR), peak systolic strain rate) derived from cardiovascular magnetic resonance tissue tracking were measured and compared. Dual-energy X-ray absorptiometer was used to measure body fat distribution. Correlations of hyperlipidemia and body fat distribution with LV strain were assessed by multivariable linear regression. RESULTS Obese individuals with preserved LV ejection fraction showed lower global LV longitudinal, circumferential, and radial PS and longitudinal and circumferential PDSR than controls (all P < 0.05). Among obese patients, those with hyperlipidemia had lower longitudinal PS and PDSR and circumferential PDSR than those without hyperlipidemia (- 12.8 ± 2.9% vs. - 14.2 ± 2.7%, 0.8 ± 0.1 s-1 vs. 0.9 ± 0.3 s-1, 1.2 ± 0.2 s-1 vs. 1.4 ± 0.2 s-1; all P < 0.05). Multivariable linear regression demonstrated that hyperlipidemia was independently associated with circumferential PDSR (β = - 0.477, P < 0.05) in obesity after controlling for growth differences, other cardiovascular risk factors, and central fat distribution. In addition, android fat had an independently negative relationship with longitudinal and radial PS (β = - 0.486 and β = - 0.408, respectively; all P < 0.05); and visceral fat was negatively associated with longitudinal PDSR (β = - 0.563, P < 0.05). Differently, gynoid fat was positively correlated with circumferential PS and PDSR and radial PDSR (β = 0.490, β = 0.481, and β = 0.413, respectively; all P < 0.05). CONCLUSION Hyperlipidemia is independently associated with subclinical LV diastolic dysfunction in obesity. Central fat distribution (android and visceral fat) has a negative association, while peripheral fat distribution (gynoid fat) has a positive association on subclinical LV function. These results suggest that appropriate management of hyperlipidemia may be beneficial for obese patients, and that the differentiation of fat distribution in different regions may facilitate the precise management of obese patients. Clinical trials registration Effect of lifestyle intervention on metabolism of obese patients based on smart phone software (ChiCTR1900026476).
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Jing Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Wenzhang He
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Xue Li
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Sumin Shen
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China
| | - Xiaoyue Zhou
- MR Collaboration, Siemens Healthineers Ltd., Shanghai, 200126, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China.
| | - Liqing Peng
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, China.
| |
Collapse
|
47
|
Hall M, de Marvao A, Schweitzer R, Cromb D, Colford K, Jandu P, O’Regan DP, Ho A, Price A, Chappell LC, Rutherford MA, Story L, Lamata P, Hutter J. Preeclampsia Associated Differences in the Placenta, Fetal Brain, and Maternal Heart Can Be Demonstrated Antenatally: An Observational Cohort Study Using MRI. Hypertension 2024; 81:836-847. [PMID: 38314606 PMCID: PMC7615760 DOI: 10.1161/hypertensionaha.123.22442] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Preeclampsia is a multiorgan disease of pregnancy that has short- and long-term implications for the woman and fetus, whose immediate impact is poorly understood. We present a novel multiorgan approach to magnetic resonance imaging (MRI) investigation of preeclampsia, with the acquisition of maternal cardiac, placental, and fetal brain anatomic and functional imaging. METHODS An observational study was performed recruiting 3 groups of pregnant women: those with preeclampsia, chronic hypertension, or no medical complications. All women underwent a cardiac MRI, and pregnant women underwent a placental-fetal MRI. Cardiac analysis for structural, morphological, and flow data were undertaken; placenta and fetal brain volumetric and T2* (which describes relative tissue oxygenation) data were obtained. All results were corrected for gestational age. A nonpregnant cohort was identified for inclusion in the statistical shape analysis. RESULTS Seventy-eight MRIs were obtained during pregnancy. Cardiac MRI analysis demonstrated higher left ventricular mass in preeclampsia with 3-dimensional modeling revealing additional specific characteristics of eccentricity and outflow track remodeling. Pregnancies affected by preeclampsia demonstrated lower placental and fetal brain T2*. Within the preeclampsia group, 23% placental T2* results were consistent with controls, these were the only cases with normal placental histopathology. Fetal brain T2* results were consistent with normal controls in 31% of cases. CONCLUSIONS We present the first holistic assessment of the immediate implications of preeclampsia on maternal heart, placenta, and fetal brain. As well as having potential clinical implications for the risk stratification and management of women with preeclampsia, this gives an insight into the disease mechanism.
Collapse
Affiliation(s)
- Megan Hall
- Department of Women and Children’s Health (M.H., A.d.M., A.H., L.C.C., L.S.), King’s College London, United Kingdom
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Antonio de Marvao
- Department of Women and Children’s Health (M.H., A.d.M., A.H., L.C.C., L.S.), King’s College London, United Kingdom
- School of Cardiovascular Medicine (A.d.M., R.S.), King’s College London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, United Kingdom (A.d.M., R.S., D.P.O.)
| | - Ronny Schweitzer
- School of Cardiovascular Medicine (A.d.M., R.S.), King’s College London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, United Kingdom (A.d.M., R.S., D.P.O.)
| | - Daniel Cromb
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Kathleen Colford
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Priya Jandu
- GKT School of Medical Education (P.J.), King’s College London, United Kingdom
| | - Declan P O’Regan
- MRC London Institute of Medical Sciences, Imperial College London, United Kingdom (A.d.M., R.S., D.P.O.)
| | - Alison Ho
- Department of Women and Children’s Health (M.H., A.d.M., A.H., L.C.C., L.S.), King’s College London, United Kingdom
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Anthony Price
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
- Centre for Medical Engineering (A.P., P.L.), King’s College London, United Kingdom
| | - Lucy C. Chappell
- Department of Women and Children’s Health (M.H., A.d.M., A.H., L.C.C., L.S.), King’s College London, United Kingdom
| | - Mary A. Rutherford
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Lisa Story
- Department of Women and Children’s Health (M.H., A.d.M., A.H., L.C.C., L.S.), King’s College London, United Kingdom
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
| | - Pablo Lamata
- Centre for Medical Engineering (A.P., P.L.), King’s College London, United Kingdom
| | - Jana Hutter
- Centre for the Developing Brain (M.H., D.C., K.C., A.H., A.P., M.A.R., L.S., J.H.), King’s College London, United Kingdom
- Smart Imaging Lab, Radiological Institute, University Hospital Erlangen, Germany (J.H.)
| |
Collapse
|
48
|
Garrity K, Docherty C, Mangion K, Woodward R, Shaw M, Roditi G, Shelley B, Quasim T, McCall P, McPeake J. Characterizing Cardiac Function in ICU Survivors of Sepsis: A Pilot Study Protocol. CHEST CRITICAL CARE 2024; 2:100050. [PMID: 38524255 PMCID: PMC10958646 DOI: 10.1016/j.chstcc.2024.100050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Background Sepsis is one of the most common reasons for ICU admission and a leading cause of mortality worldwide. More than one-half of survivors experience significant physical, psychological, or cognitive impairments, often termed post-intensive care syndrome (PICS). Sepsis is recognized increasingly as being associated with a risk of adverse cardiovascular events that is comparable with other major cardiovascular risk factors. It is plausible that sepsis survivors may be at risk of unidentified cardiovascular disease, and this may play a role in functional impairments seen after ICU discharge. Research Question What is the prevalence of myocardial dysfunction after an ICU admission with sepsis and to what extent might it be associated with physical impairments in PICS? Study Design and Methods Characterisation of Cardiovascular Function in ICU Survivors of Sepsis (CONDUCT-ICU) is a prospective, multicenter, pilot study characterizing cardiovascular function and functional impairments in survivors of sepsis taking place in the west of Scotland. Survivors of sepsis will be recruited at ICU discharge and followed up 6 to 10 weeks after hospital discharge. Biomarkers of myocardial injury or dysfunction (high sensitivity troponin and N-terminal pro B-type natriuretic peptide) and systemic inflammation (C-reactive protein, IL-1β, IL-6, IL-10, and tumor necrosis factor alpha) will be measured in 69 patients at recruitment and at follow-up. In addition, a cardiovascular magnetic resonance substudy will be performed at follow-up in 35 patients. We will explore associations between cardiovascular magenetic resonance indexes of cardiac function, biomarkers of cardiac dysfunction and inflammation, and patient-reported outcome measures. Interpretation CONDUCT-ICU will provide data regarding the cause and prevalence of cardiac dysfunction in survivors of sepsis and will explore associations with functional impairment. It will provide feasibility data and operational learning for larger studies investigating mechanisms of functional impairment after ICU admission and the association between sepsis and adverse cardiovascular events. Trial Registry ClinicalTrials.gov; No.: NCT05633290; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Kevin Garrity
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
- Glasgow Royal Infirmary, NHS Greater Glasgow & Clyde, Glasgow
| | - Christie Docherty
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
- University Hospital Crosshouse; NHS Ayrshire and Arran, Crosshouse
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow
| | - Rosie Woodward
- Imaging Centre of Excellence, Queen Elizabeth University Hospital; NHS Greater Glasgow & Clyde, Glasgow
| | - Martin Shaw
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
| | - Giles Roditi
- Imaging Centre of Excellence, Queen Elizabeth University Hospital; NHS Greater Glasgow & Clyde, Glasgow
| | - Benjamin Shelley
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
- Golden Jubilee National Hospital, NHS Scotland, Clydebank, Scotland
| | - Tara Quasim
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
- Glasgow Royal Infirmary, NHS Greater Glasgow & Clyde, Glasgow
| | - Philip McCall
- Academic Unit of Anaesthesia, Critical Care and Peri-Operative Medicine, University of Glasgow, Glasgow
- Golden Jubilee National Hospital, NHS Scotland, Clydebank, Scotland
| | - Joanne McPeake
- THIS Institute, University of Cambridge, Cambridge, England
| |
Collapse
|
49
|
Moustaki M, Markousis-Mavrogenis G, Vryonidou A, Paschou SA, Mavrogeni S. Cardiac disease in Cushing's syndrome. Emphasis on the role of cardiovascular magnetic resonance imaging. Endocrine 2024; 83:548-558. [PMID: 38129722 DOI: 10.1007/s12020-023-03623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Cushing's Syndrome (CS) is associated with increased cardiovascular morbidity and mortality. In endogenous CS, cardiovascular mortality remains increased for up to 15 years post remission of hypercortisolism. Similarly, patients with exogenous CS have 4-fold increased incidence of cardiovascular events, regardless of pre-existing cardiovascular disease (CVD). OBJECTIVE To present the pathophysiology, prognosis, clinical and imaging phenotype of cardiac disease in CS. METHODS A Pubmed search for cardiac disease in CS over the last 20 years was conducted using combinations of relevant terms. Preclinical and clinical studies, as well as review papers reporting on subclinical heart failure (HF), cardiomyopathy, coronary heart disease (CHD), and cardiovascular imaging were selected. RESULTS Cardiac disease in CS is associated with direct mineralocorticoid and glucocorticoid receptor activation, increased responsiveness to angiotensin II, ectopic epicardial adiposity, arterial stiffness and endothelial dysfunction, as well as with diabetes mellitus, hypertension, hyperlipidemia, obesity and prothrombotic diathesis. Subclinical HF and cardiomyopathy are principally related to direct glucocorticoid (GC) effects and markedly improve or regress post hypercortisolism remission. In contrast, CHD is related to both direct GC effects and CS comorbidities and persists post cure. In patients without clinical evidence of CVD, echocardiography and cardiac magnetic resonance (CMR) imaging reveal left ventricular hypertrophy, fibrosis, diastolic and systolic dysfunction, with the latter being underestimated by echocardiography. Finally, coronary microvascular disease is encountered in one third of cases. CONCLUSION Cardiovascular imaging is crucial in evaluation of cardiac involvement in CS. CMR superiority in terms of reproducibility, operator independency, unrestricted field of view and capability of tissue characterisation makes this modality ideal for future studies.
Collapse
Affiliation(s)
- Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece.
| | - George Markousis-Mavrogenis
- University Research Institute of Maternal and Child Health and Precision Medicine and UNESCO Chair in Adolescent Health Care, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophie Mavrogeni
- University Research Institute of Maternal and Child Health and Precision Medicine and UNESCO Chair in Adolescent Health Care, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
- Onassis Cardiac Surgery Center, Athens, Greece
| |
Collapse
|
50
|
Reding KW, Jordan JH. Multimodal Imaging Evidence for Optimized Blood Pressure Control Following Hypertensive Pregnancy: Mechanistic Insights Into Beneficial Cardiac Remodeling From the POP-HT Trial. Circulation 2024; 149:542-544. [PMID: 38346108 DOI: 10.1161/circulationaha.124.068282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Affiliation(s)
- Kerryn W Reding
- Department of Biobehavioral Nursing and Health Informatics, University of Washington School of Nursing, Seattle (K.W.R.)
- Division of Public Health Sciences, Fred Hutch Cancer Center, Seattle, WA (K.W.R.)
| | - Jennifer H Jordan
- Department of Biomedical Engineering, College of Engineering (J.H.J.), Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond
- Department of Internal Medicine, Division of Cardiology (J.H.J.), Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond
| |
Collapse
|