1
|
Yasuda N, Kato S, Horita N, Sekii R, Sawamura S, Nagase H, Utsunomiya D. Synthetic extracellular volume fraction as an imaging biomarker of the myocardial interstitium without blood sampling: a systematic review and meta-analysis. J Cardiovasc Magn Reson 2025; 27:101889. [PMID: 40139292 DOI: 10.1016/j.jocmr.2025.101889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The calculation of conventional extracellular volume fraction (ECV) requires blood hematocrit (Hct) measurement. Based on the relationship between Hct and blood T1 relaxivity for cardiac magnetic resonance (CMR), a synthetic ECV could be estimated without a blood sampling. The aim of this study was to evaluate the correlation and agreement in the quantification of synthetic ECV and laboratory ECV from conventional Hct measurements. METHODS Electronic database searches of PubMed, Web of Science Core Collection, Cochrane advanced search, and EMBASE were performed. The authors employed a meta-analysis using the generic inverse variance method with a random-effects model to estimate the summary correlation coefficient and mean absolute difference between synthetic and laboratory ECV. RESULTS Of 38 papers, 10 studies comprising 4492 patients were identified. Overall, there was an excellent correlation between synthetic ECV and laboratory ECV (0.95 [95% confidence interval (CI): 0.92 to 0.97]) at 1.5T CMR and (0.91 [95% CI: 0.86 to 0.94]) at 3.0T CMR. The pooled mean difference between synthetic ECV and laboratory ECV was 0.61% (95% CI: 0.23 to 0.98%, I2 = 0%, p for heterogeneity = 0.67) at 1.5T CMR and 0.24% (95% CI: -0.13 to 0.61%, I2 = 19%, p for heterogeneity = 0.25) at 3.0T CMR. CONCLUSION This study is the first comprehensive systematic review and meta-analysis of synthetic ECV evaluation at CMR. Synthetic ECV demonstrated an excellent correlation with laboratory ECV, with a mean difference of less than 1%, and offers noninvasive and instantaneous quantification of the myocardial extracellular space without the need for blood sampling.
Collapse
Affiliation(s)
- Naofumi Yasuda
- Department of Diagnostic Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Shingo Kato
- Department of Diagnostic Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.
| | - Nobuyuki Horita
- Chemotherapy Center, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | - Ryusuke Sekii
- Department of Cardiology, Yokohama Hodogaya Central Hospital, Yokohama, Kanagawa, Japan
| | - Shungo Sawamura
- Department of Diagnostic Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Hiroaki Nagase
- Department of Diagnostic Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Daisuke Utsunomiya
- Department of Diagnostic Radiology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
2
|
Li B, Gao Y, Wang J, Zhu R, Yang S, Ji C, Wang Y, Wang X, Gu H. Prognostic value of extracellular volume fraction in myocardial infarction and myocardial infarction with nonobstructive coronary arteries: A multicenter study. Acad Radiol 2025; 32:1313-1323. [PMID: 39580248 DOI: 10.1016/j.acra.2024.11.015] [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: 08/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/25/2024]
Abstract
RATIONALE AND OBJECTIVES The aim of the present retrospective study was to evaluate the prognostic role of the extracellular volume fraction (ECV) in patients with myocardial infarction (MI) and myocardial infarction with nonobstructive coronary arteries (MINOCA). The present study hypothesized that ECV is associated with major adverse cardiovascular events (MACEs) in MI and MINOCA patients. MATERIALS AND METHODS Cardiac magnetic resonance (CMR) imaging was performed on 351 consecutive patients (mean age: 58 ± 12 years; 252 [71.8%] males) who were diagnosed with MI between October 2015 and November 2023. From CMR imaging, the extent of late gadolinium enhancement (LGE), native T1 and ECV were derived. Patients were categorized into groups according to the degree of coronary artery stenosis, namely, patients with MINOCA and patients with obstructive MI. Follow-up was performed to assess MACEs. RESULTS The final cohort consisted of 61 MINOCA patients and 290 obstructive MI patients. During a mean follow-up of 27 ± 16 months, there was no statistically significant difference in the incidence of MACEs between patients with MINOCA and those with obstructive MI, and the two groups of patients had similar ECVs (32.2 ± 3.6 vs. 32.3 ± 6.0, p = 0.864). According to the multivariate Cox regression, ECV was an independent predictor of MACEs (HR: 1.13; p < 0.001) and significantly improved the prognostic value of the baseline multivariate models (C-statistic improvement: 0.816-0.864, p = 0.001). Similarly, ECV maintained an independent association with MACEs in the MINOCA (HR: 1.35; p < 0.001) and obstructive MI (HR: 1.13; p < 0.001) groups. CONCLUSION In MI and MINOCA patients, ECV is an independent predictor of MACEs. MINOCA is not a benign disease, and its long-term prognosis is as poor as that of patients with obstructive MI.
Collapse
Affiliation(s)
- Bowen Li
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan Shandong 250117, China (B.L., R.Z.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Yan Gao
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Jian Wang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (J.W.)
| | - Runze Zhu
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan Shandong 250117, China (B.L., R.Z.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Shifeng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Congshan Ji
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Ying Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.)
| | - Hui Gu
- Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China (Y.G., X.W., H.G.); Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (B.L., Y.G., R.Z., S.Y., C.J., Y.W., X.W., H.G.).
| |
Collapse
|
3
|
Wang Y, Duan X, Zhu L, Xu J, Zhou D, Yang W, Jiang M, Zhang H, Sirajuddin A, Arai AE, Zhao S, Wang H, Lu M. Prognostic Value of Myocardial Parametric Mapping in Patients with Acute Myocarditis: A Retrospective Study. Radiol Cardiothorac Imaging 2025; 7:e240125. [PMID: 39912725 PMCID: PMC11880863 DOI: 10.1148/ryct.240125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 02/07/2025]
Abstract
Purpose To investigate the prognostic value of T1 mapping, extracellular volume fraction (ECV), and T2 mapping in a large cohort of patients with acute myocarditis. Materials and Methods This retrospective study included patients with acute myocarditis who underwent cardiac MRI (3.0 T) between March 2016 and October 2022. Diagnosis was confirmed by diagnostic cardiac MRI criteria or endomyocardial biopsy. The primary end point was major adverse cardiovascular events (MACEs), defined as the composite of cardiac death, heart failure hospitalization, heart transplantation, sustained ventricular arrhythmia, and recurrent myocarditis. Univariable and multivariable Cox regression analyses were performed to assess the association of clinical and cardiac MRI variables with the primary end point. The prognostic value of each model was assessed using the Harrell C index. Results A total of 235 patients (mean age, 32 years ± 13 [SD]; 150 [63.8%] men) were included. During a mean follow-up of 1637 days (IQR: 1441-1833 days), MACEs occurred in 45 (19%) patients. Patients with MACEs had higher global native T1, ECV, and T2 values (1342 msec ± 64 vs 1263 msec ± 48; P < .001; 39.1% ± 8.7 vs 32.7% ± 5.7; P < .001; 61.1 msec ± 10.0 vs 55.3 msec ± 9.4; P = .03, respectively). In a series of multivariable Cox regression models, native T1 (per 10-msec increase: hazard ratio, 1.61; 95% CI: 1.31, 1.98; P < .001) and ECV (per 5% increase: hazard ratio, 1.70; 95% CI: 1.38, 2.08; P < .001) independently predicted MACE occurrence, and the addition of native T1 (Harrell C index = 0.76) or ECV (Harrell C index = 0.79) to the model including only clinical variables, left ventricular ejection fraction, and septal late gadolinium enhancement (Harrell C index = 0.72) improved discrimination for the primary end point. Conclusion Cardiac MRI-derived native T1 and ECV were independent predictors of MACEs in patients with acute myocarditis and provided incremental prognostic value when combined with conventional parameters. Keywords: MRI, Cardiac, Heart, Inflammation Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Yining Wang
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, State Key
Laboratory of Cardiovascular Disease, National Center for Cardiovascular
Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College,
Beijing, China
| | - Leyi Zhu
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Jing Xu
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Di Zhou
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Wenjing Yang
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Mengdi Jiang
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Huaying Zhang
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
| | - Arlene Sirajuddin
- Department of Health and Human Services, National Heart,
Lung and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Andrew E. Arai
- Johns Hopkins Medicine-Suburban Hospital, Kensington,
Md
- Division of Cardiovascular Medicine, University of Utah
School of Medicine, Salt Lake City, Utah
| | - Shihua Zhao
- Johns Hopkins Medicine-Suburban Hospital, Kensington,
Md
| | - Hongyue Wang
- Department of Pathology, Fuwai Hospital, State Key
Laboratory of Cardiovascular Disease, National Center for Cardiovascular
Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College,
Beijing, China
| | - Minjie Lu
- Department of Magnetic Resonance Imaging, Fuwai Hospital,
State Key Laboratory of Cardiovascular Disease, National Center for
Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100037, China
- Key Laboratory of Cardiovascular Imaging (Cultivation),
Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Helali J, Ramesh K, Brown J, Preciado-Ruiz C, Nguyen T, Silva LT, Ficara A, Wesbey G, Gonzalez JA, Bilchick KC, Salerno M, Robinson AA. Late gadolinium enhancement on cardiac MRI: A systematic review and meta-analysis of prognosis across cardiomyopathies. Int J Cardiol 2025; 419:132711. [PMID: 39515615 DOI: 10.1016/j.ijcard.2024.132711] [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: 07/31/2024] [Revised: 10/12/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Late gadolinium enhancement (LGE) on cardiac MRI has been shown to predict adverse outcomes in a range of cardiac diseases. However, no study has systematically reviewed and analyzed the literature across all cardiac pathologies including rare diseases. METHODS PubMed, EMBASE and Web of Science were searched for studies evaluating the relationship between LGE burden and cardiovascular outcomes. Outcomes included all-cause mortality, MACE, sudden cardiac death, sustained VT or VF, appropriate ICD shock, heart transplant, and heart failure hospitalization. Only studies reporting hazards ratios with LGE as a continuous variable were included. RESULTS Of the initial 8928 studies, 95 studies (23,313 patients) were included across 19 clinical entities. The studies included ischemic cardiomyopathy (7182 patients, 33 studies), hypertrophic cardiomyopathy (5080 patients, 17 studies), non-ischemic cardiomyopathy not otherwise specified (2627 patients, 11 studies), and dilated cardiomyopathy (2345 patients, 14 studies). Among 42 studies that quantified LGE by percent myocardium, a 1 % increase in LGE burden was associated with life-threatening ventricular arrhythmias (LTVA) with a pooled hazard ratio of 1.04 (CI 1.02-1.05), and MACE with a pooled hazard ratio of 1.06 (CI 1.04-1.07). The risk of these events was similar across disease types, with minimal heterogeneity. CONCLUSIONS Despite mechanistic differences in myocardial injury, LGE appears to have a fairly consistent, dose-dependent effect on risk of LTVA, MACE, and mortality. These data can be applied to derive a patient's absolute risk of LTVA, and therefore can be clinically useful in informing decisions on primary prevention ICD implantation irrespective of the disease etiology.
Collapse
Affiliation(s)
- Joshua Helali
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America
| | - Karthik Ramesh
- University of California San Diego School of Medicine, La Jolla, CA, United States of America
| | - John Brown
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | | | - Thornton Nguyen
- University of California Riverside, Riverside, CA, United States of America
| | - Livia T Silva
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America; University of California San Diego, La Jolla, CA, United States of America
| | - Austin Ficara
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America
| | - George Wesbey
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America; Department of Radiology, Scripps Clinic, La Jolla, CA, United States of America
| | - Jorge A Gonzalez
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America; Department of Radiology, Scripps Clinic, La Jolla, CA, United States of America
| | - Kenneth C Bilchick
- Department of Cardiovascular Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Michael Salerno
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Austin A Robinson
- Division of Cardiology, Scripps Clinic, La Jolla, CA, United States of America.
| |
Collapse
|
5
|
Unger A, Garot J, Toupin S, Duhamel S, Sanguineti F, Hovasse T, Champagne S, Unterseeh T, Chevalier B, Akodad M, Neylon A, Hamzi L, Gonçalves T, Florence J, Gall E, Martial PJ, Mirailles R, Pfeffer A, Canuti ES, Dillinger JG, Henry P, Bondue A, Bousson V, Garot P, Pezel T. Prognostic Value of Cardiac MRI Late Gadolinium Enhancement Granularity in Participants with Ischemic Cardiomyopathy. Radiology 2025; 314:e240806. [PMID: 39772797 DOI: 10.1148/radiol.240806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Background Ischemic late gadolinium enhancement (LGE) assessed with cardiac MRI is a well-established prognosticator in ischemic cardiomyopathy. However, the prognostic value of additional LGE parameters, such as extent, transmurality, location, and associated midwall LGE, remains unclear. Purpose To assess the prognostic value of ischemic LGE features to predict all-cause mortality in ischemic cardiomyopathy. Materials and Methods This study is a secondary analysis of a prospective dual-center trial of participants with ischemic cardiomyopathy and left ventricular ejection fraction (LVEF) under 50% referred for viability assessment using cardiac MRI between 2008 and 2022. The LGE granularity parameters (extent of ischemic LGE, transmurality, location, and associated midwall LGE) assessed by cardiac MRI experts were compared with traditional prognosticators of adverse events in ischemic cardiomyopathy (age, sex, body mass index, diabetes, smoking, dyslipidemia, heart failure hospitalization, atrial fibrillation, renal failure, known myocardial infarction, and LVEF). The primary outcome was all-cause mortality. Predictive value was evaluated using Cox regression analysis and assessed using time-dependent receiver operating characteristic curves at 10 years. The cardiac MRI LGE score was developed using LGE granularity parameters. Results Among 6082 participants (mean age, 64.5 years ± 11.8 [SD]; 4419 men), 3591 had ischemic LGE. During a median follow-up of 9.0 years (IQR, 6.6-11.5 years), 652 participants died. The presence of ischemic LGE was strongly associated with mortality (hazard ratio, 3.45 [99.5% CI: 2.55, 4.67]; P < .001). In the group with ischemic LGE, the LGE granularity model combining these LGE features showed the best predictive value above traditional prognosticators and ischemic LGE extent to predict all-cause mortality (area under the receiving operating characteristic curve [AUC] at 10 years, 0.89 [99.5% CI: 0.89, 0.90] vs 0.83 [99.5% CI: 0.83, 0.84]; P < .001). The cardiac MRI LGE score performed well in participants with ischemic LGE (AUC at 10 years, 0.87 [99.5% CI: 0.85, 0.90]). Conclusion In a large cohort of participants with ischemic cardiomyopathy, an LGE granularity model had a higher prognostic value over traditional prognosticators to predict mortality. © RSNA, 2025 Supplemental material is available for this article. See also the editorial by Lima and Ebrahimihoor in this issue.
Collapse
Affiliation(s)
- Alexandre Unger
- Department of Cardiology, Hôpital universitaire de Bruxelles, Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Jérôme Garot
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | | | - Suzanne Duhamel
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Francesca Sanguineti
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Thomas Hovasse
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Stéphane Champagne
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Thierry Unterseeh
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Bernard Chevalier
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Mariama Akodad
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Antoinette Neylon
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Lounis Hamzi
- Université Paris-Cité, Department of Radiology, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Trecy Gonçalves
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
- Université Paris-Cité, Department of Radiology, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Jeremy Florence
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Emmanuel Gall
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Paul-Jun Martial
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Raphael Mirailles
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Alexandre Pfeffer
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Elena Sofia Canuti
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Jean Guillaume Dillinger
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Patrick Henry
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Antoine Bondue
- Department of Cardiology, Hôpital universitaire de Bruxelles, Hôpital Erasme, Université libre de Bruxelles, Brussels, Belgium
| | - Valérie Bousson
- Université Paris-Cité, Department of Radiology, University Hospital of Lariboisière, AP-HP, Paris, France
| | - Philippe Garot
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
| | - Théo Pezel
- Université Paris-Cité, Department of Cardiology, University Hospital of Lariboisière, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- Inserm MASCOT-UMRS 942, University Hospital of Lariboisière, Paris, France
- MIRACL.ai Laboratory, Multimodality Imaging for Research and Artificial Intelligence Core Laboratory, University Hospital of Lariboisière, AP-HP, Paris, France
- Institut Cardiovasculaire Paris Sud, Cardiovascular Magnetic Resonance Laboratory, Hôpital Privé Jacques Cartier, Ramsay Santé, Massy, France
- Université Paris-Cité, Department of Radiology, University Hospital of Lariboisière, AP-HP, Paris, France
| |
Collapse
|
6
|
El Kadi S, Porter TR, Hopman LHGA, Verouden NC, van Rossum AC, Danad I, Kamp O. Echocardiography-derived regional strain for assessment of non-culprit stenosis and prediction of systolic function recovery in acute STEMI. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2025; 41:63-72. [PMID: 39630353 DOI: 10.1007/s10554-024-03287-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/11/2024] [Indexed: 01/19/2025]
Abstract
To compare echocardiographic regional longitudinal strain with quantitative coronary angiography and assess temporal changes in regional strain in patients with STEMI and multivessel coronary artery disease. Thirty-two patients with STEMI and multivessel coronary artery disease underwent coronary angiography with 3D quantification and baseline echocardiography. Regional longitudinal strain was measured as the average strain of three adjacent myocardial segments (RLS-3S) with the most impaired strain values. Forty-one stenosed vessels were identified (9 LAD [19%], 21 LCx [50%] and 11 RCA [31%]). RLS-3S did not correlate with diameter stenosis, area stenosis or minimal luminal diameter. Receiver operating curve analysis of RLS-3S for hemodynamic significant lesions (defined as positive fractional flow reserve or composite of ≥ 70% diameter stenosis and minimal luminal diameter < 1.2 mm) demonstrated an area under the curve of 0.63 (95% CI 0.45-0.76) with an optimal cut-off value of < 9.8%. Sensitivity and specificity of RLS-3S was 86% (42-100) and 48% (31-66). RLS-3S < 9.8% at baseline in remote myocardium subtended by the stenosed coronary vessel predicted benefit from percutaneous coronary intervention in terms of regional functional recovery. RLS-3S does not correlate with individual coronary angiography measures and moderately predicts hemodynamically significant lesions. RLS-3S could be used to predict regional functional recovery after additional revascularization.
Collapse
Affiliation(s)
- Soufiane El Kadi
- Amsterdam UMC, location VUmc, Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
- Department of Cardiology, Amsterdam UMC - location VUmc, ZH 4C 99, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Thomas R Porter
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Luuk H G A Hopman
- Amsterdam UMC, location VUmc, Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Niels C Verouden
- Amsterdam UMC, location VUmc, Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Albert C van Rossum
- Amsterdam UMC, location VUmc, Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Ibrahim Danad
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Otto Kamp
- Amsterdam UMC, location VUmc, Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Kamdar A, Sykes R, Thomson CR, Mangion K, Ang D, Lee MAW, Van Agtmael T, Berry C. Vascular fibrosis and extracellular matrix remodelling in post-COVID 19 conditions. INFECTIOUS MEDICINE 2024; 3:100147. [PMID: 39649442 PMCID: PMC11621938 DOI: 10.1016/j.imj.2024.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 12/10/2024]
Abstract
Causal associations between viral infections and acute myocardial injury are not fully understood, with mechanisms potentially involving direct cardiovascular involvement or systemic inflammation. This review explores plausible mechanisms of vascular fibrosis in patients with post-COVID-19 syndrome, focusing on extracellular matrix remodelling. Despite global attention, significant mechanistic or translational breakthroughs in the management of post-viral syndromes remain limited. No effective pharmacological or non-pharmacological interventions are currently available for patients experiencing persistent symptoms following COVID-19 infection. The substantial expansion of scientific knowledge resulting from collaborative efforts by medical experts, scientists, and government organisations in undertaking COVID-19 research could inform treatment strategies for other post-viral syndromes and respiratory illnesses. There is a critical need for clinical trials to evaluate potential therapeutic candidates, providing evidence to guide treatment decisions for post-COVID-19 syndromes.
Collapse
Affiliation(s)
- Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Cameron R. Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| | - Daniel Ang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Michelle AW Lee
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Tom Van Agtmael
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| |
Collapse
|
8
|
Lechner I, Reindl M, von der Emde S, Desheva A, Oberhollenzer F, Tiller C, Holzknecht M, Kremser T, Faccini J, Gollmann-Tepeköylü C, Kremser C, Mayr A, Bauer A, Metzler B, Reinstadler SJ. Hepatic Tissue Alterations in ST-Elevation Myocardial Infarction: Determinants and Prognostic Implications. Circ Cardiovasc Imaging 2024; 17:e017041. [PMID: 39611252 DOI: 10.1161/circimaging.124.017041] [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: 05/01/2024] [Accepted: 09/19/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The presence and clinical significance of hepatic tissue alterations as assessed by cardiac magnetic resonance imaging in patients with ST-segment-elevation myocardial infarction (STEMI), are unclear. This study aimed to investigate associations of hepatic T1 patterns with myocardial tissue damage and clinical outcomes in patients suffering from STEMI. METHODS We analyzed 485 patients with STEMI treated with percutaneous coronary intervention who were enrolled in the prospective MARINA STEMI study (Magnetic Resonance Imaging In Acute ST-Elevation Myocardial Infarction). Myocardial function and left and right ventricular (RV) infarct characteristics were assessed by cardiac magnetic resonance within the first week after STEMI. Native hepatic T1 times and extracellular volume were evaluated from standard cardiac T1 maps at baseline and 4 months thereafter. RESULTS Median hepatic T1 times were 559 ms (interquartile range, 514-605) at baseline and decreased to 542 ms (interquartile range, 507-577) at 4 months (P<0.001). Hepatic T1 times at baseline were independently associated with female sex (β 0.116; P=0.008), hyperlipidemia (β -0.116; P=0.008), and myocardial tissue damage (infarct size: β 0.178; P<0.001; microvascular obstruction: β 0.193; P<0.001; RV infarction: β 0.161; P<0.001). Determinants of hepatic T1 times at 4 months were female sex (β 0.123; P=0.002), multivessel disease (β 0.121; P=0.002), N-terminal pro-B-type natriuretic peptide (β 0.101; P=0.010), RV infarction (β 0.501; P<0.001), and RV end-systolic volume index (β 0.087; P=0.031). Patients without a decrease exhibited a higher frequency of major adverse cardiovascular events (13% versus 5%; P=0.003). Hepatic T1 times at baseline (hazard ratio, 1.87 [95% CI, 1.40-2.50]; P<0.001), 4 months (hazard ratio, 2.69 [95% CI, 2.15-3.36]; P<0.001), and hepatic extracellular volume at 4 months (hazard ratio, 1.59 [95% CI, 1.33-1.90]; P<0.001) were associated with major adverse cardiovascular events. After adjustment for univariable associates, only hepatic T1 times at 4 months were independently associated with adverse outcomes (hazard ratio, 2.86 [95% CI, 1.99-4.12]; P<0.001). CONCLUSIONS Hepatic tissue alterations determined by T1 mapping were associated with female sex, hyperlipidemia, multivessel disease, N-terminal pro-B-type natriuretic peptide, and left and RV myocardial tissue damage. These alterations can persist into the chronic phase after STEMI and indicate a worse clinical outcome. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04113356.
Collapse
Affiliation(s)
- Ivan Lechner
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Martin Reindl
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Sebastian von der Emde
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Alina Desheva
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Fritz Oberhollenzer
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Christina Tiller
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Magdalena Holzknecht
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Thomas Kremser
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Julian Faccini
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Can Gollmann-Tepeköylü
- University Clinic of Cardiac Surgery (C.G.-T.), Medical University of Innsbruck Innsbruck, Austria
| | - Christian Kremser
- University Clinic of Radiology (C.K., A.M.), Medical University of Innsbruck Innsbruck, Austria
| | - Agnes Mayr
- University Clinic of Radiology (C.K., A.M.), Medical University of Innsbruck Innsbruck, Austria
| | - Axel Bauer
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Bernhard Metzler
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| | - Sebastian J Reinstadler
- University Clinic of Internal Medicine III, Cardiology and Angiology (I.L., M.R., S.v.d.E., A.D., F.O., C.T., M.H., T.K., J.F., A.B., B.M., S.J.R.), Medical University of Innsbruck Innsbruck, Austria
| |
Collapse
|
9
|
Nagel E, Pappas L. The Rise of T2. JACC Cardiovasc Imaging 2024; 17:622-624. [PMID: 38727645 DOI: 10.1016/j.jcmg.2024.03.013] [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] [Received: 01/29/2024] [Accepted: 03/14/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Eike Nagel
- Institute for Experimental and Translational Cardiovascular Imaging, Goethe University Frankfurt, Frankfurt, Germany; German Center for Cardiovascular Research (DZHK), Rhine-Main, Germany.
| | - Laura Pappas
- Institute for Experimental and Translational Cardiovascular Imaging, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
10
|
Chen L, Zhang Z, Du X, Liu J, Liu Z, Chen W, Che W. Establishment and validation of an extracellular volume model without blood sampling in ST-segment elevation myocardial infarction patients. EUROPEAN HEART JOURNAL. IMAGING METHODS AND PRACTICE 2024; 2:qyae053. [PMID: 39224096 PMCID: PMC11367959 DOI: 10.1093/ehjimp/qyae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/22/2024] [Indexed: 09/04/2024]
Abstract
AIMS Recent studies have shown that extracellular volume (ECV) can also be obtained without blood sampling by the linear relationship between haematocrit (HCT) and blood pool R1 (1/T1). However, whether this relationship holds for patients with myocardial infarction is still unclear. This study established and validated an ECV model without blood sampling in ST-segment elevation myocardial infarction (STEMI) patients. METHODS AND RESULTS A total of 398 STEMI patients who underwent cardiac magnetic resonance (CMR) examination with T1 mapping and venous HCT within 24 h were retrospectively analysed. All patients were randomly divided into a derivation group and a validation group. The mean CMR scan time was 3 days after primary percutaneous coronary intervention. In the derivation group, a synthetic HCT formula was obtained by the linear regression between HCT and blood pool R1 (R 2 = 0.45, P < 0.001). The formula was used in the validation group; the results showed high concordance and correlation between synthetic ECV and conventional ECV in integral (bias = -0.12; R 2 = 0.92, P < 0.001), myocardial infarction site (bias = -0.23; R 2 = 0.93, P < 0.001), and non-myocardial infarction sites (bias = -0.09; R 2 = 0.94, P < 0.001). CONCLUSION In STEMI patients, synthetic ECV without blood sampling had good consistency and correlation with conventional ECV. This study might provide a convenient and accurate method to obtain the ECV from CMR to identify myocardial fibrosis.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai 200072, China
| | - Zeqing Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou 221002, China
| | - Xinjia Du
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou 221002, China
| | - Jiahua Liu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou 221002, China
| | - Zhongxiao Liu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou 221002, China
| | - Wensu Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Xuzhou 221002, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai 200072, China
| |
Collapse
|
11
|
Naaktgeboren WR, Stuiver MM, van Harten WH, Aaronson NK, Scott JM, Sonke G, van der Wall E, Velthuis M, Leiner T, Teske AJ, May AM, Groen WG. Effects of exercise during chemotherapy for breast cancer on long-term cardiovascular toxicity. Open Heart 2023; 10:e002464. [PMID: 37903570 PMCID: PMC10619040 DOI: 10.1136/openhrt-2023-002464] [Citation(s) in RCA: 2] [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] [Received: 08/17/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Animal data suggest that exercise during chemotherapy is cardioprotective, but clinical evidence to support this is limited. This study evaluated the effect of exercise during chemotherapy for breast cancer on long-term cardiovascular toxicity. METHODS This is a follow-up study of two previously performed randomised trials in patients with breast cancer allocated to exercise during chemotherapy or non-exercise controls. Cardiac imaging parameters, including T1 mapping (native T1, extracellular volume fraction (ECV)), left ventricular ejection fraction (LVEF) and global longitudinal strain (GLS), cardiorespiratory fitness, and physical activity levels, were acquired 8.5 years post-treatment. RESULTS In total, 185 breast cancer survivors were included (mean age 58.9±7.8 years), of whom 99% and 18% were treated with anthracyclines and trastuzumab, respectively. ECV and Native T1 were 25.3%±2.5% and 1026±51 ms in the control group, and 24.6%±2.8% and 1007±44 ms in the exercise group, respectively. LVEF was borderline normal in both groups, with an LVEF<50% prevalence of 22.5% (n=40/178) in all participants. Compared with control, native T1 was statistically significantly lower in the exercise group (β=-20.16, 95% CI -35.35 to -4.97). We found no effect of exercise on ECV (β=-0.69, 95% CI -1.62 to 0.25), LVEF (β=-1.36, 95% CI -3.45 to 0.73) or GLS (β=0.31, 95% CI -0.76 to 1.37). Higher self-reported physical activity levels during chemotherapy were significantly associated with better native T1 and ECV. CONCLUSIONS In long-term breast cancer survivors, exercise and being more physically active during chemotherapy were associated with better structural but not functional cardiac parameters. The high prevalence of cardiac dysfunction calls for additional research on cardioprotective measures, including alternative exercise regimens. TRIAL REGISTRATION NUMBER NTR7247.
Collapse
Affiliation(s)
- Willeke R Naaktgeboren
- Psychosocial Research and Epidemiology, Antoni van Leeuwenhoek Netherlands Cancer Institute, Amsterdam, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Martijn M Stuiver
- Psychosocial Research and Epidemiology, Antoni van Leeuwenhoek Netherlands Cancer Institute, Amsterdam, The Netherlands
- Center for Quality of Life, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Faculty of Health, Amsterdam University of Applied Sciences, Amsterdam, The Netherlands
| | - Wim H van Harten
- Psychosocial Research and Epidemiology, Antoni van Leeuwenhoek Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Health Technology and Services Research, University of Twente, Enschede, Netherlands
- Rijnstate Hospital, Arnhem, Netherlands
| | - Neil K Aaronson
- Psychosocial Research and Epidemiology, Antoni van Leeuwenhoek Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jessica M Scott
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Gabe Sonke
- Medical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Elsken van der Wall
- Division of Internal Medicine and Dermatology, University Medical Centre, Utrecht, The Netherlands
| | - Miranda Velthuis
- Netherlands Comprehensive Cancer Organisation, Nijmegen, The Netherlands
| | - Tim Leiner
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Radiology, UMC Utrecht, Utrecht, Netherlands
| | - Arco J Teske
- Cardiology, University Medical Centre, Utrecht, The Netherlands
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Wim G Groen
- Department of Medicine for Older People, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Aging & Later Life, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
- Amsterdam Movement Sciences, Ageing & Vitality, Rehabilitation & Development, Amstermdam, Netherlands
| |
Collapse
|
12
|
Weir-McCall JR, Alabed S. Myocardial Tissue Characterization With CT-Derived Extracellular Volume: Closing the Gap With CMR? JACC Cardiovasc Imaging 2023; 16:1318-1320. [PMID: 37354154 DOI: 10.1016/j.jcmg.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/10/2023] [Indexed: 06/26/2023]
Affiliation(s)
- Jonathan R Weir-McCall
- Department of Radiology, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Radiology, Royal Papworth Hospital, Cambridge, United Kingdom.
| | - Samer Alabed
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom; INSIGNEO, Institute for In Silico Medicine, University of Sheffield, United Kingdom; NIHR Sheffield Biomedical Research Centre, Sheffield, United Kingdom
| |
Collapse
|
13
|
Han D, Lin A, Kuronuma K, Gransar H, Dey D, Friedman JD, Berman DS, Tamarappoo BK. Cardiac Computed Tomography for Quantification of Myocardial Extracellular Volume Fraction: A Systematic Review and Meta-Analysis. JACC Cardiovasc Imaging 2023; 16:1306-1317. [PMID: 37269267 DOI: 10.1016/j.jcmg.2023.03.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND Extracellular volume (ECV) is a quantitative measure of extracellular compartment expansion, and an increase in ECV is a marker of myocardial fibrosis. Although cardiac magnetic resonance (CMR) is considered the standard imaging tool for ECV quantification, cardiac computed tomography (CT) has also been used for ECV assessment. OBJECTIVES The aim of this meta-analysis was to evaluate the correlation and agreement in the quantification of myocardial ECV by CT and CMR. METHODS PubMed and Web of Science were searched for relevant publications reporting on the use of CT for ECV quantification compared with CMR as the reference standard. The authors employed a meta-analysis using the restricted maximum-likelihood estimator with a random-effects method to estimate summary correlation and mean difference. A subgroup analysis was performed to compare the correlation and mean differences between single-energy CT (SECT) and dual-energy CT (DECT) techniques for the ECV quantification. RESULTS Of 435 papers, 13 studies comprising 383 patients were identified. The mean age range was 57.3 to 82 years, and 65% of patients were male. Overall, there was an excellent correlation between CT-derived ECV and CMR-derived ECV (mean: 0.90 [95% CI: 0.86-0.95]). The pooled mean difference between CT and CMR was 0.96% (95% CI: 0.14%-1.78%). Seven studies reported correlation values using SECT, and 4 studies reported those using DECT. The pooled correlation from studies utilizing DECT for ECV quantification was significantly higher compared with those with SECT (mean: 0.94 [95% CI: 0.91-0.98] vs 0.87 [95% CI: 0.80-0.94], respectively; P = 0.01). There was no significant difference in pooled mean differences between SECT vs DECT (P = 0.85). CONCLUSIONS CT-derived ECV showed an excellent correlation and mean difference of <1% with CMR-derived ECV. However, the overall quality of the included studies was low, and larger, prospective studies are needed to examine the accuracy and diagnostic and prognostic utility of CT-derived ECV.
Collapse
Affiliation(s)
- Donghee Han
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Andrew Lin
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Keiichiro Kuronuma
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Heidi Gransar
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - John D Friedman
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniel S Berman
- Department of Imaging and Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Balaji K Tamarappoo
- Cardiovascular Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
14
|
Zsarnóczay E, Varga-Szemes A, Emrich T, Szilveszter B, van der Werf NR, Mastrodicasa D, Maurovich-Horvat P, Willemink MJ. Characterizing the Heart and the Myocardium With Photon-Counting CT. Invest Radiol 2023; 58:505-514. [PMID: 36822653 DOI: 10.1097/rli.0000000000000956] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
ABSTRACT Noninvasive cardiac imaging has rapidly evolved during the last decade owing to improvements in computed tomography (CT)-based technologies, among which we highlight the recent introduction of the first clinical photon-counting detector CT (PCD-CT) system. Multiple advantages of PCD-CT have been demonstrated, including increased spatial resolution, decreased electronic noise, and reduced radiation exposure, which may further improve diagnostics and may potentially impact existing management pathways. The benefits that can be obtained from the initial experiences with PCD-CT are promising. The implementation of this technology in cardiovascular imaging allows for the quantification of coronary calcium, myocardial extracellular volume, myocardial radiomics features, epicardial and pericoronary adipose tissue, and the qualitative assessment of coronary plaques and stents. This review aims to discuss these major applications of PCD-CT with a focus on cardiac and myocardial characterization.
Collapse
Affiliation(s)
| | - Akos Varga-Szemes
- From the Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston
| | | | | | | | | | | | | |
Collapse
|
15
|
Segre CAW, de Lemos JA, Assunção Junior AN, Nomura CH, Favarato D, Strunz CMC, Villa AV, Parga Filho JR, Rezende PC, Hueb W, Ramires JAF, Kalil Filho R, Serrano Junior CV. Chronic troponin elevation assessed by myocardial T1 mapping in patients with stable coronary artery disease. Medicine (Baltimore) 2023; 102:e33548. [PMID: 37083772 PMCID: PMC10118361 DOI: 10.1097/md.0000000000033548] [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: 12/14/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Cardiac troponin detected with sensitive assays can be chronically elevated, in the absence of unstable coronary syndromes. In patients with chronic coronary artery disease, clinically silent ischemic episodes may cause chronic troponin release. T1 mapping is a cardiovascular magnetic resonance technique useful in quantitative cardiac tissue characterization. We selected patients with anatomically and functionally normal hearts to investigate associations between chronic troponin release and myocardial tissue characteristics assessed by T1 mapping. METHODS We investigated the relationship between cardiac troponin I concentrations and cardiovascular magnetic resonance T1 mapping parameters in patients with stable coronary artery disease enrolled in MASS V study before elective revascularization. Participants had no previous myocardial infarction, negative late gadolinium enhancement, normal left ventricular function, chamber dimensions and wall thickness. RESULTS A total of 56 patients were analyzed in troponin tertiles: nativeT1 and extracellular volume (ECV) values (expressed as means ± standard deviations) increased across tertiles: nativeT1 (1006 ± 27 ms vs 1016 ± 27 ms vs 1034 ± 37 ms, ptrend = 0.006) and ECV (22 ± 3% vs 23 ± 1.9% vs 25 ± 3%, ptrend = 0.007). Cardiac troponin I concentrations correlated with native T1(R = 0.33, P = .012) and ECV (R = 0.3, P = .025), and were independently associated with nativeT1 (P = .049) and ventricular mass index (P = .041) in multivariable analysis. CONCLUSION In patients with chronic coronary artery disease and structurally normal hearts, troponin I concentrations correlated with T1 mapping parameters, suggesting that diffuse edema or fibrosis scattered in normal myocardium might be associated with chronic troponin release.
Collapse
Affiliation(s)
| | - James A. de Lemos
- Division of Cardiology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Cesar Higa Nomura
- Heart Institute (InCor) University of São Paulo Clinics Hospital, Sao Paulo, Brazil
| | - Desiderio Favarato
- Heart Institute (InCor) University of São Paulo Clinics Hospital, Sao Paulo, Brazil
| | | | | | | | - Paulo Cury Rezende
- Heart Institute (InCor) University of São Paulo Clinics Hospital, Sao Paulo, Brazil
| | - Whady Hueb
- Heart Institute (InCor) University of São Paulo Clinics Hospital, Sao Paulo, Brazil
| | | | - Roberto Kalil Filho
- Heart Institute (InCor) University of São Paulo Clinics Hospital, Sao Paulo, Brazil
| | | |
Collapse
|
16
|
Chen BH, An DA, Wu CW, Yue T, Bautista M, Ouchi E, Xu JR, Hu J, Zhou Y, Pu J, Wu LM. Prognostic significance of non-infarcted myocardium correlated with microvascular impairment evaluated dynamically by native T1 mapping. Insights Imaging 2023; 14:50. [PMID: 36941401 PMCID: PMC10027971 DOI: 10.1186/s13244-022-01360-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/19/2022] [Indexed: 03/22/2023] Open
Abstract
OBJECTIVES This study aimed to investigate the influence of microvascular impairment on myocardial characteristic alterations in remote myocardium at multiple time points, and its prognostic significance after acute ST-segment elevation myocardial infarction (STEMI). METHODS Patients were enrolled prospectively and performed CMR at baseline, 30 days, and 6 months. The primary endpoint was major adverse cardiac events (MACE): death, myocardial reinfarction, malignant arrhythmia, and hospitalization for heart failure. Cox proportional hazards regression modeling was analyzed to estimate the correlation between T1 mapping of remote myocardium and MACE in patients with and without microvascular obstruction (MVO). RESULTS A total of 135 patients (mean age 60.72 years; 12.70% female, median follow-up 510 days) were included, of whom 86 (63.70%) had MVO and 26 (19.26%) with MACE occurred in patients. Native T1 values of remote myocardium changed dynamically. At 1 week and 30 days, T1 values of remote myocardium in the group with MVO were higher than those without MVO (p = 0.030 and p = 0.001, respectively). In multivariable cox regression analysis of 135 patients, native1w T1 (HR 1.03, 95%CI 1.01-1.04, p = 0.002), native30D T1 (HR 1.05, 95%CI 1.03-1.07, p < 0.001) and LGE (HR 1.10, 95%CI 1.05-1.15, p < 0.001) were joint independent predictors of MACE. In multivariable cox regression analysis of 86 patients with MVO, native30D T1 (HR 1.05, 95%CI 1.04-1.07, p < 0.001) and LGE (HR 1.10, 95%CI 1.05-1.15, p < 0.001) were joint independent predictors of MACE. CONCLUSIONS The evolution of native T1 in remote myocardium was associated with the extent of microvascular impairment after reperfusion injury. In patients with MVO, native30D T1 and LGE were joint independent predictors of MACE.
Collapse
Affiliation(s)
- Bing-Hua Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China
| | - Dong-Aolei An
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China
| | - Chong-Wen Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China
| | - Ting Yue
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China
| | - Matthew Bautista
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Erika Ouchi
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Jian-Rong Xu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI, 48201, USA
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China.
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China.
| | - Lian-Ming Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.160 PuJian Road, Shanghai, 200127, P. R. China.
| |
Collapse
|
17
|
Cerne JW, Shehata C, Ragin A, Pathrose A, Veer M, Subedi K, Allen BD, Avery RJ, Markl M, Carr JC. Potential Prognostic Value of Native T1 in Pulmonary Hypertension Patients. Life (Basel) 2023; 13:775. [PMID: 36983931 PMCID: PMC10051677 DOI: 10.3390/life13030775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Native T1, extracellular volume fraction (ECV), and late gadolinium enhancement (LGE) characterize myocardial tissue and relate to patient prognosis in a variety of diseases, including pulmonary hypertension. The purpose of this study was to evaluate if left ventricle (LV) fibrosis measurements have prognostic value for cardiac outcomes in pulmonary hypertension subgroups. 54 patients with suspected pulmonary hypertension underwent right-heart catheterization and were classified into pulmonary hypertension subgroups: pre-capillary component (PreCompPH) and isolated post-capillary (IpcPH). Cardiac magnetic resonance imaging (MRI) scans were performed with the acquisition of balanced cine steady-state free precession, native T1, and LGE pulse sequences to measure cardiac volumes and myocardial fibrosis. Associations between cardiac events and cardiac MRI measurements were analyzed within PreCompPH and IpcPH patients. IpcPH: LV native T1 was higher in patients who experienced a cardiac event within two years vs. those who did not. In patients with LV native T1 > 1050 ms, the rate of cardiac events was higher. ECV and quantitative LGE did not differ between groups. PreCompPH: native T1, ECV, and quantitative/qualitative LGE did not differ between patients who experienced a cardiac event within two years vs. those who did not. LV native T1 may have potential value for forecasting cardiac events in IpcPH, but not in PreCompPH, patients.
Collapse
Affiliation(s)
- John W. Cerne
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Christina Shehata
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Ann Ragin
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Ashitha Pathrose
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Manik Veer
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Kamal Subedi
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Bradley D. Allen
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Ryan J. Avery
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| | - Michael Markl
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - James C. Carr
- Department of Radiology, Feinberg School of Medicine, Northwestern, Chicago, IL 60611, USA
| |
Collapse
|
18
|
Hochhegger B, Pasini R, Roncally Carvalho A, Rodrigues R, Altmayer S, Kayat Bittencourt L, Marchiori E, Forghani R. Artificial Intelligence for Cardiothoracic Imaging: Overview of Current and Emerging Applications. Semin Roentgenol 2023; 58:184-195. [PMID: 37087139 DOI: 10.1053/j.ro.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 03/07/2023]
Abstract
Artificial intelligence algorithms can learn by assimilating information from large datasets in order to decipher complex associations, identify previously undiscovered pathophysiological states, and construct prediction models. There has been tremendous interest and increased incorporation of artificial intelligence into various industries, including healthcare. As a result, there has been an exponential rise in the number of research articles and industry participants producing models intended for a variety of applications in medical imaging, which can be challenging to navigate for radiologists. In thoracic imaging, multiple applications are being evaluated for chest radiography and computed tomography and include applications for lung nodule evaluation and cancer imaging, quantifying diffuse lung disorders, and cardiac imaging, to name a few. This review aims to provide an overview of current clinical AI models, focusing on the most common clinical applications of AI in cardiothoracic imaging.
Collapse
|
19
|
Screever EM, Gorter TM, Willems TP, Aboumsallem JP, Suthahar N, Mahmoud B, van Veldhuisen DJ, de Boer RA, Meijers WC. Diffuse Myocardial Fibrosis on Cardiac Magnetic Resonance Imaging Is Related to Galectin-3 and Predicts Outcome in Heart Failure. Biomolecules 2023; 13:410. [PMID: 36979345 PMCID: PMC10046101 DOI: 10.3390/biom13030410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
AIMS Ongoing adverse remodeling is a hallmark of heart failure (HF), which might be reflected by either focal or diffuse myocardial fibrosis. Therefore, in (pre)clinical settings, we used immunohistochemistry or cardiac magnetic resonance imaging (CMR) to investigate the association of (focal or diffuse) fibrosis with cardiac biomarkers and adverse events in HF. METHODS AND RESULTS In C57Bl/6J mice, we determined the presence and extent of myocardial fibrosis 6 weeks post-myocardial infarction (MI). Furthermore, we studied 159 outpatient HF patients who underwent CMR, and determined focal and diffuse fibrosis by late gadolinium enhancement (LGE) and post-contrast T1 time of the non-LGE myocardium, respectively. HF patients were categorized based on the presence of LGE, and by the median post-contrast T1 time. Kaplan-Meier and Cox regression analyses were used to determine the association of fibrosis with HF hospitalization and all-cause mortality. LGE was detected in 61 (38%) patients. Cardiac biomarker levels were comparable between LGE-positive and LGE-negative patients. LGE-positive patients with a short T1 time had elevated levels of both NT-proBNP and galectin-3 (1611 vs. 453 ng/L, p = 0.026 and 20 vs. 15 μg/L, p = 0.004, respectively). This was not observed in LGE-negative patients. Furthermore, a short T1 time in LGE-positive patients was associated with a higher risk of adverse events (log-rank p = 0.01). CONCLUSION This study implies that cardiac biomarkers reflect active remodeling of the non-infarcted myocardium of patients with focal myocardial scarring. Diffuse fibrosis, in contrast to focal scarring, might have a higher prognostic value regarding adverse outcomes in HF patients.
Collapse
Affiliation(s)
- Elles M. Screever
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Thomas M. Gorter
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Tineke P. Willems
- Department of Radiology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Navin Suthahar
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Belend Mahmoud
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Dirk J. van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Wouter C. Meijers
- Department of Cardiology, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
20
|
Treiber J, Hausmann CS, Wolter JS, Fischer-Rasokat U, Kriechbaum SD, Hamm CW, Nagel E, Puntmann VO, Rolf A. Native T1 is predictive of cardiovascular death/heart failure events and all-cause mortality irrespective of the patient's volume status. Front Cardiovasc Med 2023; 10:1091334. [PMID: 36865890 PMCID: PMC9971619 DOI: 10.3389/fcvm.2023.1091334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
Background Native T1 has become a pivotal parameter of tissue composition that is assessed by cardiac magnetic resonance (CMR). It characterizes diseased myocardium and can be used for prognosis estimation. Recent publications have shown that native T1 is influenced by short-term fluctuations of volume status due to hydration or hemodialysis. Methods Patients from a prospective BioCVI all-comers clinical CMR registry were included, and native T1 and plasma volume status (PVS) were determined according to Hakim's formula as surrogate markers of patient volume status. The primary endpoint was defined as combined endpoint of cardiovascular death or hospitalization for heart failure events, the secondary endpoint was defined as all-cause mortality. Results A total of 2,047 patients were included since April 2017 [median (IQR); age 63 (52-72) years, 33% female]. There was a significant although weak influence of PVS on native T1 (β = 0.11, p < 0.0001). Patients with volume expansion (PVS > -13%) showed significantly higher values for tissue markers than non-volume-overloaded patients [PVS ≤ -13%; median (IQR); native T1 1,130 (1,095-1,170) vs. 1,123 (1,086-1,166) ms, p < 0.003; and T2 39 (37-40) vs. 38 (36-40) ms, p < 0.0001]. In Cox regression analysis both native T1 and PVS were independently predictive of the primary endpoint and all-cause mortality. Conclusion Despite a weak effect of PVS on native T1, its predictive power was not affected in a large, all-comers cohort.
Collapse
Affiliation(s)
- Julia Treiber
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Carla S. Hausmann
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Jan Sebastian Wolter
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Ulrich Fischer-Rasokat
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Steffen D. Kriechbaum
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Christian W. Hamm
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany,Justus Liebig University of Giessen, Giessen, Germany
| | - Eike Nagel
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany,Institute for Experimental and Translational Cardiovascular Imaging, University Hospital Frankfurt am Main, Frankfurt, Germany
| | - Valentina O. Puntmann
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany,Institute for Experimental and Translational Cardiovascular Imaging, University Hospital Frankfurt am Main, Frankfurt, Germany
| | - Andreas Rolf
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Bad Nauheim, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany,Justus Liebig University of Giessen, Giessen, Germany,*Correspondence: Andreas Rolf, ✉
| |
Collapse
|
21
|
Lanzafame LRM, Bucolo GM, Muscogiuri G, Sironi S, Gaeta M, Ascenti G, Booz C, Vogl TJ, Blandino A, Mazziotti S, D’Angelo T. Artificial Intelligence in Cardiovascular CT and MR Imaging. Life (Basel) 2023; 13:507. [PMID: 36836864 PMCID: PMC9968221 DOI: 10.3390/life13020507] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The technological development of Artificial Intelligence (AI) has grown rapidly in recent years. The applications of AI to cardiovascular imaging are various and could improve the radiologists' workflow, speeding up acquisition and post-processing time, increasing image quality and diagnostic accuracy. Several studies have already proved AI applications in Coronary Computed Tomography Angiography and Cardiac Magnetic Resonance, including automatic evaluation of calcium score, quantification of coronary stenosis and plaque analysis, or the automatic quantification of heart volumes and myocardial tissue characterization. The aim of this review is to summarize the latest advances in the field of AI applied to cardiovascular CT and MR imaging.
Collapse
Affiliation(s)
- Ludovica R. M. Lanzafame
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Giuseppe M. Bucolo
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Giuseppe Muscogiuri
- Department of Radiology, Istituto Auxologico Italiano IRCCS, San Luca Hospital, 20149 Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, 20854 Milan, Italy
| | - Sandro Sironi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20854 Milan, Italy
- Department of Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Michele Gaeta
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Giorgio Ascenti
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Christian Booz
- Division of Experimental Imaging, Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Thomas J. Vogl
- Division of Experimental Imaging, Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Alfredo Blandino
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Silvio Mazziotti
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
| | - Tommaso D’Angelo
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital Messina, 98124 Messina, Italy
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 Rotterdam, The Netherlands
| |
Collapse
|
22
|
Treiber J, Novak D, Fischer-Rasokat U, Wolter JS, Kriechbaum S, Weferling M, von Jeinsen B, Hain A, Rieth AJ, Siemons T, Keller T, Hamm CW, Rolf A. Regional extracellular volume within late gadolinium enhancement-positive myocardium to differentiate cardiac sarcoidosis from myocarditis of other etiology: a cardiovascular magnetic resonance study. J Cardiovasc Magn Reson 2023; 25:8. [PMID: 36755275 PMCID: PMC9909902 DOI: 10.1186/s12968-023-00918-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 01/12/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Cardiovascular magnetic resonance (CMR) plays a pivotal role in diagnosing myocardial inflammation. In addition to late gadolinium enhancement (LGE), native T1 and T2 mapping as well as extracellular volume (ECV) are essential tools for tissue characterization. However, the differentiation of cardiac sarcoidosis (CS) from myocarditis of other etiology can be challenging. Positron-emission tomography-computed tomography (PET-CT) regularly shows the highest Fluordesoxyglucose (FDG) uptake in LGE positive regions. It was therefore the aim of this study to investigate, whether native T1, T2, and ECV measurements within LGE regions can improve the differentiation of CS and myocarditis compared with using global native T1, T2, and ECV values alone. METHODS PET/CT confirmed CS patients and myocarditis patients (both acute and chronic) from a prospective registry were compared with respect to regional native T1, T2, and ECV. Acute and chronic myocarditis were defined based on the 2013 European Society of Cardiology position paper on myocarditis. All parametric measures and ECV were acquired in standard fashion on three short-axis slices according to the ConSept study for global values and within PET-CT positive regions of LGE. RESULTS Between 2017 and 2020, 33 patients with CS and 73 chronic and 35 acute myocarditis patients were identified. The mean ECV (± SD) in LGE regions of CS patients was higher than in myocarditis patients (CS vs. acute and chronic, respectively: 0.65 ± 0.12 vs. 0.45 ± 0.13 and 0.47 ± 0.1; p < 0.001). Acute and chronic myocarditis patients had higher global native T1 values (1157 ± 54 ms vs. 1196 ± 63 ms vs. 1215 ± 74 ms; p = 0.001). There was no difference in global T2 and ECV values between CS and acute or chronic myocarditis patients. CONCLUSION This is the first study to show that the calculation of regional ECV within LGE-positive regions may help to differentiate CS from myocarditis. Further studies are warranted to corroborate these findings.
Collapse
Affiliation(s)
- Julia Treiber
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Dijana Novak
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
| | - Ulrich Fischer-Rasokat
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Jan Sebastian Wolter
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Steffen Kriechbaum
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Maren Weferling
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Beatrice von Jeinsen
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Andreas Hain
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
| | - Andreas J Rieth
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Tamo Siemons
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
| | - Till Keller
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- Medical Clinic 1, Justus-Liebig-Universität Giessen, Giessen, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Christian W Hamm
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany
- Medical Clinic 1, Justus-Liebig-Universität Giessen, Giessen, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany
| | - Andreas Rolf
- Department of Cardiology, Kerckhoff Heart and Thorax Center, Benekestrasse 2-8, 61231, Bad Nauheim, Germany.
- Medical Clinic 1, Justus-Liebig-Universität Giessen, Giessen, Germany.
- German Center for Cardiovascular Research (DZHK), Rhine-Main Partner Site, Frankfurt am Main, Germany.
| |
Collapse
|
23
|
Bazmpani MA, Nikolaidou C, Papanastasiou CA, Ziakas A, Karamitsos TD. Cardiovascular Magnetic Resonance Parametric Mapping Techniques for the Assessment of Chronic Coronary Syndromes. J Cardiovasc Dev Dis 2022; 9:jcdd9120443. [PMID: 36547440 PMCID: PMC9782163 DOI: 10.3390/jcdd9120443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The term chronic coronary syndromes encompasses a variety of clinical presentations of coronary artery disease (CAD), ranging from stable angina due to epicardial coronary artery disease to microvascular coronary dysfunction. Cardiac magnetic resonance (CMR) imaging has an established role in the diagnosis, prognostication and treatment planning of patients with CAD. Recent advances in parametric mapping CMR techniques have added value in the assessment of patients with chronic coronary syndromes, even without the need for gadolinium contrast administration. Furthermore, quantitative perfusion CMR techniques have enabled the non-invasive assessment of myocardial blood flow and myocardial perfusion reserve and can reliably identify multivessel coronary artery disease and microvascular dysfunction. This review summarizes the clinical applications and the prognostic value of the novel CMR parametric mapping techniques in the setting of chronic coronary syndromes and discusses their strengths, pitfalls and future directions.
Collapse
Affiliation(s)
- Maria Anna Bazmpani
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | | | - Christos A. Papanastasiou
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Antonios Ziakas
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros D. Karamitsos
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-2310994832; Fax: +30-2310994673
| |
Collapse
|
24
|
Rosa SA. Prognosis after Myocardial Infarction - A Deep Look into Myocardial Tissue. Arq Bras Cardiol 2022; 119:958-959. [PMID: 36541990 DOI: 10.36660/abc.20220798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
25
|
Zaman A, Zhao S, Kron J, Abbate A, Tomdio A, Hundley WG, Jordan JH. Role of Cardiac MRI Imaging of Focal and Diffuse Inflammation and Fibrosis in Cardiomyopathy Patients Who Have Pacemakers/ICD Devices. Curr Cardiol Rep 2022; 24:1529-1536. [PMID: 35984554 PMCID: PMC10123953 DOI: 10.1007/s11886-022-01770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW This focused report aims to discuss and summarize the use of conventional and emerging methods using cardiovascular magnetic resonance (CMR) imaging in cardiomyopathy patients with implanted cardiac devices to identify diffuse and focal inflammation and fibrosis. RECENT FINDINGS Many cardiomyopathy patients with diffuse and focal myocardial fibrosis have a unique need for cardiac imaging that is complicated by cardiovascular implantable electronic devices (CIEDs). CMR imaging can accurately image myocardial fibrosis and inflammation using T1 mapping and late gadolinium enhancement (LGE) imaging. CMR imaging in CIED patients, however, has been limited due to severe imaging artifacts associated with the devices. The emergence of wideband imaging variants of LGE and T1 mapping techniques can successfully reduce or eliminate CIED artifacts for the evaluation of myocardial substrate in cardiomyopathy patients. Wideband imaging variants of LGE and T1 mapping techniques provide new tools for imaging focal and diffuse fibrosis and imaging in cardiomyopathy patients with implanted cardiac devices. These emerging techniques have the potential for great impact in clinical care of such patients as well as clinical research where imaging endpoints are desired.
Collapse
Affiliation(s)
- Ananna Zaman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Samantha Zhao
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Jordana Kron
- Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, West Hospital, 8th Floor, 1200 E. Broad Street, Richmond, VA, 23298, USA
| | - Antonio Abbate
- Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, West Hospital, 8th Floor, 1200 E. Broad Street, Richmond, VA, 23298, USA
| | - Anna Tomdio
- Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, West Hospital, 8th Floor, 1200 E. Broad Street, Richmond, VA, 23298, USA
| | - W Gregory Hundley
- Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, West Hospital, 8th Floor, 1200 E. Broad Street, Richmond, VA, 23298, USA
| | - Jennifer H Jordan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA. .,Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, West Hospital, 8th Floor, 1200 E. Broad Street, Richmond, VA, 23298, USA.
| |
Collapse
|
26
|
Muscogiuri G, Volpato V, Cau R, Chiesa M, Saba L, Guglielmo M, Senatieri A, Chierchia G, Pontone G, Dell’Aversana S, Schoepf UJ, Andrews MG, Basile P, Guaricci AI, Marra P, Muraru D, Badano LP, Sironi S. Application of AI in cardiovascular multimodality imaging. Heliyon 2022; 8:e10872. [PMID: 36267381 PMCID: PMC9576885 DOI: 10.1016/j.heliyon.2022.e10872] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/23/2022] [Accepted: 09/27/2022] [Indexed: 12/16/2022] Open
Abstract
Technical advances in artificial intelligence (AI) in cardiac imaging are rapidly improving the reproducibility of this approach and the possibility to reduce time necessary to generate a report. In cardiac computed tomography angiography (CCTA) the main application of AI in clinical practice is focused on detection of stenosis, characterization of coronary plaques, and detection of myocardial ischemia. In cardiac magnetic resonance (CMR) the application of AI is focused on post-processing and particularly on the segmentation of cardiac chambers during late gadolinium enhancement. In echocardiography, the application of AI is focused on segmentation of cardiac chambers and is helpful for valvular function and wall motion abnormalities. The common thread represented by all of these techniques aims to shorten the time of interpretation without loss of information compared to the standard approach. In this review we provide an overview of AI applications in multimodality cardiac imaging.
Collapse
Affiliation(s)
- Giuseppe Muscogiuri
- Department of Radiology, Istituto Auxologico Italiano IRCCS, San Luca Hospital, Italy,School of Medicine, University of Milano-Bicocca, Milan, Italy,Corresponding author.
| | - Valentina Volpato
- Department of Cardiac, Neurological and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano IRCCS, Milan, Italy,IRCCS Ospedale Galeazzi - Sant'Ambrogio, University Cardiology Department, Milan, Italy
| | - Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari, Polo di Monserrato, Cagliari, Italy
| | | | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari, Polo di Monserrato, Cagliari, Italy
| | - Marco Guglielmo
- Department of Cardiology, Division of Heart and Lungs, Utrecht University, Utrecht University Medical Center, Utrecht, the Netherlands
| | | | | | | | - Serena Dell’Aversana
- Department of Radiology, Ospedale S. Maria Delle Grazie - ASL Napoli 2 Nord, Pozzuoli, Italy
| | - U. Joseph Schoepf
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, Ashley River Tower, 25 Courtenay Dr., Charleston, SC, USA
| | - Mason G. Andrews
- Division of Cardiovascular Imaging, Department of Radiology and Radiological Science, Medical University of South Carolina, Ashley River Tower, 25 Courtenay Dr., Charleston, SC, USA
| | - Paolo Basile
- University Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Andrea Igoren Guaricci
- University Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Paolo Marra
- Department of Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Denisa Muraru
- School of Medicine, University of Milano-Bicocca, Milan, Italy,Department of Cardiac, Neurological and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Luigi P. Badano
- School of Medicine, University of Milano-Bicocca, Milan, Italy,Department of Cardiac, Neurological and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Sandro Sironi
- School of Medicine, University of Milano-Bicocca, Milan, Italy,Department of Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| |
Collapse
|
27
|
Mascherbauer K, Donà C, Koschutnik M, Dannenberg V, Nitsche C, Duca F, Heitzinger G, Halavina K, Steinacher E, Kronberger C, Bardach C, Beitzke D, Loewe C, Waldmann E, Trauner M, Barkto P, Goliasch G, Mascherbauer J, Hengstenberg C, Kammerlander A. Hepatic T1-Time Predicts Cardiovascular Risk in All-Comers Referred for Cardiovascular Magnetic Resonance: A Post-Hoc Analysis. Circ Cardiovasc Imaging 2022; 15:e014716. [PMID: 36256728 DOI: 10.1161/circimaging.122.014716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Liver damage is frequently observed in patients with cardiovascular disease but infrequently quantified. We hypothesized that in patients with cardiovascular disease undergoing cardiac magnetic resonance, liver T1-times indicate liver damage and are associated with cardiovascular outcome. METHODS We measured hepatic T1-times, displayed on standard cardiac T1-maps, in an all-comer cardiac magnetic resonance-cohort. At the time of cardiac magnetic resonance, we assessed validated general liver fibrosis scores. Kaplan-Meier estimates and Cox-regression models were used to investigate the association between hepatic T1-times and a composite endpoint of non-fatal myocardial infarction, heart failure hospitalization, and death. RESULTS One thousand seventy-five participants (58±18 year old, 47% female) were included (972 patients, 50 controls, 53 participants with transient elastography). Hepatic T1-times were 590±89 ms in patients and 574±45 ms in controls (P=0.052). They were significantly correlated with cardiac size and function, presence of atrial fibrillation, NT-pro-BNP levels, and gamma-glutamyl-transferase levels (P<0.001 for all). During follow-up (58±31 months), a total of 280 (29%) events occurred. On Cox-regression, high hepatic T1-times yielded a significantly higher risk for events (adjusted hazard ratio, 1.66 [95% CI, 1.45-1.89] per 100 ms increase; P<0.001), even when adjusted for age, sex, left and right ventricular ejection fraction, NT-proBNP (N-terminal prohormone of brain natriuretic peptide), and myocardial T1-time. On receiver operating characteristic analysis and restricted cubic splines, we found that a hepatic T1-time exceeding 610 ms was associated with excessive risk. CONCLUSIONS Hepatic T1-times on standard cardiac magnetic resonance scans were significantly associated with cardiac size and function, comorbidities, natriuretic peptides, and independently predicted cardiovascular mortality and morbidity. A hepatic T1-time >610 ms seems to indicate excessive risk. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT04220450.
Collapse
Affiliation(s)
- Katharina Mascherbauer
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Carolina Donà
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Matthias Koschutnik
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Varius Dannenberg
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Christian Nitsche
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Franz Duca
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Gregor Heitzinger
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Kseniya Halavina
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Eva Steinacher
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Christina Kronberger
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Constanze Bardach
- Division of Cardiovascular and Interventional Radiology (C.B., D.B., C.L.), Medical University of Vienna
| | - Dietrich Beitzke
- Division of Cardiovascular and Interventional Radiology (C.B., D.B., C.L.), Medical University of Vienna
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology (C.B., D.B., C.L.), Medical University of Vienna
| | - Elisabeth Waldmann
- Division of Gastroenterology and Hepatology (E.W., M.T.), Medical University of Vienna
| | - Michael Trauner
- Division of Gastroenterology and Hepatology (E.W., M.T.), Medical University of Vienna
| | - Philipp Barkto
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Georg Goliasch
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Julia Mascherbauer
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna.,Karl Landsteiner University of Health Sciences, Department of Internal Medicine 3, University Hospital St. Pölten, Krems, Austria (J.M.)
| | - Christian Hengstenberg
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| | - Andreas Kammerlander
- Division of Cardiology (K.M., C.D., M.K., V.D., C.N., F.D., G.H., K.H., E.S., C.K., P.B., G.G., J.M., C.H., A.K.), Medical University of Vienna
| |
Collapse
|
28
|
Puntmann VO, Martin S, Shchendrygina A, Hoffmann J, Ka MM, Giokoglu E, Vanchin B, Holm N, Karyou A, Laux GS, Arendt C, De Leuw P, Zacharowski K, Khodamoradi Y, Vehreschild MJGT, Rohde G, Zeiher AM, Vogl TJ, Schwenke C, Nagel E. Long-term cardiac pathology in individuals with mild initial COVID-19 illness. Nat Med 2022; 28:2117-2123. [PMID: 36064600 PMCID: PMC9556300 DOI: 10.1038/s41591-022-02000-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/08/2022] [Indexed: 01/03/2023]
Abstract
Cardiac symptoms are increasingly recognized as late complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in previously well individuals with mild initial illness, but the underlying pathophysiology leading to long-term cardiac symptoms remains unclear. In this study, we conducted serial cardiac assessments in a selected population of individuals with Coronavirus Disease 2019 (COVID-19) with no previous cardiac disease or notable comorbidities by measuring blood biomarkers of heart injury or dysfunction and by performing magnetic resonance imaging. Baseline measurements from 346 individuals with COVID-19 (52% females) were obtained at a median of 109 days (interquartile range (IQR), 77-177 days) after infection, when 73% of participants reported cardiac symptoms, such as exertional dyspnea (62%), palpitations (28%), atypical chest pain (27%) and syncope (3%). Symptomatic individuals had higher heart rates and higher imaging values or contrast agent accumulation, denoting inflammatory cardiac involvement, compared to asymptomatic individuals. Structural heart disease or high levels of biomarkers of cardiac injury or dysfunction were rare in symptomatic individuals. At follow-up (329 days (IQR, 274-383 days) after infection), 57% of participants had persistent cardiac symptoms. Diffuse myocardial edema was more pronounced in participants who remained symptomatic at follow-up as compared to those who improved. Female gender and diffuse myocardial involvement on baseline imaging independently predicted the presence of cardiac symptoms at follow-up. Ongoing inflammatory cardiac involvement may, at least in part, explain the lingering cardiac symptoms in previously well individuals with mild initial COVID-19 illness.
Collapse
Affiliation(s)
- Valentina O Puntmann
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany.
| | - Simon Martin
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Anastasia Shchendrygina
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Jedrzej Hoffmann
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mame Madjiguène Ka
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eleni Giokoglu
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Byambasuren Vanchin
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Niels Holm
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Argyro Karyou
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Gerald S Laux
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christophe Arendt
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Kai Zacharowski
- Department of Anesthesiology, Intensive Care Medicine & Pain Therapy; Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Yascha Khodamoradi
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maria J G T Vehreschild
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Gernot Rohde
- Department of Internal Medicine, Respiratory Medicine, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Andreas M Zeiher
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Eike Nagel
- Institute for Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Frankfurt, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Automated Dual-energy Computed Tomography-based Extracellular Volume Estimation for Myocardial Characterization in Patients With Ischemic and Nonischemic Cardiomyopathy. J Thorac Imaging 2022; 37:307-314. [PMID: 35475983 DOI: 10.1097/rti.0000000000000656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES We aimed to validate and test a prototype algorithm for automated dual-energy computed tomography (DECT)-based myocardial extracellular volume (ECV) assessment in patients with various cardiomyopathies. METHODS This retrospective study included healthy subjects (n=9; 61±10 y) and patients with cardiomyopathy (n=109, including a validation cohort n=60; 68±9 y; and a test cohort n=49; 69±11 y), who had previously undergone cardiac DECT. Myocardial ECV was calculated using a prototype-based fully automated algorithm and compared with manual assessment. Receiver-operating characteristic analysis was performed to test the algorithm's ability to distinguish healthy subjects and patients with cardiomyopathy. RESULTS The fully automated method led to a significant reduction of postprocessing time compared with manual assessment (2.2±0.4 min and 9.4±0.7 min, respectively, P <0.001). There was no significant difference in ECV between the automated and manual methods ( P =0.088). The automated method showed moderate correlation and agreement with the manual technique ( r =0.68, intraclass correlation coefficient=0.66). ECV was significantly higher in patients with cardiomyopathy compared with healthy subjects, regardless of the method used ( P <0.001). In the test cohort, the automated method yielded an area under the curve of 0.98 for identifying patients with cardiomyopathies. CONCLUSION Automated ECV estimation based on DECT showed moderate agreement with the manual method and matched with previously reported ECV values for healthy volunteers and patients with cardiomyopathy. The automatically derived ECV demonstrated an excellent diagnostic performance to discriminate between healthy and diseased myocardium, suggesting that it could be an effective initial screening tool while significantly reducing the time of assessment.
Collapse
|
30
|
Mavrogeni S, Pepe A, Nijveldt R, Ntusi N, Sierra-Galan LM, Bratis K, Wei J, Mukherjee M, Markousis-Mavrogenis G, Gargani L, Sade LE, Ajmone-Marsan N, Seferovic P, Donal E, Nurmohamed M, Cerinic MM, Sfikakis P, Kitas G, Schwitter J, Lima JAC, Dawson D, Dweck M, Haugaa KH, Keenan N, Moon J, Stankovic I, Donal E, Cosyns B. Cardiovascular magnetic resonance in autoimmune rheumatic diseases: a clinical consensus document by the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 2022; 23:e308-e322. [PMID: 35808990 DOI: 10.1093/ehjci/jeac134] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/12/2022] Open
Abstract
Autoimmune rheumatic diseases (ARDs) involve multiple organs including the heart and vasculature. Despite novel treatments, patients with ARDs still experience a reduced life expectancy, partly caused by the higher prevalence of cardiovascular disease (CVD). This includes CV inflammation, rhythm disturbances, perfusion abnormalities (ischaemia/infarction), dysregulation of vasoreactivity, myocardial fibrosis, coagulation abnormalities, pulmonary hypertension, valvular disease, and side-effects of immunomodulatory therapy. Currently, the evaluation of CV involvement in patients with ARDs is based on the assessment of cardiac symptoms, coupled with electrocardiography, blood testing, and echocardiography. However, CVD may not become overt until late in the course of the disease, thus potentially limiting the therapeutic window for intervention. More recently, cardiovascular magnetic resonance (CMR) has allowed for the early identification of pathophysiologic structural/functional alterations that take place before the onset of clinically overt CVD. CMR allows for detailed evaluation of biventricular function together with tissue characterization of vessels/myocardium in the same examination, yielding a reliable assessment of disease activity that might not be mirrored by blood biomarkers and other imaging modalities. Therefore, CMR provides diagnostic information that enables timely clinical decision-making and facilitates the tailoring of treatment to individual patients. Here we review the role of CMR in the early and accurate diagnosis of CVD in patients with ARDs compared with other non-invasive imaging modalities. Furthermore, we present a consensus-based decision algorithm for when a CMR study could be considered in patients with ARDs, together with a standardized study protocol. Lastly, we discuss the clinical implications of findings from a CMR examination.
Collapse
Affiliation(s)
- S Mavrogeni
- Onassis Cardiac Surgery Center, Leof. Andrea Siggrou 356, Kallithea 176 74, Greece.,Exercise Physiology and Sport Medicine Clinic, Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 115 27 Athens, Greece
| | - A Pepe
- Institute of Radiology, Department of Medicine, University of Padua, 35122 Padua, Italy
| | - R Nijveldt
- Department of Cardiology, Radboud University Medical Center, 6525 GA, Nijmegen, the Netherlands
| | - N Ntusi
- University of Cape Town & Groote Schuur Hospital, City of Cape Town, 7700 Western Cape, South Africa
| | - L M Sierra-Galan
- Department of Cardiology, American British Cowdray Medical Center, 05330 Mexico City, Mexico
| | - K Bratis
- Department of Cardiology, Manchester Royal Infirmary, Manchester M13 9WL, UK
| | - J Wei
- Barbra Streisand Women's Heart Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA 90048, USA.,Preventive and Rehabilitative Cardiac Center, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA 90048, USA
| | - M Mukherjee
- Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - L Gargani
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56126 Pisa, Italy
| | - L E Sade
- University of Pittsburgh, University of Pittsburgh Medical Center, Heart and Vascular Institute, Pittsburgh, PA 15260, USA.,Department of Cardiology, Baskent University, 06790 Ankara, Turkey
| | - N Ajmone-Marsan
- Department of Cardiology, Leiden University Medical Center, 2311 EZ Leiden, the Netherlands
| | - P Seferovic
- Department of Cardiology, Belgrade University, 11000 Belgrade, Serbia
| | - E Donal
- Université RENNES-1, CHU, 35000 Rennes, France
| | - M Nurmohamed
- Amsterdam Rheumatology Immunology Center, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, the Netherlands
| | - M Matucci Cerinic
- Experimental and Clinical Medicine, Division of Internal Medicine and Rheumatology, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50121 Florence, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS, San Raffaele Hospital, 20132 Milan, Italy
| | - P Sfikakis
- First Department of Propeudeutic and Internal medicine, Laikon Hospital, Athens University Medical School, 115 27 Athens, Greece
| | - G Kitas
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - J Schwitter
- Lausanne University Hospital, CHUV, CH-1011 Lausanne, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, 1015 UniL, Switzerland.,Director CMR Center of the University Hospital Lausanne, CHUV, CH-1011 Lausanne, Switzerland
| | - J A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Comparison of Hepatic Tissue Characterization between T1-Mapping and Non-Contrast Computed Tomography. J Clin Med 2022; 11:jcm11102863. [PMID: 35628989 PMCID: PMC9144343 DOI: 10.3390/jcm11102863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 01/27/2023] Open
Abstract
Background: Non-contrast computed tomography (CT) is frequently used to assess non-alcoholic/metabolic fatty liver disease (NAFLD/MAFLD), which is associated with cardiovascular risk. Although liver biopsy is considered the gold standard for diagnosis, standardized scores and non-contrast computed tomography (CT) are used instead. On standard cardiac T1-maps on cardiovascular imaging (CMR) exams for myocardial tissue characterization hepatic tissue is also visible. We hypothesized that there is a significant correlation between hepatic tissue T1-times on CMR and Hounsfield units (HU) on non-contrast CT. Methods: We retrospectively identified patients undergoing a non-contrast CT including the abdomen, a CMR including T1-mapping, and laboratory assessment within 30 days. Patients with storage diseases were excluded. Results: We identified 271 patients (62 ± 15 y/o, 49% female) undergoing non-contrast CT and CMR T1-mapping within 30 days. Mean hepatic HU values were 54 ± 11 on CT and native T1-times were 598 ± 102 ms on CMR and there was a weak, but significant, correlation between these parameters (r = −0.136, p = 0.025). On age and sex adjusted regression analysis, lower liver HU values indicated a dismal cardiometabolic risk profile, including higher HbA1C (p = 0.005) and higher body mass index (p < 0.001). In contrast, native hepatic T1-times yielded a more pronounced cardiac risk profile, including impaired systolic function (p = 0.045) and higher NT-proBNP values (N-Terminal Brain Natriuretic Peptide) (p = 0.004). Conclusions: Hepatic T1-times are easy to assess on standard T1-maps on CMR but only weakly correlated with hepatic HU values on CT and clinical NAFLD/MAFLD scores. Liver T1-times, however, are linked to impaired systolic function and higher natriuretic peptide levels. The prognostic value and clinical usefulness of hepatic T1-times in CMR cohorts warrants further research.
Collapse
|
32
|
Frantz S, Hundertmark MJ, Schulz-Menger J, Bengel FM, Bauersachs J. Left ventricular remodelling post-myocardial infarction: pathophysiology, imaging, and novel therapies. Eur Heart J 2022; 43:2549-2561. [PMID: 35511857 PMCID: PMC9336586 DOI: 10.1093/eurheartj/ehac223] [Citation(s) in RCA: 268] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022] Open
Abstract
Most patients survive acute myocardial infarction (MI). Yet this encouraging development has certain drawbacks: heart failure (HF) prevalence is increasing and patients affected tend to have more comorbidities worsening economic strain on healthcare systems and impeding effective medical management. The heart’s pathological changes in structure and/or function, termed myocardial remodelling, significantly impact on patient outcomes. Risk factors like diabetes, chronic obstructive pulmonary disease, female sex, and others distinctly shape disease progression on the ‘road to HF’. Despite the availability of HF drugs that interact with general pathways involved in myocardial remodelling, targeted drugs remain absent, and patient risk stratification is poor. Hence, in this review, we highlight the pathophysiological basis, current diagnostic methods and available treatments for cardiac remodelling following MI. We further aim to provide a roadmap for developing improved risk stratification and novel medical and interventional therapies.
Collapse
Affiliation(s)
- Stefan Frantz
- Department of Internal Medicine I, Universitätsklinikum Würzburg, University Hospital Würzburg, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Moritz Jens Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeanette Schulz-Menger
- Department of Cardiology and Nephrology, Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine and HELIOS Hospital Berlin Buch, Berlin, Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Kolentinis M, Carerj LM, Vidalakis E, Giokoglu E, Martin S, Arendt C, Vogl TJ, Nagel E, Puntmann VO. Determination of scar area using native and post-contrast T1 mapping: Agreement with late gadolinium enhancement. Eur J Radiol 2022; 150:110242. [PMID: 35290909 DOI: 10.1016/j.ejrad.2022.110242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/28/2022] [Accepted: 03/05/2022] [Indexed: 11/15/2022]
Abstract
The purpose of this study is to ascertain agreement in measurements of the scar area between late gadolinium enhancement (LGE), native and post-contrast T1 mapping in patients with known ischemic heart disease. 132 patients (age 60 ± 11 yrs, male 82%) were included in the study. Corresponding 3 short axis slices images of LGE, native and post contrast T1 mapping were used. Scar area was evaluated semi- quantitatively with FWHM methods, in which scar is automatically determined by specialized post-processing software. Agreement per culprit vessel was also assessed. Concordance and inter- intraobserver reproducibility were assessed with Bland-Altman analysis. The results show that scar area amounted to 12.6% of myocardium for LGE, 9.1% for native (p < 0.05) and 19.4% (p < 0.05) for post-contrast T1 mapping. LAD and RCA territory infarcts showed statistical discrepancy for both T1 acquisitions. Intraobserver differences in infarct size were comparable at 0.39% ± 0.28, 2.93% ± 0.03 and 0.97% ± 0.01 respectively (p≫0.05). Interobserver differences were 5.56% ± 0.91 for LGE, 11.87% ± 3.21 (p < 0.05) for native and 5.55% ± 2.87 (p≫0.05) for post-contrast T1 mapping. In conclusion, native T1 acquisitions systematically underestimated infarct size in comparison to LGE, while post-contrast T1 overestimated it. Variances in measurements were most pronounced for LAD and RCA territory infarcts. Intraobserver reproducibility was similar with both methods, whereas interobserver variability for native T1 mapping acquisition was worse.
Collapse
Affiliation(s)
- Michael Kolentinis
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Ludovica M Carerj
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Piazza Pugliatti 1, 98122, Messina, Italy
| | - Eleftherios Vidalakis
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Eleni Giokoglu
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Department of Cardiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Simon Martin
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Christophe Arendt
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Eike Nagel
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Valentina O Puntmann
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, partner site Rhein-Main, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
34
|
Marques MD, Weinberg R, Kapoor S, Ostovaneh MR, Kato Y, Liu CY, Shea S, McClelland RL, Post WS, Bluemke DA, Lima JAC, Ambale-Venkatesh B. Myocardial fibrosis by T1 mapping magnetic resonance imaging predicts incident cardiovascular events and all-cause mortality: the Multi-Ethnic Study of Atherosclerosis. Eur Heart J Cardiovasc Imaging 2022; 23:1407-1416. [PMID: 35147665 PMCID: PMC9463991 DOI: 10.1093/ehjci/jeac010] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Indexed: 11/14/2022] Open
Abstract
AIMS To evaluate whether myocardial fibrosis predicts cardiovascular events (CVEs) and mortality in the Multi-Ethnic Study of Atherosclerosis. METHODS AND RESULTS Cardiac magnetic resonance (CMR) T1 mapping with gadolinium administration for assessment of extracellular volume fraction (ECV) was performed in 1326 participants, in whom myocardial scar was assessed by late gadolinium enhancement (LGE). The clinical outcomes were defined as all-cause mortality, atherosclerotic CVEs, and incident heart failure (HF) during an average of 8 years of follow-up after the scan. Participants' mean native T1 time was 971 ms [standard deviation (SD) 45.5], ECV was 27 (SD 2.9), and 117 (8.8%) of them had LGE. At the time of the CMR exam, participant age was 68 years (SD 9) and 48% of them were women. Ideal cut-offs were identified using classification and regression trees accounting for time-to-event outcomes for ECV (30%) and native T1 time (954 ms). Over the follow-up period, 106 participants died, 78 developed CVE, and 23 developed HF. After adjustment for risk factors, ECV >30% was associated with death [hazard ratio (HR): 1.67, P < 0.05], incident CVE (HR: 2.02, P < 0.05), and incident HF (HR: 2.85, P < 0.05). After adjustments, native T1 >954 ms was associated with incident CVE (HR: 2.09, P < 0.05). Myocardial scar by LGE was not predictive of clinical outcomes after adjustments. CONCLUSION ECV is an independent prognostic marker of incident HF, atherosclerotic CVEs, and all-cause mortality. ECV, with its ability to characterize both diffuse and focal fibrosis processes, better predicted incident events than regional myocardial abnormalities as visualized by LGE imaging in a large multi-ethnic population.
Collapse
Affiliation(s)
- Mateus D Marques
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA,Cardiology, Federal University of Santa Maria, 1000 Cidade Universitária Bairro - Camobi, Santa Maria - RS, 97105-900, Brazil
| | - Raquel Weinberg
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Shrey Kapoor
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Mohammad R Ostovaneh
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA,Pennsylvania State Milton S. Hershey Medical Center, 500 University Dr, Hershey, PA 17033, USA
| | - Yoko Kato
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Chia Ying Liu
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - Steven Shea
- Division of General Medicine, Vagelos College of Physicians & Surgeons, Columbia University, 630 W 168th St, New York, NY 10032, USA
| | - Robyn L McClelland
- Biostatistics, University of Washington, Bldg. 29, Suite 210 Seattle, WA 98115, USA
| | - Wendy S Post
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin, 3252 Clinical Science Center 600 Highland Ave Madison, WI 53792
| | - João A C Lima
- Department of Cardiology, School of Medicine, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD, 21287, USA
| | | |
Collapse
|
35
|
Muscogiuri G, Guglielmo M, Serra A, Gatti M, Volpato V, Schoepf UJ, Saba L, Cau R, Faletti R, McGill LJ, De Cecco CN, Pontone G, Dell’Aversana S, Sironi S. Multimodality Imaging in Ischemic Chronic Cardiomyopathy. J Imaging 2022; 8:jimaging8020035. [PMID: 35200737 PMCID: PMC8877428 DOI: 10.3390/jimaging8020035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
Ischemic chronic cardiomyopathy (ICC) is still one of the most common cardiac diseases leading to the development of myocardial ischemia, infarction, or heart failure. The application of several imaging modalities can provide information regarding coronary anatomy, coronary artery disease, myocardial ischemia and tissue characterization. In particular, coronary computed tomography angiography (CCTA) can provide information regarding coronary plaque stenosis, its composition, and the possible evaluation of myocardial ischemia using fractional flow reserve CT or CT perfusion. Cardiac magnetic resonance (CMR) can be used to evaluate cardiac function as well as the presence of ischemia. In addition, CMR can be used to characterize the myocardial tissue of hibernated or infarcted myocardium. Echocardiography is the most widely used technique to achieve information regarding function and myocardial wall motion abnormalities during myocardial ischemia. Nuclear medicine can be used to evaluate perfusion in both qualitative and quantitative assessment. In this review we aim to provide an overview regarding the different noninvasive imaging techniques for the evaluation of ICC, providing information ranging from the anatomical assessment of coronary artery arteries to the assessment of ischemic myocardium and myocardial infarction. In particular this review is going to show the different noninvasive approaches based on the specific clinical history of patients with ICC.
Collapse
Affiliation(s)
- Giuseppe Muscogiuri
- Department of Radiology, Istituto Auxologico Italiano IRCCS, San Luca Hospital, University Milano Bicocca, 20149 Milan, Italy
- Correspondence: ; Tel.: +39-329-404-9840
| | - Marco Guglielmo
- Department of Cardiology, Division of Heart and Lungs, Utrecht University, Utrecht University Medical Center, 3584 Utrecht, The Netherlands;
| | - Alessandra Serra
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, 09042 Cagliari, Italy; (A.S.); (L.S.); (R.C.)
| | - Marco Gatti
- Radiology Unit, Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (M.G.); (R.F.)
| | - Valentina Volpato
- Department of Cardiac, Neurological and Metabolic Sciences, Istituto Auxologico Italiano IRCCS, San Luca Hospital, University Milano Bicocca, 20149 Milan, Italy;
| | - Uwe Joseph Schoepf
- Department of Radiology and Radiological Science, MUSC Ashley River Tower, Medical University of South Carolina, 25 Courtenay Dr, Charleston, SC 29425, USA; (U.J.S.); (L.J.M.)
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, 09042 Cagliari, Italy; (A.S.); (L.S.); (R.C.)
| | - Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari-Polo di Monserrato, 09042 Cagliari, Italy; (A.S.); (L.S.); (R.C.)
| | - Riccardo Faletti
- Radiology Unit, Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (M.G.); (R.F.)
| | - Liam J. McGill
- Department of Radiology and Radiological Science, MUSC Ashley River Tower, Medical University of South Carolina, 25 Courtenay Dr, Charleston, SC 29425, USA; (U.J.S.); (L.J.M.)
| | - Carlo Nicola De Cecco
- Division of Cardiothoracic Imaging, Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA;
| | | | - Serena Dell’Aversana
- Department of Radiology, Ospedale S. Maria Delle Grazie—ASL Napoli 2 Nord, 80078 Pozzuoli, Italy;
| | - Sandro Sironi
- School of Medicine and Post Graduate School of Diagnostic Radiology, University of Milano-Bicocca, 20126 Milan, Italy;
- Department of Radiology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| |
Collapse
|
36
|
Prognostic value of non-contrast myocardial T1 mapping in cardiovascular diseases: a systematic review and meta-analysis. Heart Fail Rev 2022; 27:1899-1909. [DOI: 10.1007/s10741-021-10191-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 11/25/2022]
|
37
|
Arcari L, Camastra G, Ciolina F, Danti M, Cacciotti L. T1 and T2 Mapping in Uremic Cardiomyopathy: An Update. Card Fail Rev 2022; 8:e02. [PMID: 35111336 PMCID: PMC8790724 DOI: 10.15420/cfr.2021.19] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/15/2021] [Indexed: 11/04/2022] Open
Abstract
Uremic cardiomyopathy (UC) is the cardiac remodelling that occurs in patients with chronic kidney disease (CKD). It is characterised by a left ventricular (LV) hypertrophy phenotype, diastolic dysfunction and generally preserved LV ejection fraction. UC has a major role mediating the increased rate of cardiovascular events, especially heart failure related, observed in patients with CKD. Recently, the use of T1 and T2 mapping techniques on cardiac MRI has expanded the ability to characterise cardiac involvement in CKD. Native T1 mapping effectively tracks the progression of interstitial fibrosis in UC, whereas T2 mapping analysis suggests the contribution of myocardial oedema, at least in a subgroup of patients. Both T1 and T2 increased values were related to worsening clinical status, myocardial injury and B-type natriuretic peptide release. Studies investigating the prognostic relevance and histology validation of mapping techniques in CKD are awaited.
Collapse
Affiliation(s)
- Luca Arcari
- Cardiology Unit, Madre Giuseppina Vannini Hospital, Rome, Italy
| | | | | | | | - Luca Cacciotti
- Cardiology Unit, Madre Giuseppina Vannini Hospital, Rome, Italy
| |
Collapse
|
38
|
Gleditsch J, Jervan Ø, Tavoly M, Geier O, Holst R, Klok FA, Ghanima W, Hopp E. Association between myocardial fibrosis, as assessed with cardiac magnetic resonance T1 mapping, and persistent dyspnea after pulmonary embolism. IJC HEART & VASCULATURE 2022; 38:100935. [PMID: 35005213 PMCID: PMC8717259 DOI: 10.1016/j.ijcha.2021.100935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/19/2021] [Indexed: 11/25/2022]
Abstract
Background Persistent dyspnea is a common symptom after pulmonary embolism (PE). However, the pathophysiology of persistent dyspnea is not fully clarified. This study aimed to explore possible associations between diffuse myocardial fibrosis, as assessed by cardiac magnetic resonance (CMR) T1 mapping, and persistent dyspnea in patients with a history of PE. Methods CMR with T1 mapping and extracellular volume fraction (ECV) calculations were performed after PE in 51 patients with persistent dyspnea and in 50 non-dyspneic patients. Patients with known pulmonary disease, heart disease and CTEPH were excluded. Results Native T1 was higher in the interventricular septum in dyspneic patients compared to non-dyspneic patients; difference 13 ms (95% CI: 2–23 ms). ECV was also significantly higher in patients with dyspnea; difference 0.9 percent points (95% CI: 0.04–1.8 pp). There was no difference in native T1 or ECV in the left ventricular lateral wall. Native T1 in the interventricular septum had an adjusted Odds Ratio of 1.18 per 10 ms increase (95% CI: 0.99–1.42) in predicting dyspnea, and an adjusted Odds Ratio of 1.47 per 10 ms increase (95% CI: 1.10–1.96) in predicting Incremental Shuttle Walk Test (ISWT) score < 1020 m. Conclusion Septal native T1 and ECV values were higher in patients with dyspnea after PE compared with those who were fully recovered suggesting a possible pathological role of myocardial fibrosis in the development of dyspnea after PE. Further studies are needed to validate our findings and to explore their pathophysiological role and clinical significance.
Collapse
Affiliation(s)
- Jostein Gleditsch
- Department of Radiology, Østfold Hospital, Kalnes, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Øyvind Jervan
- Department of Cardiology, Østfold Hospital, Kalnes, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mazdak Tavoly
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Oliver Geier
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - René Holst
- Department of Research, Østfold Hospital, Kalnes, Norway.,Oslo Centre for Biostatistics and Epidemiology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Frederikus A Klok
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Waleed Ghanima
- Internal medicine clinic, Østfold Hospital, Kalnes, Norway.,Department of hematology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Einar Hopp
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
39
|
Dumitru RB, Bissell LA, Erhayiem B, Kidambi A, Dumitru AMH, Fent G, Abignano G, Donica H, Burska A, Greenwood JP, Biglands J, Schlosshan D, Del Galdo F, Plein S, Buch MH. Cardiovascular outcomes in systemic sclerosis with abnormal cardiovascular MRI and serum cardiac biomarkers. RMD Open 2021; 7:rmdopen-2021-001689. [PMID: 34663635 PMCID: PMC8524374 DOI: 10.1136/rmdopen-2021-001689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022] Open
Abstract
Objectives To explore the prognostic value of subclinical cardiovascular (CV) imaging measures and serum cardiac biomarkers in systemic sclerosis (SSc) for the development of CV outcomes of primary heart involvement (pHI). Methods Patients with SSc with no clinical SSc-pHI and no history of heart disease underwent cardiovascular magnetic resonance (CMR) imaging, and measurement of serum high-sensitivity-troponin I (hs-TnI) and N-terminal-pro-brain natriuretic peptide (NT-proBNP). Follow-up clinical and CV outcome data were recorded. CV outcomes were defined as myocarditis, arrhythmia and/or echocardiographic functional impairment including systolic dysfunction and/or diastolic dysfunction. Results Seventy-four patients with a median (IQR) age of 57 (49, 63) years, 32% diffuse cutaneous SSc, 39% interstitial lung disease, 30% Scl70+ were followed up for median (IQR) 22 (15, 54) months. Ten patients developed CV outcomes, comprising one patient with myocarditis and systolic dysfunction and nine arrhythmias: three non-sustained ventricular tachycardia and six supraventricular arrhythmias. The probability of CV outcomes was considerably higher in those with NT-proBNP >125 pg/mL versus normal NT-proBNP (X2=4.47, p=0.035). Trend for poorer time-to-event was noted in those with higher extracellular volume (ECV; indicating diffuse fibrosis) and hs-TnI levels versus those with normal values (X2=2.659, p=0.103; X2=2.530, p=0.112, respectively). In a predictive model, NT-proBNP >125 pg/mL associated with CV outcomes (OR=5.335, p=0.040), with a trend observed for ECV >29% (OR=4.347, p=0.073). Conclusion These data indicate standard serum cardiac biomarkers (notably NT-proBNP) and CMR indices of myocardial fibrosis associate with adverse CV outcomes in SSc. This forms the basis to develop a prognostic model in larger, longitudinal studies.
Collapse
Affiliation(s)
- Raluca B Dumitru
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - Lesley-Anne Bissell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - Bara Erhayiem
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK
| | - Ananth Kidambi
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK.,Leeds Teaching Hospitals NHS Trust Department of Cardiology, Leeds, West Yorkshire, UK
| | - Ana-Maria H Dumitru
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,Faculty of Business Economics and Law, University of Surrey, Guildford, Surrey, UK
| | - Graham Fent
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK
| | - Giuseppina Abignano
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - Helena Donica
- Department of Biochemical Diagnostics, Medical University of Lublin, Lublin, Lubelskie, Poland
| | - Agata Burska
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - John P Greenwood
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK.,Leeds Teaching Hospitals NHS Trust Department of Cardiology, Leeds, West Yorkshire, UK
| | - John Biglands
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - Dominik Schlosshan
- Leeds Teaching Hospitals NHS Trust Department of Cardiology, Leeds, West Yorkshire, UK
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK.,National Institute for Health Research, Leeds Biomedical Research Centre, Leeds, West Yorkshire, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK.,Leeds Teaching Hospitals NHS Trust Department of Cardiology, Leeds, West Yorkshire, UK
| | - Maya H Buch
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds Faculty of Medicine and Health, Leeds, West Yorkshire, UK .,Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
40
|
Argulian E, Narula J. Advanced Cardiovascular Imaging in Clinical Heart Failure. JACC-HEART FAILURE 2021; 9:699-709. [PMID: 34391742 DOI: 10.1016/j.jchf.2021.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Cardiovascular imaging is the cornerstone of the assessment of patients with heart failure. Although noninvasive volumetric estimation of the cardiac function is an essential and indisputably useful clinical tool, cardiac imaging has evolved and matured to offer detailed functional, hemodynamic, and tissue characterization. The adoption of a new framework to diagnose and phenotype heart failure that incorporates comprehensive imaging assessment has been lacking in clinical trials. The present review offers a general overview of available imaging strategies for patients with heart failure.
Collapse
Affiliation(s)
- Edgar Argulian
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
41
|
Almeida AG, Carpenter JP, Cameli M, Donal E, Dweck MR, Flachskampf FA, Maceira AM, Muraru D, Neglia D, Pasquet A, Plein S, Gerber BL. Multimodality imaging of myocardial viability: an expert consensus document from the European Association of Cardiovascular Imaging (EACVI). Eur Heart J Cardiovasc Imaging 2021; 22:e97-e125. [PMID: 34097006 DOI: 10.1093/ehjci/jeab053] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Indexed: 12/17/2022] Open
Abstract
In clinical decision making, myocardial viability is defined as myocardium in acute or chronic coronary artery disease and other conditions with contractile dysfunction but maintained metabolic and electrical function, having the potential to improve dysfunction upon revascularization or other therapy. Several pathophysiological conditions may coexist to explain this phenomenon. Cardiac imaging may allow identification of myocardial viability through different principles, with the purpose of prediction of therapeutic response and selection for treatment. This expert consensus document reviews current insight into the underlying pathophysiology and available methods for assessing viability. In particular the document reviews contemporary viability imaging techniques, including stress echocardiography, single photon emission computed tomography, positron emission tomography, cardiovascular magnetic resonance, and computed tomography and provides clinical recommendations for how to standardize these methods in terms of acquisition and interpretation. Finally, it presents clinical scenarios where viability assessment is clinically useful.
Collapse
Affiliation(s)
- Ana G Almeida
- Faculty of Medicine, Lisbon University, University Hospital Santa Maria/CHLN, Portugal
| | - John-Paul Carpenter
- Cardiology Department, University Hospitals Dorset, NHS Foundation Trust, Poole Hospital, Longfleet Road, Poole, Dorset BH15 2JB, United Kingdom
| | - Matteo Cameli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Viale Bracci 16, Siena, Italy
| | - Erwan Donal
- Department of Cardiology, CHU Rennes, Inserm, LTSI-UMR 1099, Université de Rennes 1, Rennes F-35000, France
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, The University of Edinburgh & Edinburgh Heart Centre, Chancellors Building Little France Crescent, Edinburgh EH16 4SB, United Kingdom
| | - Frank A Flachskampf
- Dept. of Med. Sciences, Uppsala University, and Cardiology and Clinical Physiology, Uppsala University Hospital, Akademiska, 751 85 Uppsala, Sweden
| | - Alicia M Maceira
- Cardiovascular Imaging Unit, Ascires Biomedical Group Colon St, 1, Valencia 46004, Spain; Department of Medicine, Health Sciences School, CEU Cardenal Herrera University, Lluís Vives St. 1, 46115 Alfara del Patriarca, Valencia, Spain
| | - Denisa Muraru
- Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900, Monza, Italy; Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, Piazzale Brescia 20, 20149, Milan, Italy
| | - Danilo Neglia
- Fondazione Toscana G. Monasterio-Via G. Moruzzi 1, Pisa, Italy
| | - Agnès Pasquet
- Service de Cardiologie, Département Cardiovasculaire, Cliniques Universitaires St. Luc, and Division CARD, Institut de Recherche Expérimental et Clinique (IREC), UCLouvain, Av Hippocrate 10, B-1200 Brussels, Belgium
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, United Kingdom
| | - Bernhard L Gerber
- Department of Biomedical Imaging Science, Leeds, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds LS2 9JT, United Kingdom
| | | | | | | |
Collapse
|
42
|
de Leuw P, Arendt CT, Haberl AE, Froadinadl D, Kann G, Wolf T, Stephan C, Schuettfort G, Vasquez M, Arcari L, Zhou H, Zainal H, Gawor M, Vidalakis E, Kolentinis M, Albrecht MH, Escher F, Vogl TJ, Zeiher AM, Nagel E, Puntmann VO. Myocardial Fibrosis and Inflammation by CMR Predict Cardiovascular Outcome in People Living With HIV. JACC Cardiovasc Imaging 2021; 14:1548-1557. [PMID: 33865770 DOI: 10.1016/j.jcmg.2021.01.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The goal of this study was to examine prognostic relationships between cardiac imaging measures and cardiovascular outcome in people living with human immunodeficiency virus (HIV) (PLWH) on highly active antiretroviral therapy (HAART). BACKGROUND PLWH have a higher prevalence of cardiovascular disease and heart failure (HF) compared with the noninfected population. The pathophysiological drivers of myocardial dysfunction and worse cardiovascular outcome in HIV remain poorly understood. METHODS This prospective observational longitudinal study included consecutive PLWH on long-term HAART undergoing cardiac magnetic resonance (CMR) examination for assessment of myocardial volumes and function, T1 and T2 mapping, perfusion, and scar. Time-to-event analysis was performed from the index CMR examination to the first single event per patient. The primary endpoint was an adjudicated adverse cardiovascular event (cardiovascular mortality, nonfatal acute coronary syndrome, an appropriate device discharge, or a documented HF hospitalization). RESULTS A total of 156 participants (62% male; age [median, interquartile range]: 50 years [42 to 57 years]) were included. During a median follow-up of 13 months (9 to 19 months), 24 events were observed (4 HF deaths, 1 sudden cardiac death, 2 nonfatal acute myocardial infarction, 1 appropriate device discharge, and 16 HF hospitalizations). Patients with events had higher native T1 (median [interquartile range]: 1,149 ms [1,115 to 1,163 ms] vs. 1,110 ms [1,075 to 1,138 ms]); native T2 (40 ms [38 to 41 ms] vs. 37 ms [36 to 39 ms]); left ventricular (LV) mass index (65 g/m2 [49 to 77 g/m2] vs. 57 g/m2 [49 to 64 g/m2]), and N-terminal pro-B-type natriuretic peptide (109 pg/l [25 to 337 pg/l] vs. 48 pg/l [23 to 82 pg/l]) (all p < 0.05). In multivariable analyses, native T1 was independently predictive of adverse events (chi-square test, 15.9; p < 0.001; native T1 [10 ms] hazard ratio [95% confidence interval]: 1.20 [1.08 to 1.33]; p = 0.001), followed by a model that also included LV mass (chi-square test, 17.1; p < 0.001). Traditional cardiovascular risk scores were not predictive of the adverse events. CONCLUSIONS Our findings reveal important prognostic associations of diffuse myocardial fibrosis and LV remodeling in PLWH. These results may support development of personalized approaches to screening and early intervention to reduce the burden of HF in PLWH (International T1 Multicenter Outcome Study; NCT03749343).
Collapse
Affiliation(s)
- Philipp de Leuw
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany; Infektiologikum, Frankfurt am Main, Germany
| | - Christophe T Arendt
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Annette E Haberl
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Daniel Froadinadl
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Gerrit Kann
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Timo Wolf
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christoph Stephan
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Gundolf Schuettfort
- HIV Center, Department of Infectious Diseases, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Moises Vasquez
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Cardiology Department, Enrique Baltodano Briceño Hospital, Liberia, Costa Rica
| | - Luca Arcari
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Cardiology, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Hui Zhou
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Radiology, XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Hafisyatul Zainal
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Cardiology, Universiti Teknologi MARA (UiTM), Sg. Buloh, Malaysia
| | - Monika Gawor
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Cardiology, University Hospital Warsaw, Warsaw, Poland
| | - Eleftherios Vidalakis
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Michael Kolentinis
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Moritz H Albrecht
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Andreas M Zeiher
- Department of Cardiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eike Nagel
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Valentina O Puntmann
- Institute of Experimental and Translational Cardiac Imaging, DZHK (German Centre for Cardiovascular Research) Centre for Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Cardiology, University Hospital Warsaw, Warsaw, Poland.
| |
Collapse
|
43
|
Yang MX, Shi K, Xu HY, He Y, Ma M, Zhang L, Wang JL, Li XS, Fu C, Li H, Zhou B, Zhou XY, Yang Z, Guo YK, Yang ZG. Inflammation in Remote Myocardium and Left Ventricular Remodeling After Acute Myocardial Infarction: A Pilot Study Using T2 Mapping. J Magn Reson Imaging 2021; 55:555-564. [PMID: 34245075 DOI: 10.1002/jmri.27827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The pathophysiological changes in the remote myocardium after acute myocardial infarction (MI) remains less understood. PURPOSE To assess the inflammation in the remote myocardium post-MI and its association with left ventricular (LV) remodeling using T2 mapping. STUDY TYPE Prospective. ANIMAL MODEL AND SUBJECTS Twelve pigs at 3-day post-MI, 6 pigs at 3-month post-MI, 6 healthy pigs; 54 patients at 3-day and 3-month post-MI, 31 healthy volunteers; FIELD STRENGTH/SEQUENCE: A 3 T MRI/ steady-state free-precession sequence for T2 mapping (animals: 0, 30, and 55 msec; human: 0, 25, and 55 msec), phase-sensitive inversion recovery gradient echo for late gadolinium enhancement (LGE), balanced steady free-precession sequence for cine. ASSESSMENT Infarcted myocardium was defined on LGE, remote T2 was measured on T2 maps. LV remodeling was evaluated as LV end-diastolic volume change index between two scans using cine. CD68 staining was conducted to detect monocyte/macrophage. STATISTICAL TESTS Student-t test and one-way ANOVA were used to compare remote T2 with normal controls. The association of remote T2 with LV remodeling was assessed using linear regression. P values of <0.05 were used to denote statistical significance. RESULTS Compared with healthy pigs, remote T2 significantly increased from 3 days to 3 months post-MI (31.43 ± 0.67 vs. 33.53 ± 1.15 vs. 36.43 ± 1.07 msec). CD68 staining demonstrated the inflammation in remote myocardium post-MI but not in healthy pigs. Significant remote myocardial alterations in T2 were also observed in human group (40.51 ± 1.79 vs. 41.94 ± 1.14 vs. 42.52 ± 1.71 msec). In patients, the 3-month remote T2 (β = 0.432) and remote T2 variation between two scans (β = 0.554) were both independently associated with LV remodeling. CONCLUSION T2 mapping could characterize the abnormalities in the remote myocardium post-MI, which was potentially caused by the inflammatory response. Moreover, variations in remote T2 were associated with LV remodeling. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Meng-Xi Yang
- Department of Radiology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Ke Shi
- Department of Radiology, West China Hospital, Sichuan University, Sichuan, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Yong He
- Department of Cardiology, West China Hospital, Sichuan University, Sichuan, China
| | - Min Ma
- Department of Cardiology, West China Hospital, Sichuan University, Sichuan, China.,Department of Cardiology, The Sixth People's Hospital of Chengdu, Sichuan, China
| | - Lu Zhang
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | | | - Xue-Sheng Li
- Department of Radiology, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Chuan Fu
- Department of Radiology, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Hong Li
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Bin Zhou
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Xiao-Yue Zhou
- MR Collaboration, Siemens Healthcare Ltd, Shanghai, China
| | - Zhi Yang
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China.,Department of Radiology, Chengdu Fifth People's Hospital, Sichuan, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Zhi-Gang Yang
- Department of Radiology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
44
|
Bazoukis G, Papadatos SS, Michelongona A, Lampropoulos K, Farmakis D, Vassiliou V. Contemporary Role of Cardiac Magnetic Resonance in the Management of Patients with Suspected or Known Coronary Artery Disease. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:649. [PMID: 34202588 PMCID: PMC8303732 DOI: 10.3390/medicina57070649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/21/2021] [Accepted: 06/21/2021] [Indexed: 11/19/2022]
Abstract
Cardiac magnetic resonance imaging (CMR) is a useful non-invasive radiation-free imaging modality for the management of patients with coronary artery disease (CAD). CMR cine imaging provides the "gold standard" assessment of ventricular function, late gadolinium enhancement (LGE) provides useful data for the diagnosis and extent of myocardial scar and viability, while stress imaging is an established technique for the detection of myocardial perfusion defects indicating ischemia. Beyond its role in the diagnosis of CAD, CMR allows accurate risk stratification of patients with established CAD. This review aims to summarize the data regarding the role of CMR in the contemporary management of patients with suspected or known coronary artery disease.
Collapse
Affiliation(s)
- George Bazoukis
- Department of Cardiology, Larnaca General Hospital, 6051 Larnaca, Cyprus
| | - Stamatis S. Papadatos
- Department of Anatomy, Histology and Embryology, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece;
| | | | - Konstantinos Lampropoulos
- Department of Cardiology, General Hospital of Athens “Evangelismos”, 106 76 Athens, Greece;
- Department of Pathophysiology, School of Medicine, European University of Cyprus, 1678 Nicosia, Cyprus
| | - Dimitrios Farmakis
- Shakolas Educational Center for Clinical Medicine, University of Cyprus Medical School, Palaios Dromos Lefkosias Lemesou No.215/6, Aglantzia, 2029 Nicosia, Cyprus;
| | - Vassilis Vassiliou
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK;
| |
Collapse
|
45
|
Arcari L, Engel J, Freiwald T, Zhou H, Zainal H, Gawor M, Buettner S, Geiger H, Hauser I, Nagel E, Puntmann VO. Cardiac biomarkers in chronic kidney disease are independently associated with myocardial edema and diffuse fibrosis by cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2021; 23:71. [PMID: 34092229 PMCID: PMC8183054 DOI: 10.1186/s12968-021-00762-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 04/28/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND High sensitivity cardiac troponin T (hs-cTnT) and NT-pro-brain natriuretic peptide (NT-pro BNP) are often elevated in chronic kidney disease (CKD) and associated with both cardiovascular remodeling and outcome. Relationship between these biomarkers and quantitative imaging measures of myocardial fibrosis and edema by T1 and T2 mapping remains unknown. METHODS Consecutive patients with established CKD and estimated glomerular filtration rate (eGFR) < 59 ml/min/1.73 m2 (n = 276) were compared to age/sex matched patients with eGFR ≥ 60 ml/min/1.73 m2 (n = 242) and healthy controls (n = 38). Comprehensive cardiovascular magnetic resonance (CMR) with native T1 and T2 mapping, myocardial ischemia and scar imaging was performed with venous sampling immediately prior to CMR. RESULTS Patients with CKD showed significant cardiac remodeling in comparison with both healthy individuals and non-CKD patients, including a stepwise increase of native T1 and T2 (p < 0.001 between all CKD stages). Native T1 and T2 were the sole imaging markers independently associated with worsening CKD in patients [B = 0.125 (95% CI 0.022-0.235) and B = 0.272 (95% CI 0.164-0.374) with p = 0.019 and < 0.001 respectively]. At univariable analysis, both hs-cTnT and NT-pro BNP significantly correlated with native T1 and T2 in groups with eGFR 30-59 ml/min/1.73 m2 and eGFR < 29 ml/min/1.73 m2 groups, with associations being stronger at lower eGFR (NT-pro BNP (log transformed, lg10): native T1 r = 0.43 and r = 0.57, native T2 r = 0.39 and r = 0.48 respectively; log-transformed hs-cTnT(lg10): native T1 r = 0.23 and r = 0.43, native T2 r = 0.38 and r = 0.58 respectively, p < 0.001 for all, p < 0.05 for interaction). On multivariable analyses, we found independent associations of native T1 with NT-pro BNP [(B = 0.308 (95% CI 0.129-0.407), p < 0.001 and B = 0.334 (95% CI 0.154-0.660), p = 0.002 for eGFR 30-59 ml/min/1.73 m2 and eGFR < 29 ml/min/1.73 m2, respectively] and of T2 with hs-cTnT [B = 0.417 (95% CI 0.219-0.650), p < 0.001 for eGFR < 29 ml/min/1.73 m2]. CONCLUSIONS We demonstrate independent associations between cardiac biomarkers with imaging markers of interstitial expansion, which are CKD-group specific. Our findings indicate the role of diffuse non-ischemic tissue processes, including excess of myocardial fluid in addition to diffuse fibrosis in CKD-related adverse remodeling.
Collapse
Affiliation(s)
- Luca Arcari
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
- Cardiology Unit, Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Juergen Engel
- Department of Nephrology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tilo Freiwald
- Department of Nephrology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Hui Zhou
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Radiology, XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Hafisyatul Zainal
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
- Department of Cardiology, Universiti Teknologi MARA (UiTM), Sg. Buloh, Malaysia
| | - Monika Gawor
- Department of Cardiology, University Hospital Warsaw, Warsaw, Poland
| | - Stefan Buettner
- Department of Nephrology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Helmut Geiger
- Department of Nephrology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ingeborg Hauser
- Department of Nephrology, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eike Nagel
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Valentina O Puntmann
- Institute of Experimental and Translational Cardiovascular Imaging, DZHK Centre for Cardiovascular Imaging, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
46
|
Grigoratos C, Gueli I, Arendt CT, Leithner D, Meloni A, Nugara C, Barison A, Todiere G, Puntmann VO, Novo G, Pepe A, Emdin M, Nagel E, Aquaro GD. Prevalence and prognostic impact of nonischemic late gadolinium enhancement in stress cardiac magnetic resonance. J Cardiovasc Med (Hagerstown) 2021; 21:980-985. [PMID: 33156590 DOI: 10.2459/jcm.0000000000001016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIM To assess the prevalence and prognostic significance of NI-LGE in patients undergoing stress-CMR. METHODS Stress-CMR with either dipyridamole or adenosine was performed in 283 patients (228 men, 81%) including perfusion imaging, wall motion evaluation and LGE. Follow-up was completed in all enrolled patients (median time: 1850 days; interquartile range: 1225-2705 days). Composite endpoint included cardiac death, ventricular tachycardia, myocardial infarction, stroke, hospitalization for cardiac cause and coronary revascularization performed beyond 90 days from stress-CMR scans. RESULTS One hundred and twelve patients (40%) had negative LGE (no-LGE), 140 patients (49%) I-LGE and 31 patients (11%) NI-LGE. Twenty-five events occurred in the no-LGE group, 68 in I-LGE and 11 in the NI-LGE group. On survival curves, patients with NI-LGE had worse prognosis than patients with no-LGE regardless of the presence of inducible perfusion defects. No significant prognostic differences were found between I-LGE and NI-LGE. CONCLUSION NI-LGE can be detected in 11% of patients during stress-CMR providing a diagnosis of nonischemic cardiac disease. Patients with NI-LGE have worse prognosis than those with no-LGE.
Collapse
Affiliation(s)
- Chrysanthos Grigoratos
- Fondazione Gabriele Monasterio CNR/Regione Toscana.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Christophe T Arendt
- Institute for Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Doris Leithner
- Institute for Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Cinzia Nugara
- Fondazione Gabriele Monasterio CNR/Regione Toscana.,Division of Cardiology and Cardiovascular Rehabilitation, Department of Internal Medicine and Cardiovascular Disease, University Hospital Paolo Giaccone, Palermo, Italy
| | | | | | - Valentina O Puntmann
- DZHK Centre for Cardiovascular Imaging, Institute for Experimental and Translational Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Giuseppina Novo
- Division of Cardiology and Cardiovascular Rehabilitation, Department of Internal Medicine and Cardiovascular Disease, University Hospital Paolo Giaccone, Palermo, Italy
| | - Alessia Pepe
- Fondazione Gabriele Monasterio CNR/Regione Toscana
| | - Michele Emdin
- Fondazione Gabriele Monasterio CNR/Regione Toscana.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Eike Nagel
- DZHK Centre for Cardiovascular Imaging, Institute for Experimental and Translational Cardiovascular Imaging, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
47
|
Gupta S, Ge Y, Singh A, Gräni C, Kwong RY. Multimodality Imaging Assessment of Myocardial Fibrosis. JACC Cardiovasc Imaging 2021; 14:2457-2469. [PMID: 34023250 DOI: 10.1016/j.jcmg.2021.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Myocardial fibrosis, seen in ischemic and nonischemic cardiomyopathies, is associated with adverse cardiac outcomes. Noninvasive imaging plays a key role in early identification and quantification of myocardial fibrosis with the use of an expanding array of techniques including cardiac magnetic resonance, computed tomography, and nuclear imaging. This review discusses currently available noninvasive imaging techniques, provides insights into their strengths and limitations, and examines novel developments that will affect the future of noninvasive imaging of myocardial fibrosis.
Collapse
Affiliation(s)
- Sumit Gupta
- Department of Radiology Brigham and Women's Hospital, Boston, Massachusetts, USA; Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yin Ge
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Cardiology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Amitoj Singh
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christoph Gräni
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine and Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - 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
|
48
|
Anthony C, Imran M, Pouliopoulos J, Emmanuel S, Iliff JW, Moffat KJ, Ross J, Graham RM, Kotlyar E, Muthiah K, Keogh AM, Hayward CS, Macdonald P, Jabbour A. Everolimus for the Prevention of Calcineurin-Inhibitor-Induced Left Ventricular Hypertrophy After Heart Transplantation (RADTAC Study). JACC-HEART FAILURE 2021; 9:301-313. [PMID: 33795116 DOI: 10.1016/j.jchf.2021.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/04/2021] [Accepted: 01/24/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES This study aimed to determine the safety and efficacy of combined low-dose everolimus and low-dose tacrolimus compared with standard-dose tacrolimus in attenuating left ventricular hypertrophy (LVH) after orthotopic heart transplantation (OHT). BACKGROUND Calcineurin inhibitors (CNIs) such as tactrolimus are important in preventing cardiac allograft rejection and reducing mortality after OHT. However CNIs are causatively linked to the development of LVH, and are associated with nephrotoxicity and vasculopathy. CNI-sparing agents such as everolimus have been hypothesized to inhibit adverse effects of CNIs. METHODS In this prospective, randomized, open-label study, OHT recipients were randomized at 12 weeks after OHT to a combination of low-dose everolimus and tacrolimus (the RADTAC group) or standard-dose tacrolimus (the TAC group), with both groups coadministered mycophenolate and prednisolone. The primary endpoint was LVH indexed as the change in left ventricular mass (ΔLVM) by cardiovascular magnetic resonance (CMR) imaging from 12 to 52 weeks. Secondary endpoints included CMR-based myocardial performance, T1 fibrosis mapping, blood pressure, and renal function. Safety endpoints included episodes of allograft rejection and infection. RESULTS Forty stable OHT recipients were randomized. Recipients in the RADTAC group had significantly lower tacrolimus levels compared with the TAC group (6.5 ± 3.5 μg/l vs. 8.6 ± 2.8 μg/l; p = 0.02). The mean everolimus level in the RADTAC group was 4.2 ± 1.7 μg/l. A significant reduction in LVM was observed in the RADTAC group compared with an increase in LVM in the TAC group (ΔLVM = -13.0 ± 16.8 g vs. 2.1 ± 8.4 g; p < 0.001). Significant differences were also noted in secondary endpoints measuring function and fibrosis (Δ circumferential strain = -2.9 ± 2.8 vs. 2.1 ± 2.3; p < 0.001; ΔT1 mapping values = -32.7 ± 51.3 ms vs. 26.3 ± 90.4 ms; p = 0.003). No significant differences were observed in blood pressure (Δ mean arterial pressure = 4.2 ± 18.8 mm Hg vs. 2.8 ± 13.8 mm Hg; p = 0.77), renal function (Δ creatinine = 3.1 ± 19.9 μmol/l vs. 9 ± 21.8 μmol/l; p = 0.31), frequency of rejection episodes (p = 0.69), or frequency of infections (p = 0.67) between groups. CONCLUSIONS The combination of low-dose everolimus and tacrolimus compared with standard-dose tacrolimus safely attenuates LVH in the first year after cardiac transplantation with an observed reduction in CMR-measured fibrosis and an improvement in myocardial strain.
Collapse
Affiliation(s)
- Chris Anthony
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia
| | - Muhammad Imran
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia
| | - Jim Pouliopoulos
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Victor Chang Cardiac Research Institute
| | - Sam Emmanuel
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia
| | - James W Iliff
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia
| | - Kirsten J Moffat
- Medical Imaging Department, St. Vincent's Hospital, Sydney, Australia
| | - Joanne Ross
- Medical Imaging Department, St. Vincent's Hospital, Sydney, Australia
| | - Robert M Graham
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Victor Chang Cardiac Research Institute; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Eugene Kotlyar
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kavitha Muthiah
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Anne M Keogh
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Christopher S Hayward
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Victor Chang Cardiac Research Institute; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter Macdonald
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Victor Chang Cardiac Research Institute; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Jabbour
- Heart and Lung Transplant Unit, St. Vincent's Hospital, Sydney, Australia; Victor Chang Cardiac Research Institute; Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
49
|
Vietheer JM, Hamm CW, Rolf A. [Current role of cardiac magnetic resonance (CMR) in diagnosis and therapy of myocardial diseases]. Dtsch Med Wochenschr 2021; 146:461-465. [PMID: 33780992 DOI: 10.1055/a-1218-8972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cardiac magnetic resonance is the only imaging modality, that allows for characterising myocardial tissue with respect to fibrosis and edema. It has therefore become gold standard in diagnosing myocardial inflammation by combining scar, fibrosis and edema imaging. Recent developements in T1- and T2 mapping have improved diagnostic accuracy and prognostic information.
Collapse
|
50
|
Emrich T, Halfmann M, Schoepf UJ, Kreitner KF. CMR for myocardial characterization in ischemic heart disease: state-of-the-art and future developments. Eur Radiol Exp 2021; 5:14. [PMID: 33763757 PMCID: PMC7990980 DOI: 10.1186/s41747-021-00208-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/22/2021] [Indexed: 01/25/2023] Open
Abstract
Ischemic heart disease and its sequelae are one of the major contributors to morbidity and mortality worldwide. Over the last decades, technological developments have strengthened the role of noninvasive imaging for detection, risk stratification, and management of patients with ischemic heart disease. Cardiac magnetic resonance (CMR) imaging incorporates both functional and morphological characterization of the heart to determine presence, acuteness, and severity of ischemic heart disease by evaluating myocardial wall motion and function, the presence and extent of myocardial edema, ischemia, and scarring. Currently established clinical protocols have already demonstrated their diagnostic and prognostic value. Nevertheless, there are emerging imaging technologies that provide additional information based on advanced quantification of imaging biomarkers and improved diagnostic accuracy, therefore potentially allowing reduction or avoidance of contrast and/or stressor agents. The aim of this review is to summarize the current state of the art of CMR imaging for ischemic heart disease and to provide insights into promising future developments.
Collapse
Affiliation(s)
- Tilman Emrich
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz; Langenbeckstraße 1, 55131, Mainz, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Langenbeckstraße 1, 55131, Mainz, Germany. .,Department of Radiology and Radiological Science, Medical University of South Carolina, 25 Courtenay Drive, Charleston, SC, 29425, USA.
| | - Moritz Halfmann
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz; Langenbeckstraße 1, 55131, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - U Joseph Schoepf
- Department of Radiology and Radiological Science, Medical University of South Carolina, 25 Courtenay Drive, Charleston, SC, 29425, USA
| | - Karl-Friedrich Kreitner
- Department of Diagnostic and Interventional Radiology, University Medical Center, Mainz; Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|