1
|
Ahn JC, Rattan P, Starlinger P, Juanola A, Moreta MJ, Colmenero J, Aqel B, Keaveny AP, Mullan AF, Liu K, Attia ZI, Allen AM, Friedman PA, Shah VH, Noseworthy PA, Heimbach JK, Kamath PS, Gines P, Simonetto DA. AI-Cirrhosis-ECG (ACE) score for predicting decompensation and liver outcomes. JHEP Rep 2025; 7:101356. [PMID: 40276480 PMCID: PMC12018547 DOI: 10.1016/j.jhepr.2025.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 04/26/2025] Open
Abstract
Background & Aims Accurate prediction of disease severity and prognosis are challenging in patients with cirrhosis. We evaluated whether the deep learning-based AI-Cirrhosis-ECG (ACE) score could detect hepatic decompensation and predict clinical outcomes in cirrhosis. Methods We analyzed 2,166 ECGs from 472 patients in a retrospective Mayo Clinic cohort, 420 patients in a prospective Mayo transplant cohort, and 341 patients in an external validation cohort from Hospital Clínic de Barcelona. The ACE score's performance was assessed using receiver-operating characteristic analysis for decompensation detection and competing risks Cox regression for outcome prediction. Results The ACE score showed high accuracy in detecting hepatic decompensation (area under the curve 0.933, 95% CI: 0.923-0.942) with 88.0% sensitivity and 84.3% specificity at an optimal threshold of 0.25. In multivariable analysis, each 0.1-point increase in ACE score was independently associated with increased risk of liver-related death (hazard ratio [HR] 1.44, 95% CI 1.32-1.58, p <0.001). Adding ACE to model for end-stage liver disease-sodium significantly improved prediction of adverse outcomes across all cohorts (c-statistics: retrospective cohort 0.903 vs. 0.844; prospective cohort 0.779 vs. 0.735; external validation 0.744 vs. 0.732; all p <0.001). Conclusions The ACE score accurately identifies hepatic decompensation and independently predicts liver-related outcomes in cirrhosis. This non-invasive tool enhances current prognostic models and may improve risk stratification in cirrhosis management. Impact and implications This study demonstrates the potential of artificial intelligence to enhance prognostication in liver disease, addressing the critical need for improved risk stratification in cirrhosis management. The AI-Cirrhosis-ECG (ACE) score, derived from widely available ECGs, shows promise as a non-invasive tool for detecting hepatic decompensation and predicting liver-related outcomes, which could significantly impact clinical decision-making and resource allocation in hepatology. These findings are particularly important for hepatologists, transplant surgeons, and patients with cirrhosis, as they offer a novel approach to complement existing prognostic models such as model for end-stage liver disease-sodium. In practical terms, the ACE score could be integrated into routine clinical assessments to provide more accurate risk predictions, potentially improving the timing of interventions, optimizing transplant listing decisions, and ultimately enhancing patient outcomes. However, further validation in diverse populations and integration with other established predictors is necessary before widespread clinical implementation.
Collapse
Affiliation(s)
- Joseph C. Ahn
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| | - Puru Rattan
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| | - Patrick Starlinger
- Department of Surgery, Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, MA, USA
| | - Adrià Juanola
- Liver Unit, Hospital Clínic, Barcelona, Catalonia, Spain
- Institut d’Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Maria José Moreta
- Liver Unit, Hospital Clínic, Barcelona, Catalonia, Spain
- Institut d’Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jordi Colmenero
- Liver Unit, Hospital Clínic, Barcelona, Catalonia, Spain
- Institut d’Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Faculty of Medicine and Health Sciences, Barcelona, Catalonia, Spain
| | - Bashar Aqel
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Aidan F. Mullan
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Kan Liu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zachi I. Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alina M. Allen
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| | - Paul A. Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Vijay H. Shah
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| | | | - Julie K. Heimbach
- Department of Surgery, Division of Transplantation Surgery, Mayo Clinic, Rochester, MN, USA
| | - Patrick S. Kamath
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| | - Pere Gines
- Liver Unit, Hospital Clínic, Barcelona, Catalonia, Spain
- Institut d’Investigacions Biomèdiques August Pi-Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Faculty of Medicine and Health Sciences, Barcelona, Catalonia, Spain
| | - Douglas A. Simonetto
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MA, USA
| |
Collapse
|
2
|
Presacan O, Dorobanţiu A, Isaksen JL, Willi T, Graff C, Riegler MA, Sridhar AR, Kanters JK, Thambawita V. Evaluating the feasibility of 12-lead electrocardiogram reconstruction from limited leads using deep learning. COMMUNICATIONS MEDICINE 2025; 5:139. [PMID: 40281134 PMCID: PMC12032410 DOI: 10.1038/s43856-025-00814-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Wearables with integrated electrocardiogram (ECG) acquisition have made single-lead ECGs widely accessible to patients and consumers. However, the 12-lead ECG remains the gold standard for most clinical cardiac assessments. In this study, we developed a neural network to reconstruct 12-lead ECGs from single-lead and dual-lead ECGs, and evaluated the mathematical accuracy. METHODS We used lead I or leads I and II from 9514 individuals from the Physikalisch-Technische Bundesanstalt (PTB-XL) cohort and a generative adversarial network, with the aim of recreating the missing leads from the 12-lead ECG. ECGs were divided into training, validation, and testing (10%). Original and recreated leads were measured with a commercially available algorithm. Differences in means and variances were assessed with Student's t-tests and F-tests, respectively. Calibration and bias were assessed with Bland-Altman plots. Inter-lead correlations were compared in original and recreated ECGs. RESULTS The variability of precordial ECG amplitudes is significantly reduced in recreated ECGs compared to real ECGs (all p < 0.05), indicating regression-to-the-mean. Amplitude averages are recreated with bias (p < 0.05 for most leads). Reconstruction errors depend on the real amplitudes, suggesting regression-to-the-mean (R2 between target and error in R-peak amplitude in lead V3: 0.92). The relations between lead markers have a similar slope but are much stronger due to reduced variance (R-peak amplitude R2 between leads I and V3, real ECGs: 0.04, recreated ECGs: 0.49). Using two leads does not significantly improve 12-lead recreation. CONCLUSIONS AI-based 12-lead ECG reconstruction results in a regression-to-the-mean effect rather than personalized output, rendering it unsuitable for clinical use.
Collapse
Affiliation(s)
| | | | | | - Tobias Willi
- KTH Royal Institute of Technology, 11428, Stockholm, Sweden
| | - Claus Graff
- Aalborg University, 9220, Aalborg Ø, Denmark
| | - Michael A Riegler
- Simula Research Laboratory, Kristian Augusts gate 23, 0164, Oslo, Norway
| | - Arun R Sridhar
- Pulse Heart Institute, Multicare Health System, Tacoma, WA, USA
| | - Jørgen K Kanters
- University of Copenhagen, 2200, Copenhagen N, Denmark
- University of California, San Francisco, USA
| | | |
Collapse
|
3
|
Wiedmann F, Schmidt C. Precision medicine in the management of cardiac arrhythmias. Herz 2025; 50:88-95. [PMID: 40056164 DOI: 10.1007/s00059-025-05298-x] [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] [Accepted: 02/03/2025] [Indexed: 03/10/2025]
Abstract
Precision medicine in cardiac electrophysiology tailors diagnosis, treatment, and prevention by integrating genetic, environmental, and lifestyle factors. Unlike traditional, generalized strategies, precision medicine focuses on individual patient characteristics to enhance care. Significant progress has been made, especially in managing channelopathies, where genetic insights now already drive personalized therapies. Identifying specific mutations has clarified molecular mechanisms and enabled targeted interventions, improving outcomes in conditions such as long QT syndrome. The integration of big data from clinical records, omics datasets, and biosignals from devices such as cardiac implantable electronic devices (CIEDs) or wearables may be on the verge of revolutionizing the diagnosis of cardiac arrhythmias once again. Progress is also expected in the field of human-induced pluripotent stem cells (hiPSCs) and in silico modeling, which may overcome the limitations of traditional expression systems for the functional evaluation of patient-specific mutations. Genome-wide association studies (GWAS) and polygenic risk scores (PRS) provide deeper insights into complex arrhythmogenic disorders, aiding in risk stratification and targeted treatment strategies. Finally, emerging technologies such as CRISPR/Cas9 promise gene editing for inherited and acquired arrhythmias. In summary, precision medicine offers the potential for individualized treatment of cardiac arrhythmias.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, Medical University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
4
|
Disrud LW, Swain WH, Davison H, Gosse T, Kubler MM, Harmon DM, Friedman PA, Noseworthy PA, Kashou AH. A Pilot Study of the Home-Based 12-Lead Electrocardiogram in Clinical Practice. Mayo Clin Proc Innov Qual Outcomes 2025; 9:100598. [PMID: 40092494 PMCID: PMC11909748 DOI: 10.1016/j.mayocpiqo.2025.100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 03/19/2025] Open
Abstract
Telehealth consultation with a physician can be an attractive option for eligible patients. In this pilot study, we evaluate the feasibility and efficiency of an FDA approved 12-lead electrocardiogram (ECG) device, Smeartheart, that can be used remotely in the patients' home before telehealth appointments with a cardiac electrophysiologist. We scheduled a phone call with 10 patients who used this device as part of their care. Eight patients were able to obtain a diagnostic quality ECG. Telephone call appointments with ECG technicians took a median of 51 minutes, and it took patients an average of 2.2 attempts to record a usable ECG. We also identified barriers to the use of the Smartheart device, including internet accessibility, training material, patient functional status, and motion artifact that may inform more widespread study and utilization of remote-recorded 12-lead ECGs. We conclude that the Smartheart device may have clinical use with remote use in routine clinical care, although the best use of this technology requires further study.
Collapse
Affiliation(s)
- Levi W. Disrud
- Department of Cardiovascular Research, Mayo Clinic, Rochester, MN
| | | | - Halley Davison
- Department of Cardiovascular Research, Mayo Clinic, Rochester, MN
| | - Tara Gosse
- Department of Transformational/Digital Strategy, Mayo Clinic, Rochester, MN
| | | | - David M. Harmon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Paul A. Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | | |
Collapse
|
5
|
Toba S, Mitani Y, Sugitani Y, Ohashi H, Sawada H, Takeoka M, Tsuboya N, Ohya K, Yodoya N, Yamasaki T, Nakayama Y, Ito H, Hirayama M, Takao M. Deep learning-based analysis of 12-lead electrocardiograms in school-age children: a proof of concept study. Front Cardiovasc Med 2025; 12:1471989. [PMID: 40109297 PMCID: PMC11919894 DOI: 10.3389/fcvm.2025.1471989] [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: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction The diagnostic performance of automated analysis of electrocardiograms for screening children with pediatric heart diseases at risk of sudden cardiac death is unknown. In this study, we aimed to develop and validate a deep learning-based model for automated analysis of ECGs in children. Methods Wave data of 12-lead electrocardiograms were transformed into a tensor sizing 2 × 12 × 400 using signal processing methods. A deep learning-based model to classify abnormal electrocardiograms based on age, sex, and the transformed wave data was developed using electrocardiograms performed in patients at the age of 6-18 years during 2003-2006 at a tertiary referral hospital in Japan. Eighty-three percent of the patients were assigned to a training group, and 17% to a test group. The diagnostic performance of the model and a conventional algorithm (ECAPS12C, Nihon Kohden, Japan) for classifying abnormal electrocardiograms were evaluated using the cross-tabulation, McNemar's test, and decision curve analysis. Results We included 1,842 ECGs performed in 1,062 patients in this study, and 310 electrocardiograms performed in 177 patients were included in the test group. The specificity of the deep learning-based model for detecting abnormal electrocardiograms was not significantly different from that of the conventional algorithm. For detecting electrocardiograms with ST-T abnormality, complete right bundle branch block, QRS axis abnormality, left ventricular hypertrophy, incomplete right bundle branch block, WPW syndrome, supraventricular tachyarrhythmia, and Brugada-type electrocardiograms, the specificity of the deep learning-based model was higher than that of the conventional algorithm at the same sensitivity. Conclusions The present new deep learning-based method of screening for abnormal electrocardiograms in children showed at least a similar diagnostic performance compared to that of a conventional algorithm. Further studies are warranted to develop an automated analysis of electrocardiograms in school-age children.
Collapse
Affiliation(s)
- Shuhei Toba
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Yoshihide Mitani
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Yusuke Sugitani
- Department of Clinical Engineering, Mie University Hospital, Tsu, Mie, Japan
- Department of Electrical and Electronic Engineering, Mie University, Tsu, Mie, Japan
| | - Hiroyuki Ohashi
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hirofumi Sawada
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Mami Takeoka
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Naoki Tsuboya
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Kazunobu Ohya
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Noriko Yodoya
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Takato Yamasaki
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Yuki Nakayama
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hisato Ito
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Masahiro Hirayama
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Motoshi Takao
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
6
|
Singh M, Friedman PA, Gulati R, El Sabbagh A, Lewis BR, Kanwar A, Raphael CE, Al-Hijji MA, Attia ZI, Behfar A, Kirkland JL. Role of Biological Age in the Determination of Long-Term Cause-Specific Death Following Percutaneous Coronary Interventions. J Am Heart Assoc 2025; 14:e036876. [PMID: 40008514 DOI: 10.1161/jaha.124.036876] [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: 05/31/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND We tested whether biologic age, as estimated by deficits, functional impairments, or Age-Gap or their combination, provide improved estimation of cause-specific death as compared with chronological age. METHODS Cardiovascular and noncardiovascular deficits, functional impairments, and Age-Gap were prospectively collected in 535 patients aged ≥55 years undergoing percutaneous coronary interventions between August 1, 2014, and March 31, 2018. Age-Gap was calculated as the difference between chronological age and age estimated by artificial intelligence ECG using a convolutional neural network. The full biological age model included deficits, functional impairments, and Age-Gap >2 SD. A multivariable reduced model with the least number of variables was also created to provide a comparable C index to the full model. RESULTS The average chronological age was 72.1±9.5 years, and there were 68% of men. During a median follow-up of 2.61 years, 124 (23%) patients died. There was a modest correlation between Age-Gap and biological age (r=0.28 [95% CI, 0.20-0.35]; P<0.001). When modeled with chronologic age as a covariate, Age-Gap predicted all-cause (hazard ratio [HR], 1.07 [95% CI, 1.04-1.10]; P<0.001) and cardiovascular (HR, 1.07 [95% CI, 1.04-1.11]; P<0.001) mortality. As compared with chronological age, the full biological age model noted significant improvement in the prediction of long-term overall (95% CI, 0.65-0.78), cardiovascular (95% CI, 0.69-0.77), and noncardiovascular (95% CI, 0.55-0.86) mortality. In the reduced models, most prognostic information for noncardiovascular mortality (C index: 0.79) was obtained by subjective difficulty in performing tasks, whereas the deficit-based estimation predicted cardiovascular mortality (C index: 0.72). CONCLUSIONS Estimated biological age from deficits and functional impairments was superior to chronological age in predicting long-term cause-specific mortality following percutaneous coronary interventions.
Collapse
Affiliation(s)
- Mandeep Singh
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | - Paul A Friedman
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | - Rajiv Gulati
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | | | | | - Amrit Kanwar
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | - Claire E Raphael
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | | | - Zachi I Attia
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | - Atta Behfar
- Department of Cardiovascular Diseases Mayo Clinic Rochester MN USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| |
Collapse
|
7
|
Ribeiro AH, Ribeiro ALP. AI-ECG and prediction of new atrial fibrillation: when the heart tells the age. Eur Heart J 2025; 46:853-855. [PMID: 39657904 DOI: 10.1093/eurheartj/ehae809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Affiliation(s)
- Antonio H Ribeiro
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Antonio Luiz P Ribeiro
- Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena 110, 1° Andar, Ala Sul, Sala 107, Belo Horizonte MG 30130-100, Brazil
- Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
8
|
Cho S, Eom S, Kim D, Kim TH, Uhm JS, Pak HN, Lee MH, Yang PS, Lee E, Attia ZI, Friedman PA, You SC, Yu HT, Joung B. Artificial intelligence-derived electrocardiographic aging and risk of atrial fibrillation: a multi-national study. Eur Heart J 2025; 46:839-852. [PMID: 39626169 DOI: 10.1093/eurheartj/ehae790] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/26/2024] [Accepted: 10/31/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND AND AIMS Artificial intelligence (AI) algorithms in 12-lead electrocardiogram (ECG) provides promising age prediction methods. This study investigated whether the discrepancy between ECG-derived AI-predicted age (AI-ECG age) and chronological age, termed electrocardiographic aging (ECG aging), is associated with atrial fibrillation (AF) risk. METHODS An AI-ECG age prediction model was developed using a large-scale dataset (1 533 042 ECGs from 689 639 participants) and validated with six independent and multi-national datasets (737 133 ECGs from 330 794 participants). The AI-ECG age gap was calculated across two South Korean cohorts [mean (standard deviation) follow-up: 4.1 (4.3) years for 111 483 participants and 6.1 (3.8) years for 37 517 participants], one UK cohort [3.0 (1.6) years; 40 973 participants], and one US cohort [12.9 (8.6) years; 90 639 participants]. Participants were classified into two groups: normal group (age gap < 7 years) and ECG-aged group (age gap ≥ 7 years). The predictive capability of ECG aging for new- and early-onset AF risk was assessed. RESULTS The mean AI-ECG ages were 51.9 (16.2), 47.4 (12.5), 68.4 (7.8), and 56.7 (14.6) years with age gaps of .0 (6.8), -.1 (6.0), 4.7 (8.7), and -1.4 (8.9) years in the two South Korean, UK, and US cohorts, respectively. In the ECG-aged group, increased risks of new-onset AF were observed with hazard ratios (95% confidence intervals) of 2.50 (2.24-2.78), 1.89 (1.46-2.43), 1.90 (1.55-2.33), and 1.76 (1.67-1.86) in the two South Korean, UK, and US cohorts, respectively. For early-onset AF, odds ratios were 2.89 (2.47-3.37), 1.94 (1.39-2.70), 1.58 (1.06-2.35), and 1.79 (1.62-1.97) in these cohorts compared with the normal group. CONCLUSIONS The AI-derived ECG aging was associated with the risk of new- and early-onset AF, suggesting its potential utility to identify individuals for AF prevention across diverse populations.
Collapse
Affiliation(s)
- Seunghoon Cho
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Sujeong Eom
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Institute for Innovation in Digital Healthcare, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Daehoon Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Jae-Sun Uhm
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Moon-Hyoung Lee
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Pil-Sung Yang
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Eunjung Lee
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Seng Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Institute for Innovation in Digital Healthcare, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Hee Tae Yu
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Boyoung Joung
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
9
|
Udompap P, Liu K, Attia IZ, Canning RE, Benson JT, Therneau TM, Noseworthy PA, Friedman PA, Rattan P, Ahn JC, Simonetto DA, Shah VH, Kamath PS, Allen AM. Performance of AI-Enabled Electrocardiogram in the Prediction of Metabolic Dysfunction-Associated Steatotic Liver Disease. Clin Gastroenterol Hepatol 2025; 23:574-582.e3. [PMID: 39209186 DOI: 10.1016/j.cgh.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Accessible noninvasive screening tools for metabolic dysfunction-associated steatotic liver disease (MASLD) are needed. We aim to explore the performance of a deep learning-based artificial intelligence (AI) model in distinguishing the presence of MASLD using 12-lead electrocardiogram (ECG). METHODS This is a retrospective study of adults diagnosed with MASLD in Olmsted County, Minnesota, between 1996 and 2019. Both cases and controls had ECGs performed within 6 years before and 1 year after study entry. An AI-based ECG model using a convolutional neural network was trained, validated, and tested in 70%, 10%, and 20% of the cohort, respectively. External validation was performed in an independent cohort from Mayo Clinic Enterprise. The primary outcome was the performance of ECG to identify MASLD, alone or when added to clinical parameters. RESULTS A total of 3468 MASLD cases and 25,407 controls were identified. The AI-ECG model predicted the presence of MASLD with an area under the curve (AUC) of 0.69 (original cohort) and 0.62 (validation cohort). The performance was similar or superior to age- and sex-adjusted models using body mass index (AUC, 0.71), presence of diabetes, hypertension or hyperlipidemia (AUC, 0.68), or diabetes alone (AUC, 0.66). The model combining ECG, age, sex, body mass index, diabetes, and alanine aminotransferase had the highest AUC: 0.76 (original) and 0.72 (validation). CONCLUSIONS This is a proof-of-concept study that an AI-based ECG model can detect MASLD with a comparable or superior performance as compared with the models using a single clinical parameter but not superior to the combination of clinical parameters. ECG can serve as another screening tool for MASLD in the nonhepatology space.
Collapse
Affiliation(s)
- Prowpanga Udompap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kan Liu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Itzhak Zachi Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Rachel E Canning
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Joanne T Benson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Terry M Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Peter A Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Puru Rattan
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joseph C Ahn
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Douglas A Simonetto
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Patrick S Kamath
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
10
|
de Melo JF, Mangold KE, Debertin J, Rosenbaum A, Bois JP, Attia ZI, Friedman PA, Deshmukh AJ, Kapa S, Cooper LT, Abou Ezzeddine OF, Siontis KC. Detection of cardiac sarcoidosis with the artificial intelligence-enhanced electrocardiogram. Heart Rhythm 2025; 22:859-861. [PMID: 39127231 DOI: 10.1016/j.hrthm.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Affiliation(s)
- Jose F de Melo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kathryn E Mangold
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Julia Debertin
- Mayo Clinic Alix School of Medicine, Rochester, Minnesota
| | - Andrew Rosenbaum
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - John P Bois
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Zachi I Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Suraj Kapa
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, Florida
| | | | | |
Collapse
|
11
|
Holt DB, El-Bokl A, Stromberg D, Taylor MD. Role of Artificial Intelligence in Congenital Heart Disease and Interventions. JOURNAL OF THE SOCIETY FOR CARDIOVASCULAR ANGIOGRAPHY & INTERVENTIONS 2025; 4:102567. [PMID: 40230672 PMCID: PMC11993855 DOI: 10.1016/j.jscai.2025.102567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 04/16/2025]
Abstract
Artificial intelligence has promising impact on patients with congenital heart disease, a vulnerable population with life-long health care needs and, often, a substantially higher risk of death than the general population. This review explores the role artificial intelligence has had on cardiac imaging, electrophysiology, interventional procedures, and intensive care monitoring as it relates to children and adults with congenital heart disease. Machine learning and deep learning algorithms have enhanced not only imaging segmentation and processing but also diagnostic accuracy namely reducing interobserver variability. This has a meaningful impact in complex congenital heart disease improving anatomic diagnosis, assessment of cardiac function, and predicting long-term outcomes. Image processing has benefited procedural planning for interventional cardiology, allowing for a higher quality and density of information to be extracted from the same imaging modalities. In electrophysiology, deep learning models have enhanced the diagnostic potential of electrocardiograms, detecting subtle yet meaningful variation in signals that enable early diagnosis of cardiac dysfunction, risk stratification of mortality, and more accurate diagnosis and prediction of arrhythmias. In the congenital heart disease population, this has the potential for meaningful prolongation of life. Postoperative care in the cardiac intensive care unit is a data-rich environment that is often overwhelming. Detection of subtle data trends in this environment for early detection of morbidity is a ripe avenue for artificial intelligence algorithms to be used. Examples like early detection of catheter-induced thrombosis have already been published. Despite their great promise, artificial intelligence algorithms are still limited by hurdles such as data standardization, algorithm validation, drift, and explainability.
Collapse
Affiliation(s)
- Dudley Byron Holt
- Department of Pediatrics, University of Texas at Austin Dell Medical School, Austin, Texas
- Texas Center for Pediatric and Congenital Heart Disease, Dell Children’s Medical Center, Austin, Texas
| | - Amr El-Bokl
- Department of Pediatrics, University of Texas at Austin Dell Medical School, Austin, Texas
- Texas Center for Pediatric and Congenital Heart Disease, Dell Children’s Medical Center, Austin, Texas
| | - Daniel Stromberg
- Department of Pediatrics, University of Texas at Austin Dell Medical School, Austin, Texas
- Texas Center for Pediatric and Congenital Heart Disease, Dell Children’s Medical Center, Austin, Texas
| | - Michael D. Taylor
- Department of Pediatrics, University of Texas at Austin Dell Medical School, Austin, Texas
- Texas Center for Pediatric and Congenital Heart Disease, Dell Children’s Medical Center, Austin, Texas
| |
Collapse
|
12
|
Sau A, Sieliwonczyk E, Patlatzoglou K, Pastika L, McGurk KA, Ribeiro AH, Ribeiro ALP, Ho JE, Peters NS, Ware JS, Tayal U, Kramer DB, Waks JW, Ng FS. Artificial intelligence-enhanced electrocardiography for the identification of a sex-related cardiovascular risk continuum: a retrospective cohort study. Lancet Digit Health 2025; 7:e184-e194. [PMID: 40015763 DOI: 10.1016/j.landig.2024.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 11/23/2024] [Accepted: 12/06/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Females are typically underserved in cardiovascular medicine. The use of sex as a dichotomous variable for risk stratification fails to capture the heterogeneity of risk within each sex. We aimed to develop an artificial intelligence-enhanced electrocardiography (AI-ECG) model to investigate sex-specific cardiovascular risk. METHODS In this retrospective cohort study, we trained a convolutional neural network to classify sex using the 12-lead electrocardiogram (ECG). The Beth Israel Deaconess Medical Center (BIDMC) secondary care dataset, comprising data from individuals who had clinically indicated ECGs performed in a hospital setting in Boston, MA, USA collected between May, 2000, and March, 2023, was the derivation cohort (1 163 401 ECGs). 50% of this dataset was used for model training, 10% for validation, and 40% for testing. External validation was performed using the UK Biobank cohort, comprising data from volunteers aged 40-69 years at the time of enrolment in 2006-10 (42 386 ECGs). We examined the difference between AI-ECG-predicted sex (continuous) and biological sex (dichotomous), termed sex discordance score. FINDINGS AI-ECG accurately identified sex (area under the receiver operating characteristic 0·943 [95% CI 0·942-0·943] for BIDMC and 0·971 [0·969-0·972] for the UK Biobank). In BIDMC outpatients with normal ECGs, an increased sex discordance score was associated with covariate-adjusted increased risk of cardiovascular death in females (hazard ratio [HR] 1·78 [95% CI 1·18-2·70], p=0·006) but not males (1·00 [0·63-1·58], p=0·996). In the UK Biobank cohort, the same pattern was seen (HR 1·33 [95% CI 1·06-1·68] for females, p=0·015; 0·98 [0·80-1·20] for males, p=0·854). Females with a higher sex discordance score were more likely to have future heart failure or myocardial infarction in the BIDMC cohort and had more male cardiac (increased left ventricular mass and chamber volumes) and non-cardiac phenotypes (increased muscle mass and reduced body fat percentage) in both cohorts. INTERPRETATION Sex discordance score is a novel AI-ECG biomarker capable of identifying females with disproportionately elevated cardiovascular risk. AI-ECG has the potential to identify female patients who could benefit from enhanced risk factor modification or surveillance. FUNDING British Heart Foundation.
Collapse
Affiliation(s)
- Arunashis Sau
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Imperial College Healthcare NHS Trust, London, UK.
| | - Ewa Sieliwonczyk
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK; University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | | | - Libor Pastika
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Antônio H Ribeiro
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, and Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jennifer E Ho
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicholas S Peters
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Imperial College Healthcare NHS Trust, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Upasana Tayal
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Royal Brompton & Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Daniel B Kramer
- National Heart and Lung Institute, Imperial College London, London, UK; Richard A and Susan F Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan W Waks
- Harvard-Thorndike Electrophysiology Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, London, UK; Department of Cardiology, Imperial College Healthcare NHS Trust, London, UK; Department of Cardiology, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
13
|
Adedinsewo DA, Shufelt CL, Carter RE. Unmasking hidden risk: an AI approach to improve cardiovascular risk assessment in females. Lancet Digit Health 2025; 7:e170-e171. [PMID: 40015761 DOI: 10.1016/j.landig.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 03/01/2025]
Affiliation(s)
| | - Chrisandra L Shufelt
- Women's Health Research Center, Mayo Clinic, Jacksonville, FL 32224, USA; Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rickey E Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
14
|
Wu Z, Guo C. Deep learning and electrocardiography: systematic review of current techniques in cardiovascular disease diagnosis and management. Biomed Eng Online 2025; 24:23. [PMID: 39988715 PMCID: PMC11847366 DOI: 10.1186/s12938-025-01349-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
This paper reviews the recent advancements in the application of deep learning combined with electrocardiography (ECG) within the domain of cardiovascular diseases, systematically examining 198 high-quality publications. Through meticulous categorization and hierarchical segmentation, it provides an exhaustive depiction of the current landscape across various cardiovascular ailments. Our study aspires to furnish interested readers with a comprehensive guide, thereby igniting enthusiasm for further, in-depth exploration and research in this realm.
Collapse
Affiliation(s)
- Zhenyan Wu
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Saran Khalid M, Shahid Quraishi I, Wasim Nawaz M, Sajjad H, Yaseen H, Mehmood A, Mahboob Ur Rahman M, Abbasi QH. A low-cost PPG sensor-based empirical study on healthy aging based on changes in PPG morphology. Physiol Meas 2025; 13:025005. [PMID: 39706154 DOI: 10.1088/1361-6579/ada246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/20/2024] [Indexed: 12/23/2024]
Abstract
Objective. We study the changes in morphology of the photoplethysmography (PPG) signals-acquired from a select group of South Asian origin-through a low-cost PPG sensor, and correlate it with healthy aging which allows us to reliably estimate the vascular age and chronological age of a healthy person as well as the age group he/she belongs to.Approach. Raw infrared PPG data is collected from the finger-tip of 173 apparently healthy subjects, aged 3-61 years, via a non-invasive low-cost MAX30102 PPG sensor. In addition, the following metadata is recorded for each subject: age, gender, height, weight, family history of cardiac disease, smoking history, vitals (heart rate and SpO2). The raw PPG data is conditioned and 62 features are then extracted based upon the first four PPG derivatives. Then, correlation-based feature-ranking is performed which retains 26 most important features. Finally, the feature set is fed to three machine learning classifiers, i.e. logistic regression, random forest, eXtreme Gradient Boosting (XGBoost), and two shallow neural networks: a feedforward neural network and a convolutional neural network.Main results. For the age group classification problem, the ensemble method XGboost stands out with an accuracy of 99% for both binary classification (3-20 years vs. 20+ years) and three-class classification (3-18 years, 18-23 years, 23+ years). For the vascular/chronological age prediction problem, the ensemble random forest method stands out with a mean absolute error of 6.97 years.Significance. The results demonstrate that PPG is indeed a promising (i.e. low-cost, non-invasive) biomarker to study the healthy aging phenomenon.
Collapse
Affiliation(s)
- Muhammad Saran Khalid
- Electrical engineering department, Information Technology University, Lahore, Pakistan
| | | | | | - Hadia Sajjad
- Electrical engineering department, Information Technology University, Lahore, Pakistan
| | - Hira Yaseen
- Electrical engineering department, Information Technology University, Lahore, Pakistan
| | - Ahsan Mehmood
- Electrical engineering department, Information Technology University, Lahore, Pakistan
| | - M Mahboob Ur Rahman
- Electrical engineering department, Information Technology University, Lahore, Pakistan
| | - Qammer H Abbasi
- James Watt School of Engineering, University of Glasgow, Glasgow, United Kingdom
- Artificial Intelligence Research Centre, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
16
|
Liu CM, Kuo MJ, Kuo CY, Wu IC, Chen PF, Hsu WT, Liao LL, Chen SA, Tsao HM, Liu CL, Hu YF. Reclassification of the conventional risk assessment for aging-related diseases by electrocardiogram-enabled biological age. NPJ AGING 2025; 11:7. [PMID: 39915530 PMCID: PMC11802786 DOI: 10.1038/s41514-025-00198-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025]
Abstract
An artificial intelligence (AI)-enabled electrocardiogram (ECG) model has been developed in a healthy adult population to predict ECG biological age (ECG-BA). This ECG-BA exhibited a robust correlation with chronological age (CA) in healthy adults and additionally significantly enhanced the prediction of aging-related diseases' onset in adults with subclinical diseases. The model showed particularly strong predictive power for cardiovascular and non-cardiovascular diseases such as stroke, coronary artery disease, peripheral arterial occlusive disease, myocardial infarction, Alzheimer's disease, osteoarthritis, and cancers. When combined with CA, ECG-BA improved diagnostic accuracy and risk classification by 21% over using CA alone, notably offering the greatest improvements in cancer prediction. The net reclassification improvement significantly reduced misclassification rates for disease onset predictions. This comprehensive study validates ECG-BA as an effective supplement to CA, advancing the precision of risk assessments for aging-related conditions and suggesting broad implications for enhancing preventive healthcare strategies, potentially leading to better patient outcomes.
Collapse
Affiliation(s)
- Chih-Min Liu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Jen Kuo
- Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chin-Yu Kuo
- Department of Industrial Engineering and Management, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - I-Chien Wu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Fen Chen
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Wan-Ting Hsu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Lien Liao
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
- National Chung Hsing University, Taichung, Taiwan
| | - Hsuan-Ming Tsao
- Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Division of Cardiology, Department of Medicine, National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan.
| | - Chien-Liang Liu
- Department of Industrial Engineering and Management, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| | - Yu-Feng Hu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
17
|
Griné M, Guerreiro C, Moscoso Costa F, Nobre Menezes M, Ladeiras-Lopes R, Ferreira D, Oliveira-Santos M. Digital health in cardiovascular medicine: An overview of key applications and clinical impact by the Portuguese Society of Cardiology Study Group on Digital Health. Rev Port Cardiol 2025; 44:107-119. [PMID: 39393635 DOI: 10.1016/j.repc.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/13/2024] Open
Abstract
Digital health interventions including telehealth, mobile health, artificial intelligence, big data, robotics, extended reality, computational and high-fidelity bench simulations are an integral part of the path toward precision medicine. Current applications encompass risk factor modification, chronic disease management, clinical decision support, diagnostics interpretation, preprocedural planning, evidence generation, education, and training. Despite the acknowledged potential, their development and implementation have faced several challenges and constraints, meaning few digital health tools have reached daily clinical practice. As a result, the Portuguese Society of Cardiology Study Group on Digital Health set out to outline the main digital health applications, address some of the roadblocks hampering large-scale deployment, and discuss future directions in support of cardiovascular health at large.
Collapse
Affiliation(s)
- Mafalda Griné
- Serviço de Cardiologia, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal.
| | - Cláudio Guerreiro
- Serviço de Cardiologia, Centro Hospitalar de Vila Nova de Gaia, Vila Nova de Gaia, Portugal
| | | | - Miguel Nobre Menezes
- Structural and Coronary Heart Disease Unit, Cardiovascular Center of the University of Lisbon, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Serviço de Cardiologia, Departamento de Coração e Vasos, CHULN Hospital de Santa Maria, Lisboa, Portugal
| | - Ricardo Ladeiras-Lopes
- UnIC@RISE, Cardiovascular Research and Development Center, Department of Surgery and Physiology, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; Hospital da Luz, Lisboa, Portugal
| | - Daniel Ferreira
- Serviço de Medicina Intensiva, Hospital da Luz, Lisboa, Portugal; Hospital da Luz Digital, Lisboa, Portugal
| | - Manuel Oliveira-Santos
- Serviço de Cardiologia, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, Coimbra, Portugal; Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
18
|
Nakayama M, Yagi R, Goto S. Deep Learning Applications in 12-lead Electrocardiogram and Echocardiogram. JMA J 2025; 8:102-112. [PMID: 39926090 PMCID: PMC11799486 DOI: 10.31662/jmaj.2024-0195] [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: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 02/11/2025] Open
Abstract
Artificial intelligence (AI), empowered by advances in deep learning technology, has demonstrated its capabilities in the medical field to automate tedious tasks that are otherwise performed by humans or to detect or predict diseases with higher accuracy compared with experts. Given the ability to take complex multidimensional data as input, AI models have primarily been applied to complex medical imaging and time-series data. Another prominent strength of AI applications is its large scalability. The field of cardiovascular medicine uses various noninvasive and accessible metrics that produce a large amount of complex multidimensional data, such as electrocardiograms (ECGs) and echocardiograms. AI models can increase the utility of such modalities. Simple automation of conventional tasks using AI models provides significant opportunities for cost reduction and capacity expansion. The ability to improve disease detection or prediction at scale may provide novel opportunities for disease screening, enabling early intervention in asymptomatic patients. For example, AI-enabled pipelines can accurately identify cardiomyopathies and congenital heart diseases from a single ECG or echocardiogram recording. The detection of these diseases using the conventional approach usually requires complicated diagnostic strategies or expensive tests. Therefore, underdiagnosis is a huge problem. Using AI models to screen these diseases will provide opportunities for reducing missed cases. The utility of AI models in the medical field is not limited to the development of clinically useful models. Recent research has shown the promise of AI models in mechanism research by combining them with genetic and structural analyses. In this review, we provide an update on the current achievements of the innovative AI application for ECG and echocardiogram and provide insights into the future direction of AI in cardiovascular care and research settings.
Collapse
Affiliation(s)
- Masamitsu Nakayama
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - Ryuichiro Yagi
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - Shinichi Goto
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA
- Division of General Internal Medicine & Family Medicine, Department of General and Acute Medicine, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
19
|
Friedman SF, Khurshid S, Venn RA, Wang X, Diamant N, Di Achille P, Weng LC, Choi SH, Reeder C, Pirruccello JP, Singh P, Lau ES, Philippakis A, Anderson CD, Maddah M, Batra P, Ellinor PT, Ho JE, Lubitz SA. Unsupervised deep learning of electrocardiograms enables scalable human disease profiling. NPJ Digit Med 2025; 8:23. [PMID: 39799251 PMCID: PMC11724961 DOI: 10.1038/s41746-024-01418-9] [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: 07/29/2024] [Accepted: 12/21/2024] [Indexed: 01/15/2025] Open
Abstract
The 12-lead electrocardiogram (ECG) is inexpensive and widely available. Whether conditions across the human disease landscape can be detected using the ECG is unclear. We developed a deep learning denoising autoencoder and systematically evaluated associations between ECG encodings and ~1,600 Phecode-based diseases in three datasets separate from model development, and meta-analyzed the results. The latent space ECG model identified associations with 645 prevalent and 606 incident Phecodes. Associations were most enriched in the circulatory (n = 140, 82% of category-specific Phecodes), respiratory (n = 53, 62%) and endocrine/metabolic (n = 73, 45%) categories, with additional associations across the phenome. The strongest ECG association was with hypertension (p < 2.2×10-308). The ECG latent space model demonstrated more associations than models using standard ECG intervals, and offered favorable discrimination of prevalent disease compared to models comprising age, sex, and race. We further demonstrate how latent space models can be used to generate disease-specific ECG waveforms and facilitate individual disease profiling.
Collapse
Grants
- U01NS069763 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K24HL105780 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 21SFRN812095 American Heart Association (American Heart Association, Inc.)
- 18SFRN34250007 American Heart Association (American Heart Association, Inc.)
- 23CDA1050571 American Heart Association (American Heart Association, Inc.)
- 18SFRN34110082 American Heart Association (American Heart Association, Inc.)
- R01HL140224 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL139731 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K23 HL159243 NHLBI NIH HHS
- K23HL159243 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08HL159346 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL160003 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1R01HL092577 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K23HL169839 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01NS103924 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K24HL153669 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1R01HL139731 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL134893 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 853922 American Heart Association (American Heart Association, Inc.)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NHLBI BioData Catalyst Fellows program
- European Union MAESTRIA 965286
Collapse
Affiliation(s)
- Sam F Friedman
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shaan Khurshid
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Rachael A Venn
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Xin Wang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nate Diamant
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher Reeder
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Division of Cardiology, University of California San Francisco, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emily S Lau
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Christopher D Anderson
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Mahnaz Maddah
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer E Ho
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Steven A Lubitz
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
Adel FW, Sang P, Walsh C, Maheshwari A, Cummings P, Attia Z, Mangold K, Davidge-Pitts C, Lopez-Jimenez F, Friedman P, Noseworthy PA, Mankad R. Artificial intelligence evaluation of electrocardiographic characteristics and interval changes in transgender patients on gender-affirming hormone therapy. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2025; 6:55-62. [PMID: 39846073 PMCID: PMC11750187 DOI: 10.1093/ehjdh/ztae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/06/2024] [Accepted: 08/25/2024] [Indexed: 01/24/2025]
Abstract
Aims Gender-affirming hormone therapy (GAHT) is used by some transgender individuals (TG), who comprise 1.4% of US population. However, the effects of GAHT on electrocardiogram (ECG) remain unknown. The objective is to assess the effects of GAHT on ECG changes in TG. Methods and results Twelve-lead ECGs of TG on GAHT at the Mayo Clinic were inspected using a validated artificial intelligence (AI) algorithm. The algorithm assigns a patient's ECG male pattern probability on a scale of 0 (female) to 1 (male). In the primary analysis, done separately for transgender women (TGW) and transgender men (TGM), 12-lead ECGs were used to estimate the male pattern probability before and after GAHT. In a subanalysis, only patients with both pre- and post-GAHT EGCs were included. Further, the autopopulated PR, QRS, and QTc intervals were compared before and after GAHT. Among TGW (n = 86), the probability (mean ± SD) of an ECG male pattern was 0.84 ± 0.25 in the pre-GAHT group, and it was lowered to 0.59 ± 0.36 in the post-GAHT group (n = 173, P < 7.8 × 10-10). Conversely, among TGM, male pattern probability was 0.16 ± 0.28 (n = 47) in the pre-GAHT group, and it was higher at 0.41 ± 0.38 in the post-GAHT group (n = 53, P < 2.4×10-4). The trend persisted in the subanalysis. Furthermore, both the PR (P = 5.68 × 10-4) and QTc intervals (P = 6.65×10-6) prolonged among TGW. Among TGM, the QTc interval shortened (P = 4.8 × 10-2). Conclusion Among TG, GAHT is associated with ECG changes trending towards gender congruence, as determined by the AI algorithm and ECG intervals. Prospective studies are warranted to understand GAHT effects on cardiac structure and function.
Collapse
Affiliation(s)
- Fadi W Adel
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Philip Sang
- Department of Internal Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Connor Walsh
- Department of Internal Medicine, University of Washington, 2505 2nd Ave, Seattle, WA 98121, USA
| | - Arvind Maheshwari
- Advocate Medical Group, 27750 West Highway 22 Suite 110, Barrington, IL 60010, USA
| | - Paige Cummings
- Mayo Clinic Alix School of Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Zachi Attia
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Kathryn Mangold
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Caroline Davidge-Pitts
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | | | - Paul Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Peter A Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Rekha Mankad
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| |
Collapse
|
21
|
Çamkıran V, Tunç H, Achmar B, Ürker TS, Kutlu İ, Torun A. Artificial intelligence (ChatGPT) ready to evaluate ECG in real life? Not yet! Digit Health 2025; 11:20552076251325279. [PMID: 40078449 PMCID: PMC11898233 DOI: 10.1177/20552076251325279] [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: 11/14/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Objective This study aims at evaluating if ChatGPT-based artificial intelligence (AI) models are effective in interpreting electrocardiograms (ECGs) and determine their accuracy as compared to those of cardiologists. The purpose is therefore to explore if ChatGPT can be employed for clinical setting, particularly where there are no available cardiologists. Methods A total of 107 ECG cases classified according to difficulty (simple, intermediate, complex) were analyzed using three AI models (GPT-ECGReader, GPT-ECGAnalyzer, GPT-ECGInterpreter) and compared with the performance of two cardiologists. The statistical analysis was conducted using chi-square and Fisher exact tests using scikit-learn library in Python 3.8. Results Cardiologists demonstrated superior accuracy (92.52%) compared to ChatGPT-based models (GPT-ECGReader: 57.94%, GPT-ECGInterpreter: 62.62%, GPT-ECGAnalyzer: 62.62%). Statistically significant differences were observed between cardiologists and AI models (p < 0.05). ChatGPT models exhibited enhanced performance with female patients; however, the differences found were not statistically significant. Cardiologists significantly outperformed AI models across all difficulty levels. When it comes to diagnosing patients with arrhythmia (A) and cardiac structural disease ECG patterns, cardiologists gave the best results though there was no statistical difference between them and AI models in diagnosing people with normal (N) ECG patterns. Conclusions ChatGPT-based models have potential in ECG interpretation; however, they currently lack adequate reliability beyond oversight from a doctor. Additionally, further studies that would improve the accuracy of these models, especially in intricate diagnoses are needed.
Collapse
Affiliation(s)
- Volkan Çamkıran
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| | - Hüseyin Tunç
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| | - Batool Achmar
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| | - Tuğçe Simay Ürker
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| | - İlhan Kutlu
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| | - Akin Torun
- Bahçeşehir Üniversite Hastanesi Medical Park Göztepe, İstanbul, Turkey
| |
Collapse
|
22
|
Al-Falahi ZS, Schlegel TT, Palencia-Lamela I, Li A, Schelbert EB, Niklasson L, Maanja M, Lindow T, Ugander M. Advanced electrocardiography heart age: a prognostic, explainable machine learning approach applicable to sinus and non-sinus rhythms. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2025; 6:45-54. [PMID: 39846063 PMCID: PMC11750191 DOI: 10.1093/ehjdh/ztae075] [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: 06/03/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 01/24/2025]
Abstract
Aims An explainable advanced electrocardiography (A-ECG) Heart Age gap is the difference between A-ECG Heart Age and chronological age. This gap is an estimate of accelerated cardiovascular aging expressed in years of healthy human aging, and can intuitively communicate cardiovascular risk to the general population. However, existing A-ECG Heart Age requires sinus rhythm. We aim to develop and prognostically validate a revised, explainable A-ECG Heart Age applicable to both sinus and non-sinus rhythms. Methods and results An A-ECG Heart Age excluding P-wave measures was derived from the 10-s 12-lead ECG in a derivation cohort using multivariable regression machine learning with Bayesian 5-min 12-lead A-ECG Heart Age as reference. The Heart Age was externally validated in a separate cohort of patients referred for cardiovascular magnetic resonance imaging by describing its association with heart failure hospitalization or death using Cox regression, and its association with comorbidities. In the derivation cohort (n = 2771), A-ECG Heart Age agreed with the 5-min Heart Age (R 2 = 0.91, bias 0.0 ± 6.7 years), and increased with increasing comorbidity. In the validation cohort [n = 731, mean age 54 ± 15 years, 43% female, n = 139 events over 5.7 (4.8-6.7) years follow-up], increased A-ECG Heart Age gap (≥10 years) associated with events [hazard ratio, HR (95% confidence interval, CI) 2.04 (1.38-3.00), C-statistic 0.58 (0.54-0.62)], and the presence of hypertension, diabetes mellitus, hypercholesterolaemia, and heart failure (P ≤ 0.009 for all). Conclusion An explainable A-ECG Heart Age gap applicable to both sinus and non-sinus rhythm associates with cardiovascular risk, cardiovascular morbidity, and survival.
Collapse
Affiliation(s)
- Zaidon S Al-Falahi
- Kolling Institute, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW 2065, Australia
- Department of Cardiology, Campbelltown Hospital, South West Sydney Local Health District, NSW 2560, Australia
| | - Todd T Schlegel
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, SE-17176 Stockholm, Sweden
- Nicollier-Schlegel SARL, Trélex 1270, Switzerland
| | - Israel Palencia-Lamela
- Kolling Institute, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW 2065, Australia
| | - Annie Li
- Kolling Institute, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW 2065, Australia
| | - Erik B Schelbert
- Minneapolis Heart Institute East, United Hospital, Minneapolis, MN 55407, USA
| | - Louise Niklasson
- Minneapolis Heart Institute East, United Hospital, Minneapolis, MN 55407, USA
| | - Maren Maanja
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Thomas Lindow
- Kolling Institute, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW 2065, Australia
- Department of Medicine, Research and Development, Växjö Central Hospital, 35188 Region Kronoberg, Sweden
- Respiratory Medicine, Allergology and Palliative Medicine, Clinical Sciences, Lund University, 22100 Lund, Sweden
| | - Martin Ugander
- Kolling Institute, Royal North Shore Hospital, University of Sydney, St Leonards, Sydney, NSW 2065, Australia
- Department of Clinical Physiology, Karolinska University Hospital, and Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
23
|
Mayourian J, van Boxtel JPA, Sleeper LA, Diwanji V, Geva A, O'Leary ET, Triedman JK, Ghelani SJ, Wald RM, Valente AM, Geva T. Electrocardiogram-Based Deep Learning to Predict Mortality in Repaired Tetralogy of Fallot. JACC Clin Electrophysiol 2024; 10:2600-2612. [PMID: 39297841 DOI: 10.1016/j.jacep.2024.07.015] [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: 06/26/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Artificial intelligence-enhanced electrocardiogram (AI-ECG) analysis shows promise to predict mortality in adults with acquired cardiovascular diseases. However, its application to the growing repaired tetralogy of Fallot (rTOF) population remains unexplored. OBJECTIVES This study aimed to develop and externally validate an AI-ECG model to predict 5-year mortality in rTOF. METHODS A convolutional neural network was trained on electrocardiograms (ECGs) obtained at Boston Children's Hospital and tested on Boston (internal testing) and Toronto (external validation) INDICATOR (International Multicenter TOF Registry) cohorts to predict 5-year mortality. Model performance was evaluated on single ECGs per patient using area under the receiver operating (AUROC) and precision recall (AUPRC) curves. RESULTS The internal testing and external validation cohorts comprised of 1,054 patients (13,077 ECGs at median age 17.8 [Q1-Q3: 7.9-30.5] years; 54% male; 6.1% mortality) and 335 patients (5,014 ECGs at median age 38.3 [Q1-Q3: 29.1-48.7] years; 57% male; 8.4% mortality), respectively. Model performance was similar during internal testing (AUROC 0.83, AUPRC 0.18) and external validation (AUROC 0.81, AUPRC 0.21). AI-ECG performed similarly to the biventricular global function index (an imaging biomarker) and outperformed QRS duration. AI-ECG 5-year mortality prediction, but not QRS duration, was a significant independent predictor when added into a Cox regression model with biventricular global function index to predict shorter time-to-death on internal and external cohorts. Saliency mapping identified QRS fragmentation, wide and low amplitude QRS complexes, and flattened T waves as high-risk features. CONCLUSIONS This externally validated AI-ECG model may complement imaging biomarkers to improve risk stratification in patients with rTOF.
Collapse
Affiliation(s)
- Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Juul P A van Boxtel
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Lynn A Sleeper
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Vedang Diwanji
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alon Geva
- Department of Anesthesiology, Critical Care, and Pain Medicine, and Computational Health Informatics Program, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Anesthesia, Harvard Medical School, Boston, Massachusetts, USA
| | - Edward T O'Leary
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - John K Triedman
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunil J Ghelani
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel M Wald
- Division of Cardiology, University of Toronto, Peter Munk Cardiac Centre, Toronto, Ontario, Canada
| | - Anne Marie Valente
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Tal Geva
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
24
|
Anjewierden S, O'Sullivan D, Mangold KE, Greason G, Attia IZ, Lopez‐Jimenez F, Friedman PA, Asirvatham SJ, Anderson J, Eidem BW, Johnson JN, Havangi Prakash S, Niaz T, Madhavan M. Detection of Right and Left Ventricular Dysfunction in Pediatric Patients Using Artificial Intelligence-Enabled ECGs. J Am Heart Assoc 2024; 13:e035201. [PMID: 39494568 PMCID: PMC11935708 DOI: 10.1161/jaha.124.035201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Early detection of left and right ventricular systolic dysfunction (LVSD and RVSD respectively) in children can lead to intervention to reduce morbidity and death. Existing artificial intelligence algorithms can identify LVSD and RVSD in adults using a 12-lead ECG; however, its efficacy in children is uncertain. We aimed to develop novel artificial intelligence-enabled ECG algorithms for LVSD and RVSD detection in pediatric patients. METHODS AND RESULTS We identified 10 142 unique pediatric patients (age≤18) with a 10-second, 12-lead surface ECG within 14 days of a transthoracic echocardiogram, performed between 2002 and 2022. LVSD was defined quantitatively by left ventricular ejection fraction (LVEF). RVSD was defined semiquantitatively. Novel pediatric models for LVEF ≤35% and LVEF <50% achieved excellent test areas under the curve of 0.93 (95% CI, 0.89-0.98) and 0.88 (95% CI, 0.83-0.94) respectively. The model to detect LVEF <50% had a sensitivity of 0.85, specificity of 0.80, positive predictive value of 0.095, and negative predictive value of 0.995. In comparison, the previously validated adult data-derived model for LVEF <35% achieved an area under the curve of 0.87 (95% CI, 0.84-0.90) for LVEF ≤35% in children. A novel pediatric model for any RVSD detection reached a test area under the curve of 0.90 (0.87-0.94). CONCLUSIONS An artificial intelligence-enabled ECG demonstrates accurate detection of both LVSD and RVSD in pediatric patients. While adult-trained models offer good performance, improvements are seen when training pediatric-specific models.
Collapse
Affiliation(s)
- Scott Anjewierden
- Department of Pediatrics and Adolescent MedicineMayo ClinicRochesterMNUSA
| | | | | | - Grace Greason
- Department of Cardiovascular MedicineMayo ClinicRochesterMNUSA
| | | | | | | | | | - Jason Anderson
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent MedicineMayo ClinicRochesterMNUSA
| | - Benjamin W. Eidem
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent MedicineMayo ClinicRochesterMNUSA
| | - Jonathan N. Johnson
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent MedicineMayo ClinicRochesterMNUSA
| | | | - Talha Niaz
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent MedicineMayo ClinicRochesterMNUSA
| | - Malini Madhavan
- Department of Cardiovascular MedicineMayo ClinicRochesterMNUSA
| |
Collapse
|
25
|
Lee SH, Jeon KL, Lee YJ, You SC, Lee SJ, Hong SJ, Ahn CM, Kim JS, Kim BK, Ko YG, Choi D, Hong MK. Development of Clinically Validated Artificial Intelligence Model for Detecting ST-segment Elevation Myocardial Infarction. Ann Emerg Med 2024; 84:540-548. [PMID: 39066765 DOI: 10.1016/j.annemergmed.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
STUDY OBJECTIVE Although the importance of primary percutaneous coronary intervention has been emphasized for ST-segment elevation myocardial infarction (STEMI), the appropriateness of the cardiac catheterization laboratory activation remains suboptimal. This study aimed to develop a precise artificial intelligence (AI) model for the diagnosis of STEMI and accurate cardiac catheterization laboratory activation. METHODS We used electrocardiography (ECG) waveform data from a prospective percutaneous coronary intervention registry in Korea in this study. Two independent board-certified cardiologists established a criterion standard (STEMI or Not STEMI) for each ECG based on corresponding coronary angiography data. We developed a deep ensemble model by combining 5 convolutional neural networks. In addition, we performed clinical validation based on a symptom-based ECG data set, comparisons with clinical physicians, and external validation. RESULTS We used 18,697 ECGs for the model development data set, and 1,745 (9.3%) were STEMI. The AI model achieved an accuracy of 92.1%, sensitivity of 95.4%, and specificity of 91.8 %. The performances of the AI model were well balanced and outstanding in the clinical validation, comparison with clinical physicians, and the external validation. CONCLUSION The deep ensemble AI model showed a well-balanced and outstanding performance. As visualized with gradient-weighted class activation mapping, the AI model has a reasonable explainability. Further studies with prospective validation regarding clinical benefit in a real-world setting should be warranted.
Collapse
Affiliation(s)
- Sang-Hyup Lee
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyu Lee Jeon
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, South Korea
| | - Yong-Joon Lee
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Seng Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, South Korea.
| | - Seung-Jun Lee
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Jin Hong
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chul-Min Ahn
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung-Sun Kim
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Byeong-Keuk Kim
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Young-Guk Ko
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Donghoon Choi
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Myeong-Ki Hong
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
26
|
Alahdab F, Saad MB, Ahmed AI, Al Tashi Q, Aminu M, Han Y, Moody JB, Murthy VL, Wu J, Al-Mallah MH. Development and validation of a machine learning model to predict myocardial blood flow and clinical outcomes from patients' electrocardiograms. Cell Rep Med 2024; 5:101746. [PMID: 39326409 PMCID: PMC11513811 DOI: 10.1016/j.xcrm.2024.101746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/24/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
We develop a machine learning (ML) model using electrocardiography (ECG) to predict myocardial blood flow reserve (MFR) and assess its prognostic value for major adverse cardiovascular events (MACEs). Using 3,639 ECG-positron emission tomography (PET) and 17,649 ECG-single-photon emission computed tomography (SPECT) data pairs, the ML model is trained with a swarm intelligence approach and support vector regression (SVR). The model achieves a receiver-operator curve (ROC) area under the curve (AUC) of 0.83, with a sensitivity and specificity of 0.75. An ECG-MFR value below 2 is significantly associated with MACE, with hazard ratios (HRs) of 3.85 and 3.70 in the discovery and validation phases, respectively. The model's C-statistic is 0.76, with a net reclassification improvement (NRI) of 0.35. Validated in an independent cohort, the ML model using ECG data offers superior MACE prediction compared to baseline clinical models, highlighting its potential for risk stratification in patients with coronary artery disease (CAD) using the accessible 12-lead ECG.
Collapse
Affiliation(s)
- Fares Alahdab
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA; Departments of Biomedical Informatics, Biostatistics, Epidemiology, and Cardiology, University of Missouri, Columbia, MO
| | - Maliazurina Binti Saad
- Department of Imaging Physics, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Qasem Al Tashi
- Department of Imaging Physics, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Yushui Han
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Jonathan B Moody
- INVIA Medical Imaging Solutions, 3025 Boardwalk Dr., Suite 200, Ann Arbor, MI 48108, USA
| | - Venkatesh L Murthy
- Division of Cardiovascular Medicine, Department of Medicine, and Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Jia Wu
- Department of Imaging Physics, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX, USA. //
| | - Mouaz H Al-Mallah
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA.
| |
Collapse
|
27
|
Nolin-Lapalme A, Corbin D, Tastet O, Avram R, Hussin JG. Advancing Fairness in Cardiac Care: Strategies for Mitigating Bias in Artificial Intelligence Models Within Cardiology. Can J Cardiol 2024; 40:1907-1921. [PMID: 38735528 DOI: 10.1016/j.cjca.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
In the dynamic field of medical artificial intelligence (AI), cardiology stands out as a key area for its technological advancements and clinical application. In this review we explore the complex issue of data bias, specifically addressing those encountered during the development and implementation of AI tools in cardiology. We dissect the origins and effects of these biases, which challenge their reliability and widespread applicability in health care. Using a case study, we highlight the complexities involved in addressing these biases from a clinical viewpoint. The goal of this review is to equip researchers and clinicians with the practical knowledge needed to identify, understand, and mitigate these biases, advocating for the creation of AI solutions that are not just technologically sound, but also fair and effective for all patients.
Collapse
Affiliation(s)
- Alexis Nolin-Lapalme
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada; Mila - Québec AI Institute, Montreal, Quebec, Canada; Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Quebec, Canada.
| | - Denis Corbin
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Olivier Tastet
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Robert Avram
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada; Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Quebec, Canada
| | - Julie G Hussin
- Department of Medicine, Montreal Heart Institute, Montreal, Quebec, Canada; Faculté de Médecine, Université de Montréal, Montreal, Quebec, Canada; Mila - Québec AI Institute, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Islam MS, Kalmady SV, Hindle A, Sandhu R, Sun W, Sepehrvand N, Greiner R, Kaul P. Diagnostic and Prognostic Electrocardiogram-Based Models for Rapid Clinical Applications. Can J Cardiol 2024; 40:1788-1803. [PMID: 38992812 DOI: 10.1016/j.cjca.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
Leveraging artificial intelligence (AI) for the analysis of electrocardiograms (ECGs) has the potential to transform diagnosis and estimate the prognosis of not only cardiac but, increasingly, noncardiac conditions. In this review, we summarize clinical studies and AI-enhanced ECG-based clinical applications in the early detection, diagnosis, and estimating prognosis of cardiovascular diseases in the past 5 years (2019-2023). With advancements in deep learning and the rapid increased use of ECG technologies, a large number of clinical studies have been published. However, most of these studies are single-centre, retrospective, proof-of-concept studies that lack external validation. Prospective studies that progress from development toward deployment in clinical settings account for < 15% of the studies. Successful implementations of ECG-based AI applications that have received approval from the Food and Drug Administration have been developed through commercial collaborations, with approximately half of them being for mobile or wearable devices. The field is in its early stages, and overcoming several obstacles is essential, such as prospective validation in multicentre large data sets, addressing technical issues, bias, privacy, data security, model generalizability, and global scalability. This review concludes with a discussion of these challenges and potential solutions. By providing a holistic view of the state of AI in ECG analysis, this review aims to set a foundation for future research directions, emphasizing the need for comprehensive, clinically integrated, and globally deployable AI solutions in cardiovascular disease management.
Collapse
Affiliation(s)
- Md Saiful Islam
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Sunil Vasu Kalmady
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Abram Hindle
- Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Roopinder Sandhu
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Smidt Heart Institute, Cedars-Sinai Medical Center Hospital System, Los Angeles, California, USA
| | - Weijie Sun
- Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada
| | - Nariman Sepehrvand
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Russell Greiner
- Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada; Alberta Machine Intelligence Institute, Edmonton, Alberta, Canada
| | - Padma Kaul
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
29
|
Adedinsewo DA, Morales-Lara AC, Afolabi BB, Kushimo OA, Mbakwem AC, Ibiyemi KF, Ogunmodede JA, Raji HO, Ringim SH, Habib AA, Hamza SM, Ogah OS, Obajimi G, Saanu OO, Jagun OE, Inofomoh FO, Adeolu T, Karaye KM, Gaya SA, Alfa I, Yohanna C, Venkatachalam KL, Dugan J, Yao X, Sledge HJ, Johnson PW, Wieczorek MA, Attia ZI, Phillips SD, Yamani MH, Tobah YB, Rose CH, Sharpe EE, Lopez-Jimenez F, Friedman PA, Noseworthy PA, Carter RE. Artificial intelligence guided screening for cardiomyopathies in an obstetric population: a pragmatic randomized clinical trial. Nat Med 2024; 30:2897-2906. [PMID: 39223284 PMCID: PMC11485252 DOI: 10.1038/s41591-024-03243-9] [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: 06/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Nigeria has the highest reported incidence of peripartum cardiomyopathy worldwide. This open-label, pragmatic clinical trial randomized pregnant and postpartum women to usual care or artificial intelligence (AI)-guided screening to assess its impact on the diagnosis left ventricular systolic dysfunction (LVSD) in the perinatal period. The study intervention included digital stethoscope recordings with point of-care AI predictions and a 12-lead electrocardiogram with asynchronous AI predictions for LVSD. The primary end point was identification of LVSD during the study period. In the intervention arm, the primary end point was defined as the number of identified participants with LVSD as determined by a positive AI screen, confirmed by echocardiography. In the control arm, this was the number of participants with clinical recognition and documentation of LVSD on echocardiography in keeping with current standard of care. Participants in the intervention arm had a confirmatory echocardiogram at baseline for AI model validation. A total of 1,232 (616 in each arm) participants were randomized and 1,195 participants (587 intervention arm and 608 control arm) completed the baseline visit at 6 hospitals in Nigeria between August 2022 and September 2023 with follow-up through May 2024. Using the AI-enabled digital stethoscope, the primary study end point was met with detection of 24 out of 587 (4.1%) versus 12 out of 608 (2.0%) patients with LVSD (intervention versus control odds ratio 2.12, 95% CI 1.05-4.27; P = 0.032). With the 12-lead AI-electrocardiogram model, the primary end point was detected in 20 out of 587 (3.4%) versus 12 out of 608 (2.0%) patients (odds ratio 1.75, 95% CI 0.85-3.62; P = 0.125). A similar direction of effect was observed in prespecified subgroup analysis. There were no serious adverse events related to study participation. In pregnant and postpartum women, AI-guided screening using a digital stethoscope improved the diagnosis of pregnancy-related cardiomyopathy. ClinicalTrials.gov registration: NCT05438576.
Collapse
Affiliation(s)
| | | | - Bosede B Afolabi
- Department of Obstetrics and Gynaecology, College of Medicine and Centre for Clinical Trials, Research and Implementation Science, University of Lagos, Lagos, Nigeria
| | - Oyewole A Kushimo
- Cardiology Unit, Department of Medicine, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Amam C Mbakwem
- Cardiology Unit, Department of Medicine, Lagos University Teaching Hospital, Lagos, Nigeria
| | - Kehinde F Ibiyemi
- Department of Obstetrics & Gynaecology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | | | - Hadijat Olaide Raji
- Department of Obstetrics & Gynaecology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Sadiq H Ringim
- Department of Medicine, Rasheed Shekoni Specialist Hospital, Dutse, Nigeria
| | - Abdullahi A Habib
- Department of Obstetrics and Gynaecology, Rasheed Shekoni Specialist Hospital, Dutse, Nigeria
| | - Sabiu M Hamza
- Department of Medicine, Rasheed Shekoni Specialist Hospital, Dutse, Nigeria
| | | | - Gbolahan Obajimi
- Department of Obstetrics and Gynaecology, University College Hospital Ibadan, Oyo, Nigeria
| | | | - Olusoji E Jagun
- Department of Obstetrics and Gynaecology, Olabisi Onabanjo University Teaching Hospital, Sagamu, Nigeria
| | - Francisca O Inofomoh
- Cardiology Unit, Department of Medicine, Olabisi Onabanjo University Teaching Hospital, Sagamu, Nigeria
| | - Temitope Adeolu
- Cardiology Unit, Department of Medicine, Olabisi Onabanjo University Teaching Hospital, Sagamu, Nigeria
| | - Kamilu M Karaye
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Sule A Gaya
- Department of Obstetrics and Gynaecology, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Isiaka Alfa
- Department of Medicine, Bayero University and Aminu Kano Teaching Hospital, Kano, Nigeria
| | - Cynthia Yohanna
- Lakeside Healthcare at Yaxley, the Health Centre, Peterborough, United Kingdom
| | - K L Venkatachalam
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Jennifer Dugan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiaoxi Yao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Hanna J Sledge
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Patrick W Johnson
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Mikolaj A Wieczorek
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Zachi I Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sabrina D Phillips
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamad H Yamani
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Carl H Rose
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Emily E Sharpe
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Rickey E Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
30
|
Sigfstead S, Jiang R, Avram R, Davies B, Krahn AD, Cheung CC. Applying Artificial Intelligence for Phenotyping of Inherited Arrhythmia Syndromes. Can J Cardiol 2024; 40:1841-1851. [PMID: 38670456 DOI: 10.1016/j.cjca.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/08/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
Inherited arrhythmia disorders account for a significant proportion of sudden cardiac death, particularly among young individuals. Recent advances in our understanding of these syndromes have improved patient diagnosis and care, yet certain clinical gaps remain, particularly within case ascertainment, access to genetic testing, and risk stratification. Artificial intelligence (AI), specifically machine learning and its subset deep learning, present promising solutions to these challenges. The capacity of AI to process vast amounts of patient data and identify disease patterns differentiates them from traditional methods, which are time- and resource-intensive. To date, AI models have shown immense potential in condition detection (including asymptomatic/concealed disease) and genotype and phenotype identification, exceeding expert cardiologists in these tasks. Additionally, they have exhibited applicability for general population screening, improving case ascertainment in a set of conditions that are often asymptomatic such as left ventricular dysfunction. Third, models have shown the ability to improve testing protocols; through model identification of disease and genotype, specific clinical testing (eg, drug challenges or further diagnostic imaging) can be avoided, reducing health care expenses, speeding diagnosis, and possibly allowing for more incremental or targeted genetic testing approaches. These significant benefits warrant continued investigation of AI, particularly regarding the development and implementation of clinically applicable screening tools. In this review we summarize key developments in AI, including studies in long QT syndrome, Brugada syndrome, hypertrophic cardiomyopathy, and arrhythmogenic cardiomyopathies, and provide direction for effective future AI implementation in clinical practice.
Collapse
Affiliation(s)
- Sophie Sigfstead
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - River Jiang
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert Avram
- Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Quebec, Canada; Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Brianna Davies
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew D Krahn
- Center for Cardiovascular Innovation, Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Christopher C Cheung
- Division of Cardiology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Elyamani HA, Salem MA, Melgani F, Yhiea NM. Deep residual 2D convolutional neural network for cardiovascular disease classification. Sci Rep 2024; 14:22040. [PMID: 39327440 PMCID: PMC11427665 DOI: 10.1038/s41598-024-72382-3] [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: 03/20/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Cardiovascular disease (CVD) continues to be a major global health concern, underscoring the need for advancements in medical care. The use of electrocardiograms (ECGs) is crucial for diagnosing cardiac conditions. However, the reliance on professional expertise for manual ECG interpretation poses challenges for expanding accessible healthcare, particularly in community hospitals. To address this, there is a growing interest in leveraging automated and AI-driven ECG analysis systems, which can enhance diagnostic accuracy and efficiency, making quality cardiac care more accessible to a broader population. In this study, we implemented a novel deep two-dimensional convolutional neural network (2D-CNN) on a dataset of PTB-XL for cardiac disorder detection. The studies were performed on 2, 5, and 23 classes of cardiovascular diseases. The our network in classifying healthy/sick patients achived an AUC of 95% and an average accuracy of 87.85%. In 5-classes classification, our model achieved an AUC of 93.46% with an average accuracy of 89.87%. In a more complex scenario involving classification into 23 different classes, the model achieved an AUC of 92.18% and an accuracy of 96.88%. According to the experimental results, our model obtained the best classification result compared to the other methods based on the same public dataset. This indicates that our method can aid healthcare professionals in the clinical analysis of ECGs, offering valuable assistance in diagnosing CVD and contributing to the advancement of computer-aided diagnosis technology.
Collapse
Affiliation(s)
- Haneen A Elyamani
- Department of Mathematics, Faculty of Science, Suez Canal University, Ismailia, 44745, Egypt.
| | - Mohammed A Salem
- Media Engineering and Technology, German University in Cairo (GUC), Cairo, Egypt
| | - Farid Melgani
- Department of Information Engineering and Computer Science, University of Trento, Via Sommarive, 14, I-3812, Trento, Italy
| | - N M Yhiea
- Department of Mathematics, Faculty of Science, Suez Canal University, Ismailia, 44745, Egypt
- Faculty of Informatics and Computer Science, The British University in Egypt (BUE), Cairo, Egypt
| |
Collapse
|
32
|
Park H, Kwon OS, Shim J, Kim D, Park JW, Kim YG, Yu HT, Kim TH, Uhm JS, Choi JI, Joung B, Lee MH, Pak HN. Artificial intelligence estimated electrocardiographic age as a recurrence predictor after atrial fibrillation catheter ablation. NPJ Digit Med 2024; 7:234. [PMID: 39237703 PMCID: PMC11377779 DOI: 10.1038/s41746-024-01234-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
The application of artificial intelligence (AI) algorithms to 12-lead electrocardiogram (ECG) provides promising age prediction models. We explored whether the gap between the pre-procedural AI-ECG age and chronological age can predict atrial fibrillation (AF) recurrence after catheter ablation. We validated a pre-trained residual network-based model for age prediction on four multinational datasets. Then we estimated AI-ECG age using a pre-procedural sinus rhythm ECG among individuals on anti-arrhythmic drugs who underwent de-novo AF catheter ablation from two independent AF ablation cohorts. We categorized the AI-ECG age gap based on the mean absolute error of the AI-ECG age gap obtained from four model validation datasets; aged-ECG (≥10 years) and normal ECG age (<10 years) groups. In the two AF ablation cohorts, aged-ECG was associated with a significantly increased risk of AF recurrence compared to the normal ECG age group. These associations were independent of chronological age or left atrial diameter. In summary, a pre-procedural AI-ECG age has a prognostic value for AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Hanjin Park
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Oh-Seok Kwon
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Jaemin Shim
- Division of Cardiology, Department of Internal Medicine, Korea University Medical Center, Seoul, Republic of Korea.
| | - Daehoon Kim
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Je-Wook Park
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Yun-Gi Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Medical Center, Seoul, Republic of Korea
| | - Hee Tae Yu
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Jae-Sun Uhm
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Medical Center, Seoul, Republic of Korea
| | - Boyoung Joung
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Moon-Hyoung Lee
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Hui-Nam Pak
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Rajai N, Medina-Inojosa JR, Lewis BR, Sheffeh MA, Baez-Suarez A, Nyman M, Attia ZI, Lerman LO, Medina-Inojosa BJ, Friedman PA, Lopez-Jimenez F, Lerman A. Association Between Social Isolation With Age-Gap Determined by Artificial Intelligence-Enabled Electrocardiography. JACC. ADVANCES 2024; 3:100890. [PMID: 39372468 PMCID: PMC11450907 DOI: 10.1016/j.jacadv.2024.100890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 10/08/2024]
Abstract
Background Loneliness and social isolation are associated with poor health outcomes such as an increased risk of cardiovascular diseases. Objectives The authors aimed to explore the association between social isolation with biological aging which was determined by artificial intelligence-enabled electrocardiography (AI-ECG) as well as the risk of all-cause mortality. Methods The study included adults aged ≥18 years seen at Mayo Clinic from 2019 to 2022 who respond to a survey for social isolation assessment and had a 12-lead ECG within 1 year of completing the questionnaire. Biological age was determined from ECGs using a previously developed and validated convolutional neural network (AI-ECG age). Age-Gap was defined as AI-ECG age minus chronological age, where positive values reflect an older-than-expected age. The status of social isolation was measured by the previously validated multiple-choice questions based on Social Network Index (SNI) with score ranges between 0 (most isolated) and 4 (least isolated). Results A total of 280,324 subjects were included (chronological age 59.8 ± 16.4 years, 50.9% female). The mean Age-Gap was -0.2 ± 9.16 years. A higher SNI was associated with a lower Age-Gap (β of SNI = 4 was -0.11; 95% CI: -0.22 to -0.01; P < 0.001, adjusted to covariates). Cox proportional hazard analysis revealed the association between social connection and all-cause mortality (HR for SNI = 4, 0.47; 95% CI: 0.43-0.5; P < 0.001). Conclusions Social isolation is associated with accelerating biological aging and all-cause mortality independent of conventional cardiovascular risk factors. This observation underscores the need to address social connection as a health care determinant.
Collapse
Affiliation(s)
- Nazanin Rajai
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Bradley R. Lewis
- Division of Biomedical Statistics and Informatics, Mayo College of Medicine, Rochester, Minnesota, USA
| | | | - Abraham Baez-Suarez
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark Nyman
- Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Zachi I. Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Paul A. Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
34
|
Saleh G, Sularz A, Liu CH, Lo Russo GV, Adi MZ, Attia Z, Friedman P, Gulati R, Alkhouli M. Artificial Intelligence Electrocardiogram-Derived Heart Age Predicts Long-Term Mortality After Transcatheter Aortic Valve Replacement. JACC. ADVANCES 2024; 3:101171. [PMID: 39372454 PMCID: PMC11450920 DOI: 10.1016/j.jacadv.2024.101171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Affiliation(s)
- Ghasaq Saleh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Agata Sularz
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Chia-Hao Liu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Gerardo V. Lo Russo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mahmoud Zhour Adi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Zachi Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Rajiv Gulati
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mohamad Alkhouli
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
35
|
Pencovich N, Smith BH, Attia ZI, Jimenez FL, Bentall AJ, Schinstock CA, Khamash HA, Jadlowiec CC, Jarmi T, Mao SA, Park WD, Diwan TS, Friedman PA, Stegall MD. Electrocardiography-based Artificial Intelligence Algorithms Aid in Prediction of Long-term Mortality After Kidney Transplantation. Transplantation 2024; 108:1976-1985. [PMID: 38557657 DOI: 10.1097/tp.0000000000005023] [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: 04/04/2024]
Abstract
BACKGROUND Predicting long-term mortality postkidney transplantation (KT) using baseline clinical data presents significant challenges. This study aims to evaluate the predictive power of artificial intelligence (AI)-enabled analysis of preoperative electrocardiograms (ECGs) in forecasting long-term mortality following KT. METHODS We analyzed preoperative ECGs from KT recipients at three Mayo Clinic sites (Minnesota, Florida, and Arizona) between January 1, 2006, and July 30, 2021. The study involved 6 validated AI algorithms, each trained to predict future development of atrial fibrillation, aortic stenosis, low ejection fraction, hypertrophic cardiomyopathy, amyloid heart disease, and biological age. These algorithms' outputs based on a single preoperative ECG were correlated with patient mortality data. RESULTS Among 6504 KT recipients included in the study, 1764 (27.1%) died within a median follow-up of 5.7 y (interquartile range: 3.00-9.29 y). All AI-ECG algorithms were independently associated with long-term all-cause mortality ( P < 0.001). Notably, few patients had a clinical cardiac diagnosis at the time of transplant, indicating that AI-ECG scores were predictive even in asymptomatic patients. When adjusted for multiple clinical factors such as recipient age, diabetes, and pretransplant dialysis, AI algorithms for atrial fibrillation and aortic stenosis remained independently associated with long-term mortality. These algorithms also improved the C-statistic for predicting overall (C = 0.74) and cardiac-related deaths (C = 0.751). CONCLUSIONS The findings suggest that AI-enabled preoperative ECG analysis can be a valuable tool in predicting long-term mortality following KT and could aid in identifying patients who may benefit from enhanced cardiac monitoring because of increased risk.
Collapse
Affiliation(s)
- Niv Pencovich
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
- Department of General Surgery and Transplantation, Sheba Medical Center, Tel Hashomer, Tel-Aviv University, Tel-Aviv, Israel
| | - Byron H Smith
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Zachi I Attia
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Andrew J Bentall
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Carrie A Schinstock
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | | | | | - Tambi Jarmi
- Department of Transplant, Mayo Clinic Florida, Jacksonville, FL
| | - Shennen A Mao
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Phoenix, AZ
| | - Walter D Park
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Tayyab S Diwan
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Mark D Stegall
- Departments of Surgery and Immunology, William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN
| |
Collapse
|
36
|
Tao Y, Zhang D, Tan C, Wang Y, Shi L, Chi H, Geng S, Ma Z, Hong S, Liu XP. An artificial intelligence-enabled electrocardiogram algorithm for the prediction of left atrial low-voltage areas in persistent atrial fibrillation. J Cardiovasc Electrophysiol 2024; 35:1849-1858. [PMID: 39054663 DOI: 10.1111/jce.16373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVES We aimed to construct an artificial intelligence-enabled electrocardiogram (ECG) algorithm that can accurately predict the presence of left atrial low-voltage areas (LVAs) in patients with persistent atrial fibrillation. METHODS The study included 587 patients with persistent atrial fibrillation who underwent catheter ablation procedures between March 2012 and December 2023 and 942 scanned images of 12-lead ECGs obtained before the ablation procedures were performed. Artificial intelligence-based algorithms were used to construct models for predicting the presence of LVAs. The DR-FLASH and APPLE clinical scores for LVA prediction were calculated. We used a receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis to evaluate model performance. RESULTS The data obtained from the participants were split into training (n = 469), validation (n = 58), and test sets (n = 60). LVAs were detected in 53.7% of all participants. Using ECG alone, the deep learning algorithm achieved an area under the ROC curve (AUROC) of 0.752, outperforming both the DR-FLASH score (AUROC = 0.610) and the APPLE score (AUROC = 0.510). The random forest classification model, which integrated a probabilistic deep learning model and clinical features, showed a maximum AUROC of 0.759. Moreover, the ECG-based deep learning algorithm for predicting extensive LVAs achieved an AUROC of 0.775, with a sensitivity of 0.816 and a specificity of 0.896. The random forest classification model for predicting extensive LVAs achieved an AUROC of 0.897, with a sensitivity of 0.862, and a specificity of 0.935. CONCLUSION The deep learning model based exclusively on ECG data and the machine learning model that combined a probabilistic deep learning model and clinical features both predicted the presence of LVAs with a higher degree of accuracy than the DR-FLASH and the APPLE risk scores.
Collapse
Affiliation(s)
- Yirao Tao
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Deyun Zhang
- HeartVoice Medical Technology, Hefei, China
- HeartRhythm-HeartVoice Joint Laboratory, Beijing, China
| | - Chen Tan
- Department of Cardiology, Hebei Yanda Hospital, Hebei, Hebei Province, China
| | - Yanjiang Wang
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Liang Shi
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hongjie Chi
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shijia Geng
- HeartVoice Medical Technology, Hefei, China
- HeartRhythm-HeartVoice Joint Laboratory, Beijing, China
| | - Zhimin Ma
- Department of Cardiology, Heart Rhythm Cardiovascular Hospital, Shandong, China
| | - Shenda Hong
- National Institute of Health Data Science, Peking University, Beijing, China
- Health Science Center of Peking University, Institute of Medical Technology, Beijing, China
| | - Xing Peng Liu
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Wysokinski WE, Meverden RA, Lopez-Jimenez F, Harmon DM, Medina Inojosa BJ, Suarez AB, Liu K, Medina Inojosa JR, Casanegra AI, McBane RD, Houghton DE. Electrocardiogram Signal Analysis With a Machine Learning Model Predicts the Presence of Pulmonary Embolism With Accuracy Dependent on Embolism Burden. MAYO CLINIC PROCEEDINGS. DIGITAL HEALTH 2024; 2:453-462. [PMID: 40206108 PMCID: PMC11975982 DOI: 10.1016/j.mcpdig.2024.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Objective To develop an artificial intelligence deep neural network (AI-DNN) algorithm to analyze 12-lead electrocardiogram (ECG) for detection of acute pulmonary embolism (PE) and PE categories. Patients and Methods A cohort of patients seen between January 1, 1999, and December 31, 2020, from across the Mayo Clinic Enterprise with computed tomography pulmonary angiogram (CTPA) and ECG performed ±6 hours was identified. Natural language processing algorithms were applied to radiology reports to determine the diagnosis of acute PE, acute right ventricular strain pulmonary embolism (RVSPE), saddle pulmonary embolism (SADPE), or no PE. Diagnostic performance parameters of the AI-DNN reported were area under the receiver operating characteristics curve (AUROC), sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). Results A cohort of patients with CTPA report and ECG consisted of 79,894 patients including 7423 (9.3%) with acute PE, among whom 1138 patients had RVSPE or SADPE. Artificial intelligence deep neural network predicted acute PE with a modest accuracy of AUROC of 0.69 (95% CI, 0.68-0.71), sensitivity of 63.5%, specificity of 64.7%, PPV of 15.6%, and NPV of 94.5%. The AI-DNN prediction using the same algorithm for RVSPE or SADPE was higher (AUROC, 0.84; 95% CI, 0.81-0.86) with a sensitivity of 80.8%, specificity of 64.7.8%, PPV of 3.5%, and NPV of 99.5%. Conclusion An AI-based analysis of 12-lead ECG shows modest detection power for acute PE in patients who underwent CTPA, with higher accuracy for high-risk PE. Moreover, with the high NPV, it has the clinical potential to exclude high-risk PE quickly and correctly.
Collapse
Affiliation(s)
| | - Ryan A. Meverden
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - David M. Harmon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Kan Liu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Ana I. Casanegra
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Robert D. McBane
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
38
|
Poterucha TJ, Cheng S, Ouyang D. Exploring the Social Contributors to Biological Aging With Medical AI. JACC. ADVANCES 2024; 3:100889. [PMID: 39372463 PMCID: PMC11451070 DOI: 10.1016/j.jacadv.2024.100889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Affiliation(s)
- Timothy J. Poterucha
- Division of Cardiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David Ouyang
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
39
|
Takase B, Ikeda T, Shimizu W, Abe H, Aiba T, Chinushi M, Koba S, Kusano K, Niwano S, Takahashi N, Takatsuki S, Tanno K, Watanabe E, Yoshioka K, Amino M, Fujino T, Iwasaki YK, Kohno R, Kinoshita T, Kurita Y, Masaki N, Murata H, Shinohara T, Yada H, Yodogawa K, Kimura T, Kurita T, Nogami A, Sumitomo N. JCS/JHRS 2022 Guideline on Diagnosis and Risk Assessment of Arrhythmia. Circ J 2024; 88:1509-1595. [PMID: 37690816 DOI: 10.1253/circj.cj-22-0827] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Affiliation(s)
| | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Haruhiko Abe
- Department of Heart Rhythm Management, University of Occupational and Environmental Health, Japan
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center
| | - Masaomi Chinushi
- School of Health Sciences, Niigata University School of Medicine
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Shinichi Niwano
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Seiji Takatsuki
- Department of Cardiology, Keio University School of Medicine
| | - Kaoru Tanno
- Cardiology Division, Cardiovascular Center, Showa University Koto-Toyosu Hospital
| | - Eiichi Watanabe
- Division of Cardiology, Department of Internal Medicine, Fujita Health University Bantane Hospital
| | | | - Mari Amino
- Department of Cardiology, Tokai University School of Medicine
| | - Tadashi Fujino
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Yu-Ki Iwasaki
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Ritsuko Kohno
- Department of Heart Rhythm Management, University of Occupational and Environmental Health, Japan
| | - Toshio Kinoshita
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Yasuo Kurita
- Cardiovascular Center, International University of Health and Welfare, Mita Hospital
| | - Nobuyuki Masaki
- Department of Intensive Care Medicine, National Defense Medical College
| | | | - Tetsuji Shinohara
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hirotaka Yada
- Department of Cardiology, International University of Health and Welfare, Mita Hospital
| | - Kenji Yodogawa
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Takeshi Kimura
- Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | | | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba
| | - Naokata Sumitomo
- Department of Pediatric Cardiology, Saitama Medical University International Medical Center
| |
Collapse
|
40
|
Barros A, German Mesner I, Nguyen NR, Moorman JR. Age prediction from 12-lead electrocardiograms using deep learning: a comparison of four models on a contemporary, freely available dataset. Physiol Meas 2024; 45:08NT01. [PMID: 39048099 PMCID: PMC11334242 DOI: 10.1088/1361-6579/ad6746] [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: 02/27/2024] [Revised: 06/05/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024]
Abstract
Objective.The 12-lead electrocardiogram (ECG) is routine in clinical use and deep learning approaches have been shown to have the identify features not immediately apparent to human interpreters including age and sex. Several models have been published but no direct comparisons exist.Approach.We implemented three previously published models and one unpublished model to predict age and sex from a 12-lead ECG and then compared their performance on an open-access data set.Main results.All models converged and were evaluated on the holdout set. The best preforming age prediction model had a hold-out set mean absolute error of 8.06 years. The best preforming sex prediction model had a hold-out set area under the receiver operating curve of 0.92.Significance.We compared performance of four models on an open-access dataset.
Collapse
Affiliation(s)
- Andrew Barros
- Center for Advanced Medical Analytics (CAMA), School of Medicine, University of Virginia, Charlottesville, VA, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - Ian German Mesner
- Center for Advanced Medical Analytics (CAMA), School of Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - N Rich Nguyen
- Center for Advanced Medical Analytics (CAMA), School of Medicine, University of Virginia, Charlottesville, VA, United States of America
- Department of Computer Science, University of Virginia, Charlottesville, VA, United States of America
| | - J Randall Moorman
- Center for Advanced Medical Analytics (CAMA), School of Medicine, University of Virginia, Charlottesville, VA, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA, United States of America
| |
Collapse
|
41
|
Petzl AM, Jabbour G, Cadrin-Tourigny J, Pürerfellner H, Macle L, Khairy P, Avram R, Tadros R. Innovative approaches to atrial fibrillation prediction: should polygenic scores and machine learning be implemented in clinical practice? Europace 2024; 26:euae201. [PMID: 39073570 PMCID: PMC11332604 DOI: 10.1093/europace/euae201] [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: 07/02/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Atrial fibrillation (AF) prediction and screening are of important clinical interest because of the potential to prevent serious adverse events. Devices capable of detecting short episodes of arrhythmia are now widely available. Although it has recently been suggested that some high-risk patients with AF detected on implantable devices may benefit from anticoagulation, long-term management remains challenging in lower-risk patients and in those with AF detected on monitors or wearable devices as the development of clinically meaningful arrhythmia burden in this group remains unknown. Identification and prediction of clinically relevant AF is therefore of unprecedented importance to the cardiologic community. Family history and underlying genetic markers are important risk factors for AF. Recent studies suggest a good predictive ability of polygenic risk scores, with a possible additive value to clinical AF prediction scores. Artificial intelligence, enabled by the exponentially increasing computing power and digital data sets, has gained traction in the past decade and is of increasing interest in AF prediction using a single or multiple lead sinus rhythm electrocardiogram. Integrating these novel approaches could help predict AF substrate severity, thereby potentially improving the effectiveness of AF screening and personalizing the management of patients presenting with conditions such as embolic stroke of undetermined source or subclinical AF. This review presents current evidence surrounding deep learning and polygenic risk scores in the prediction of incident AF and provides a futuristic outlook on possible ways of implementing these modalities into clinical practice, while considering current limitations and required areas of improvement.
Collapse
Affiliation(s)
- Adrian M Petzl
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Gilbert Jabbour
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Canada
| | - Julia Cadrin-Tourigny
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Helmut Pürerfellner
- Department of Internal Medicine 2/Cardiology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Laurent Macle
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Paul Khairy
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Robert Avram
- Heartwise (heartwise.ai), Montreal Heart Institute, Montreal, Canada
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - Rafik Tadros
- Electrophysiology Service, Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
- Cardiovascular Genetics Center, Montreal Heart Institute, Université de Montréal, 5000 rue Bélanger, Montreal, QC H1T 1C8, Canada
| |
Collapse
|
42
|
Grenne B, Østvik A. Beyond Years: Is Artificial Intelligence Ready to Predict Biological Age and Cardiovascular Risk Using Echocardiography? J Am Soc Echocardiogr 2024; 37:736-739. [PMID: 38797330 DOI: 10.1016/j.echo.2024.05.013] [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: 05/18/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Affiliation(s)
- Bjørnar Grenne
- Clinic of Cardiology, St. Olav's University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Andreas Østvik
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Medical Image Analysis, Health Research, SINTEF Digital, Trondheim, Norway
| |
Collapse
|
43
|
Faierstein K, Fiman M, Loutati R, Rubin N, Manor U, Am-Shalom A, Cohen-Shelly M, Blank N, Lotan D, Zhao Q, Schwammenthal E, Klempfner R, Zimlichman E, Raanani E, Maor E. Artificial Intelligence Assessment of Biological Age From Transthoracic Echocardiography: Discrepancies with Chronologic Age Predict Significant Excess Mortality. J Am Soc Echocardiogr 2024; 37:725-735. [PMID: 38740271 DOI: 10.1016/j.echo.2024.04.017] [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: 03/04/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Age and sex can be estimated using artificial intelligence on the basis of various sources. The aims of this study were to test whether convolutional neural networks could be trained to estimate age and predict sex using standard transthoracic echocardiography and to evaluate the prognostic implications. METHODS The algorithm was trained on 76,342 patients, validated in 22,825 patients, and tested in 20,960 patients. It was then externally validated using data from a different hospital (n = 556). Finally, a prospective cohort of handheld point-of-care ultrasound devices (n = 319; ClinicalTrials.gov identifier NCT05455541) was used to confirm the findings. A multivariate Cox regression model was used to investigate the association between age estimation and chronologic age with overall survival. RESULTS The mean absolute error in age estimation was 4.9 years, with a Pearson correlation coefficient of 0.922. The probabilistic value of sex had an overall accuracy of 96.1% and an area under the curve of 0.993. External validation and prospective study cohorts yielded consistent results. Finally, survival analysis demonstrated that age prediction ≥5 years vs chronologic age was associated with an independent 34% increased risk for death during follow-up (P < .001). CONCLUSIONS Applying artificial intelligence to standard transthoracic echocardiography allows the prediction of sex and the estimation of age. Machine-based estimation is an independent predictor of overall survival and, with further evaluation, can be used for risk stratification and estimation of biological age.
Collapse
Affiliation(s)
- Kobi Faierstein
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | | | - Ranel Loutati
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Uri Manor
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Nimrod Blank
- Echocardiography Unit, Division of Cardiovascular Medicine, Baruch-Padeh Medical Center, Poria, Israel
| | - Dor Lotan
- Division of Cardiology, Department of Medicine, NewYork-Presbyterian Hospital, Columbia University Irving Medical Center, New York, New York
| | - Qiong Zhao
- Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, Virginia
| | - Ehud Schwammenthal
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Aisap.ai, Ramat Gan, Israel
| | - Robert Klempfner
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Aisap.ai, Ramat Gan, Israel
| | - Eyal Zimlichman
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ehud Raanani
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Aisap.ai, Ramat Gan, Israel
| | - Elad Maor
- Leviev Cardiovascular Institute, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Aisap.ai, Ramat Gan, Israel
| |
Collapse
|
44
|
Takase B, Ikeda T, Shimizu W, Abe H, Aiba T, Chinushi M, Koba S, Kusano K, Niwano S, Takahashi N, Takatsuki S, Tanno K, Watanabe E, Yoshioka K, Amino M, Fujino T, Iwasaki Y, Kohno R, Kinoshita T, Kurita Y, Masaki N, Murata H, Shinohara T, Yada H, Yodogawa K, Kimura T, Kurita T, Nogami A, Sumitomo N. JCS/JHRS 2022 Guideline on Diagnosis and Risk Assessment of Arrhythmia. J Arrhythm 2024; 40:655-752. [PMID: 39139890 PMCID: PMC11317726 DOI: 10.1002/joa3.13052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024] Open
Affiliation(s)
| | - Takanori Ikeda
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular MedicineNippon Medical School
| | - Haruhiko Abe
- Department of Heart Rhythm ManagementUniversity of Occupational and Environmental HealthJapan
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and GeneticsNational Cerebral and Cardiovascular Center
| | | | - Shinji Koba
- Division of Cardiology, Department of MedicineShowa University School of Medicine
| | - Kengo Kusano
- Department of Cardiovascular MedicineNational Cerebral and Cardiovascular Center
| | - Shinichi Niwano
- Department of Cardiovascular MedicineKitasato University School of Medicine
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of MedicineOita University
| | | | - Kaoru Tanno
- Cardiovascular Center, Cardiology DivisionShowa University Koto‐Toyosu Hospital
| | - Eiichi Watanabe
- Division of Cardiology, Department of Internal MedicineFujita Health University Bantane Hospital
| | | | - Mari Amino
- Department of CardiologyTokai University School of Medicine
| | - Tadashi Fujino
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Yu‐ki Iwasaki
- Department of Cardiovascular MedicineNippon Medical School
| | - Ritsuko Kohno
- Department of Heart Rhythm ManagementUniversity of Occupational and Environmental HealthJapan
| | - Toshio Kinoshita
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Yasuo Kurita
- Cardiovascular Center, Mita HospitalInternational University of Health and Welfare
| | - Nobuyuki Masaki
- Department of Intensive Care MedicineNational Defense Medical College
| | | | - Tetsuji Shinohara
- Department of Cardiology and Clinical Examination, Faculty of MedicineOita University
| | - Hirotaka Yada
- Department of CardiologyInternational University of Health and Welfare Mita Hospital
| | - Kenji Yodogawa
- Department of Cardiovascular MedicineNippon Medical School
| | - Takeshi Kimura
- Cardiovascular MedicineKyoto University Graduate School of Medicine
| | | | - Akihiko Nogami
- Department of Cardiology, Faculty of MedicineUniversity of Tsukuba
| | - Naokata Sumitomo
- Department of Pediatric CardiologySaitama Medical University International Medical Center
| | | |
Collapse
|
45
|
Barros A, German-Mesner I, Rich Nguyen N, Moorman JR. Age Prediction From 12-lead Electrocardiograms Using Deep Learning: A Comparison of Four Models on a Contemporary, Freely Available Dataset. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.02.24302201. [PMID: 38352374 PMCID: PMC10862990 DOI: 10.1101/2024.02.02.24302201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Objective The 12-lead electrocardiogram (ECG) is routine in clinical use and deep learning approaches have been shown to have the identify features not immediately apparent to human interpreters including age and sex. Several models have been published but no direct comparisons exist. Approach We implemented three previously published models and one unpublished model to predict age and sex from a 12-lead ECG and then compared their performance on an open-access data set. Main results All models converged and were evaluated on the holdout set. The best preforming age prediction model had a hold-out set mean absolute error of 8.06 years. The best preforming sex prediction model had a hold-out set area under the receiver operating curve of 0.92. Significance We compared performance of four models on an open-access dataset.
Collapse
|
46
|
Ansari MY, Qaraqe M, Righetti R, Serpedin E, Qaraqe K. Enhancing ECG-based heart age: impact of acquisition parameters and generalization strategies for varying signal morphologies and corruptions. Front Cardiovasc Med 2024; 11:1424585. [PMID: 39027006 PMCID: PMC11254851 DOI: 10.3389/fcvm.2024.1424585] [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: 04/28/2024] [Accepted: 06/04/2024] [Indexed: 07/20/2024] Open
Abstract
Electrocardiogram (ECG) is a non-invasive approach to capture the overall electrical activity produced by the contraction and relaxation of the cardiac muscles. It has been established in the literature that the difference between ECG-derived age and chronological age represents a general measure of cardiovascular health. Elevated ECG-derived age strongly correlates with cardiovascular conditions (e.g., atherosclerotic cardiovascular disease). However, the neural networks for ECG age estimation are yet to be thoroughly evaluated from the perspective of ECG acquisition parameters. Additionally, deep learning systems for ECG analysis encounter challenges in generalizing across diverse ECG morphologies in various ethnic groups and are susceptible to errors with signals that exhibit random or systematic distortions To address these challenges, we perform a comprehensive empirical study to determine the threshold for the sampling rate and duration of ECG signals while considering their impact on the computational cost of the neural networks. To tackle the concern of ECG waveform variability in different populations, we evaluate the feasibility of utilizing pre-trained and fine-tuned networks to estimate ECG age in different ethnic groups. Additionally, we empirically demonstrate that finetuning is an environmentally sustainable way to train neural networks, and it significantly decreases the ECG instances required (by more than 100 × ) for attaining performance similar to the networks trained from random weight initialization on a complete dataset. Finally, we systematically evaluate augmentation schemes for ECG signals in the context of age estimation and introduce a random cropping scheme that provides best-in-class performance while using shorter-duration ECG signals. The results also show that random cropping enables the networks to perform well with systematic and random ECG signal corruptions.
Collapse
Affiliation(s)
- Mohammed Yusuf Ansari
- Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
- Electrical and Computer Engineering, Texas A&M University at Qatar, Doha, Qatar
| | - Marwa Qaraqe
- Electrical and Computer Engineering, Texas A&M University at Qatar, Doha, Qatar
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Raffaella Righetti
- Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Erchin Serpedin
- Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Khalid Qaraqe
- Electrical and Computer Engineering, Texas A&M University at Qatar, Doha, Qatar
| |
Collapse
|
47
|
Khera R, Oikonomou EK, Nadkarni GN, Morley JR, Wiens J, Butte AJ, Topol EJ. Transforming Cardiovascular Care With Artificial Intelligence: From Discovery to Practice: JACC State-of-the-Art Review. J Am Coll Cardiol 2024; 84:97-114. [PMID: 38925729 DOI: 10.1016/j.jacc.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024]
Abstract
Artificial intelligence (AI) has the potential to transform every facet of cardiovascular practice and research. The exponential rise in technology powered by AI is defining new frontiers in cardiovascular care, with innovations that span novel diagnostic modalities, new digital native biomarkers of disease, and high-performing tools evaluating care quality and prognosticating clinical outcomes. These digital innovations promise expanded access to cardiovascular screening and monitoring, especially among those without access to high-quality, specialized care historically. Moreover, AI is propelling biological and clinical discoveries that will make future cardiovascular care more personalized, precise, and effective. The review brings together these diverse AI innovations, highlighting developments in multimodal cardiovascular AI across clinical practice and biomedical discovery, and envisioning this new future backed by contemporary science and emerging discoveries. Finally, we define the critical path and the safeguards essential to realizing this AI-enabled future that helps achieve optimal cardiovascular health and outcomes for all.
Collapse
Affiliation(s)
- Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA; Center for Outcomes Research and Evaluation (CORE), New Haven, Connecticut, USA; Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Connecticut, USA; Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA.
| | - Evangelos K Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Girish N Nadkarni
- The Samuel Bronfman Department of Medicine, Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, New York, USA; The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica R Morley
- Digital Ethics Center, Yale University, New Haven, Connecticut, USA
| | - Jenna Wiens
- Electrical Engineering and Computer Science, Computer Science and Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA; Center for Data-Driven Insights and Innovation, University of California Health, Oakland, California, USA
| | - Eric J Topol
- Molecular Medicine, Scripps Research Translational Institute, Scripps Research, La Jolla, California, USA
| |
Collapse
|
48
|
O'Sullivan D, Anjewierden S, Greason G, Attia IZ, Lopez-Jimenez F, Friedman PA, Noseworthy P, Anderson J, Kashou A, Asirvatham SJ, Eidem BW, Johnson JN, Niaz T, Madhavan M. Pediatric sex estimation using AI-enabled ECG analysis: influence of pubertal development. NPJ Digit Med 2024; 7:176. [PMID: 38956410 PMCID: PMC11220019 DOI: 10.1038/s41746-024-01165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
AI-enabled ECGs have previously been shown to accurately predict patient sex in adults and correlate with sex hormone levels. We aimed to test the ability of AI-enabled ECGs to predict sex in the pediatric population and study the influence of pubertal development. AI-enabled ECG models were created using a convolutional neural network trained on pediatric 10-second, 12-lead ECGs. The first model was trained de novo using pediatric data. The second model used transfer learning from a previously validated adult data-derived algorithm. We analyzed the first ECG from 90,133 unique pediatric patients (aged ≤18 years) recorded between 1987-2022, and divided the cohort into training, validation, and testing datasets. Subgroup analysis was performed on prepubertal (0-7 years), peripubertal (8-14 years), and postpubertal (15-18 years) patients. The cohort was 46.7% male, with 21,678 prepubertal, 26,740 peripubertal, and 41,715 postpubertal children. The de novo pediatric model demonstrated 81% accuracy and an area under the curve (AUC) of 0.91. Model sensitivity was 0.79, specificity was 0.83, positive predicted value was 0.84, and the negative predicted value was 0.78, for the entire test cohort. The model's discriminatory ability was highest in postpubertal (AUC = 0.98), lower in the peripubertal age group (AUC = 0.91), and poor in the prepubertal age group (AUC = 0.67). There was no significant performance difference observed between the transfer learning and de novo models. AI-enabled interpretation of ECG can estimate sex in peripubertal and postpubertal children with high accuracy.
Collapse
Affiliation(s)
- Donnchadh O'Sullivan
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA.
| | - Scott Anjewierden
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Grace Greason
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Paul A Friedman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Peter Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jason Anderson
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Anthony Kashou
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Benjamin W Eidem
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jonathan N Johnson
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Talha Niaz
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Malini Madhavan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
49
|
Strodthoff N, Lopez Alcaraz JM, Haverkamp W. Prospects for artificial intelligence-enhanced electrocardiogram as a unified screening tool for cardiac and non-cardiac conditions: an explorative study in emergency care. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2024; 5:454-460. [PMID: 39081937 PMCID: PMC11284007 DOI: 10.1093/ehjdh/ztae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 08/02/2024]
Abstract
Aims Current deep learning algorithms for automatic ECG analysis have shown notable accuracy but are typically narrowly focused on singular diagnostic conditions. This exploratory study aims to investigate the capability of a single deep learning model to predict a diverse range of both cardiac and non-cardiac discharge diagnoses based on a single ECG collected in the emergency department. Methods and results In this study, we assess the performance of a model trained to predict a broad spectrum of diagnoses. We find that the model can reliably predict 253 ICD codes (81 cardiac and 172 non-cardiac) in the sense of exceeding an AUROC score of 0.8 in a statistically significant manner. Conclusion The model demonstrates proficiency in handling a wide array of cardiac and non-cardiac diagnostic scenarios, indicating its potential as a comprehensive screening tool for diverse medical encounters.
Collapse
Affiliation(s)
- Nils Strodthoff
- Carl von Ossietzky Universität Oldenburg, School VI Medicine and Health Services, Department of Health Services Research, Ammerländer Heerstr. 114-118, 26129 Oldenburg, Germany
| | - Juan Miguel Lopez Alcaraz
- Carl von Ossietzky Universität Oldenburg, School VI Medicine and Health Services, Department of Health Services Research, Ammerländer Heerstr. 114-118, 26129 Oldenburg, Germany
| | - Wilhelm Haverkamp
- Charité Universitätsmedizin Berlin, Department of Cardiology and Metabolism, Clinic for Cardiology, Angiology, and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
50
|
Sharma V, Ghose A. BioAgeNet: An Age-Informed Convolutional Autoencoder for ECG Clustering Indicating Health. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039457 DOI: 10.1109/embc53108.2024.10781506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological Age (BA) indicates the authentic ageing progression of an individual in relation to their quality of life. The noninvasive identification of BA is crucial in predicting longevity and early age-related diseases and enabling personalized healthcare. Potential biomarkers of BA are vague and need attention. The ageing process stands out as a prominent risk factor for cardiovascular diseases. Consequently, an Electrocardiogram (ECG), the most popular and easily accessible signal, is explored to analyze the effect of age. Numerous studies have delved into supervised deep-learning approaches for ECG analysis, particularly in predicting age. These studies rely on regression-based methods and necessitate additional analysis for extracting health-related insights, such as the correlation of error between Chronological Age and AI-predicted Age with mortality. Moreover, as the shortage of cardiologists' annotated data is apparent, we propose an Age-Informed Convolutional Autoencoder that clusters ECG deep features associated with age to assess the quality of life possessed at the current age. We also proposed a three-step training strategy combining model training and deep ECG features clustering with a controlled initialization. We find that a combination of age and ECG reveals the heart's BA and is a contributing biomarker for estimating the overall BA of the body. This approach marks substantial progress in analyzing age-related impacts on ECG. It provides new perspectives on different cardiovascular disorders and can potentially transform personalized healthcare in the future.
Collapse
|