151
|
Lareyre F, Behrendt CA, Chaudhuri A, Lee R, Carrier M, Adam C, Lê CD, Raffort J. Applications of artificial intelligence for patients with peripheral artery disease. J Vasc Surg 2023; 77:650-658.e1. [PMID: 35921995 DOI: 10.1016/j.jvs.2022.07.160] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/06/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Applications of artificial intelligence (AI) have been reported in several cardiovascular diseases but its interest in patients with peripheral artery disease (PAD) has been so far less reported. The aim of this review was to summarize current knowledge on applications of AI in patients with PAD, to discuss current limits, and highlight perspectives in the field. METHODS We performed a narrative review based on studies reporting applications of AI in patients with PAD. The MEDLINE database was independently searched by two authors using a combination of keywords to identify studies published between January 1995 and December 2021. Three main fields of AI were investigated including natural language processing (NLP), computer vision and machine learning (ML). RESULTS NLP and ML brought new tools to improve the screening, the diagnosis and classification of the severity of PAD. ML was also used to develop predictive models to better assess the prognosis of patients and develop real-time prediction models to support clinical decision-making. Studies related to computer vision mainly aimed at creating automatic detection and characterization of arterial lesions based on Doppler ultrasound examination or computed tomography angiography. Such tools could help to improve screening programs, enhance diagnosis, facilitate presurgical planning, and improve clinical workflow. CONCLUSIONS AI offers various applications to support and likely improve the management of patients with PAD. Further research efforts are needed to validate such applications and investigate their accuracy and safety in large multinational cohorts before their implementation in daily clinical practice.
Collapse
Affiliation(s)
- Fabien Lareyre
- Department of Vascular Surgery, Hospital of Antibes Juan-les-Pins, Antibes, France; Université Côte d'Azur, INSERM U1065, C3M, Nice, France.
| | - Christian-Alexander Behrendt
- Research Group GermanVasc, Department of Vascular Medicine, University Heart and Vascular Centre UKE Hamburg, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Arindam Chaudhuri
- Bedfordshire-Milton Keynes Vascular Centre, Bedfordshire Hospitals NHS Foundation Trust, Bedford, UK
| | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Marion Carrier
- Laboratory of Applied Mathematics and Computer Science (MICS), CentraleSupélec, Université Paris-Saclay, Paris, France
| | - Cédric Adam
- Laboratory of Applied Mathematics and Computer Science (MICS), CentraleSupélec, Université Paris-Saclay, Paris, France
| | - Cong Duy Lê
- Department of Vascular Surgery, Hospital of Antibes Juan-les-Pins, Antibes, France; Université Côte d'Azur, INSERM U1065, C3M, Nice, France
| | - Juliette Raffort
- Université Côte d'Azur, INSERM U1065, C3M, Nice, France; Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France; AI Institute 3IA Côte d'Azur, Université Côte d'Azur, Côte d'Azur, France
| |
Collapse
|
152
|
Moradi H, Al-Hourani A, Concilia G, Khoshmanesh F, Nezami FR, Needham S, Baratchi S, Khoshmanesh K. Recent developments in modeling, imaging, and monitoring of cardiovascular diseases using machine learning. Biophys Rev 2023; 15:19-33. [PMID: 36909958 PMCID: PMC9995635 DOI: 10.1007/s12551-022-01040-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are the leading cause of mortality, morbidity, and hospitalization around the world. Recent technological advances have facilitated analyzing, visualizing, and monitoring cardiovascular diseases using emerging computational fluid dynamics, blood flow imaging, and wearable sensing technologies. Yet, computational cost, limited spatiotemporal resolution, and obstacles for thorough data analysis have hindered the utility of such techniques to curb cardiovascular diseases. We herein discuss how leveraging machine learning techniques, and in particular deep learning methods, could overcome these limitations and offer promise for translation. We discuss the remarkable capacity of recently developed machine learning techniques to accelerate flow modeling, enhance the resolution while reduce the noise and scanning time of current blood flow imaging techniques, and accurate detection of cardiovascular diseases using a plethora of data collected by wearable sensors.
Collapse
Affiliation(s)
- Hamed Moradi
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Akram Al-Hourani
- School of Engineering, RMIT University, Melbourne, Victoria Australia
| | | | - Farnaz Khoshmanesh
- School of Allied Health, Human Services & Sport, La Trobe University, Melbourne, Victoria Australia
| | - Farhad R. Nezami
- Division of Thoracic and Cardiac Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Scott Needham
- Leading Technology Group, Melbourne, Victoria Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria Australia
| | | |
Collapse
|
153
|
Singh A, Miller RJH, Otaki Y, Kavanagh P, Hauser MT, Tzolos E, Kwiecinski J, Van Kriekinge S, Wei CC, Sharir T, Einstein AJ, Fish MB, Ruddy TD, Kaufmann PA, Sinusas AJ, Miller EJ, Bateman TM, Dorbala S, Di Carli M, Liang JX, Huang C, Han D, Dey D, Berman DS, Slomka PJ. Direct Risk Assessment From Myocardial Perfusion Imaging Using Explainable Deep Learning. JACC Cardiovasc Imaging 2023; 16:209-220. [PMID: 36274041 PMCID: PMC10980287 DOI: 10.1016/j.jcmg.2022.07.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/21/2022] [Accepted: 07/21/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Myocardial perfusion imaging (MPI) is frequently used to provide risk stratification, but methods to improve the accuracy of these predictions are needed. OBJECTIVES The authors developed an explainable deep learning (DL) model (HARD MACE [major adverse cardiac events]-DL) for the prediction of death or nonfatal myocardial infarction (MI) and validated its performance in large internal and external testing groups. METHODS Patients undergoing single-photon emission computed tomography MPI were included, with 20,401 patients in the training and internal testing group (5 sites) and 9,019 in the external testing group (2 different sites). HARD MACE-DL uses myocardial perfusion, motion, thickening, and phase polar maps combined with age, sex, and cardiac volumes. The primary outcome was all-cause mortality or nonfatal MI. Prognostic accuracy was evaluated using area under the receiver-operating characteristic curve (AUC). RESULTS During internal testing, patients with normal perfusion and elevated HARD MACE-DL risk were at higher risk than patients with abnormal perfusion and low HARD MACE-DL risk (annualized event rate, 2.9% vs 1.2%; P < 0.001). Patients in the highest quartile of HARD MACE-DL score had an annual rate of death or MI (4.8%) 10-fold higher than patients in the lowest quartile (0.48% per year). In external testing, the AUC for HARD MACE-DL (0.73; 95% CI: 0.71-0.75) was higher than a logistic regression model (AUC: 0.70), stress total perfusion deficit (TPD) (AUC: 0.65), and ischemic TPD (AUC: 0.63; all P < 0.01). Calibration, a measure of how well predicted risk matches actual risk, was excellent in both groups (Brier score, 0.079 for internal and 0.070 for external). CONCLUSIONS The DL model predicts death or MI directly from MPI, by estimating patient-level risk with good calibration and improved accuracy compared with traditional quantitative approaches. The model incorporates mechanisms to explain to the physician which image regions contribute to the adverse event prediction.
Collapse
Affiliation(s)
- Ananya Singh
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert J H Miller
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Cardiac Sciences, University of Calgary and Libin Cardiovascular Institute, Calgary, Alberta, Canada
| | - Yuka Otaki
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paul Kavanagh
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael T Hauser
- Department of Nuclear Cardiology, Oklahoma Heart Hospital, Oklahoma City, Oklahoma, USA
| | - Evangelos Tzolos
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA; BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Jacek Kwiecinski
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | - Serge Van Kriekinge
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chih-Chun Wei
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tali Sharir
- Department of Nuclear Cardiology, Assuta Medical Center, Tel Aviv, Israel; Department of Nuclear Cardiology, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Andrew J Einstein
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, New York, USA; Department of Radiology, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, New York, USA
| | - Mathews B Fish
- Oregon Heart and Vascular Institute, Sacred Heart Medical Center, Springfield, Oregon, USA
| | - Terrence D Ruddy
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Philipp A Kaufmann
- Division of Cardiac Imaging, Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Albert J Sinusas
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Sharmila Dorbala
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Marcelo Di Carli
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Joanna X Liang
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Cathleen Huang
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Donghee Han
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Damini Dey
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniel S Berman
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Piotr J Slomka
- Division of Artificial Intelligence in Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA; Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
154
|
Kampaktsis PN, Emfietzoglou M, Al Shehhi A, Fasoula NA, Bakogiannis C, Mouselimis D, Tsarouchas A, Vassilikos VP, Kallmayer M, Eckstein HH, Hadjileontiadis L, Karlas A. Artificial intelligence in atherosclerotic disease: Applications and trends. Front Cardiovasc Med 2023; 9:949454. [PMID: 36741834 PMCID: PMC9896100 DOI: 10.3389/fcvm.2022.949454] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most common cause of death globally. Increasing amounts of highly diverse ASCVD data are becoming available and artificial intelligence (AI) techniques now bear the promise of utilizing them to improve diagnosis, advance understanding of disease pathogenesis, enable outcome prediction, assist with clinical decision making and promote precision medicine approaches. Machine learning (ML) algorithms in particular, are already employed in cardiovascular imaging applications to facilitate automated disease detection and experts believe that ML will transform the field in the coming years. Current review first describes the key concepts of AI applications from a clinical standpoint. We then provide a focused overview of current AI applications in four main ASCVD domains: coronary artery disease (CAD), peripheral arterial disease (PAD), abdominal aortic aneurysm (AAA), and carotid artery disease. For each domain, applications are presented with refer to the primary imaging modality used [e.g., computed tomography (CT) or invasive angiography] and the key aim of the applied AI approaches, which include disease detection, phenotyping, outcome prediction, and assistance with clinical decision making. We conclude with the strengths and limitations of AI applications and provide future perspectives.
Collapse
Affiliation(s)
- Polydoros N. Kampaktsis
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY, United States,*Correspondence: Polydoros N. Kampaktsis,
| | - Maria Emfietzoglou
- Heart Centre, John Radcliffe Hospital, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Aamna Al Shehhi
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nikolina-Alexia Fasoula
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany,School of Medicine, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Constantinos Bakogiannis
- Third Department of Cardiology, Hippokration University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Mouselimis
- Third Department of Cardiology, Hippokration University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasios Tsarouchas
- Third Department of Cardiology, Hippokration University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios P. Vassilikos
- Third Department of Cardiology, Hippokration University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michael Kallmayer
- Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Leontios Hadjileontiadis
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Healthcare Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Department of Electrical and Computer Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Angelos Karlas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany,School of Medicine, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany,Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
155
|
Kharlamov A, Lamberts M. Digital medicine: the next big leap advancing cardiovascular science. BMC Cardiovasc Disord 2023; 23:30. [PMID: 36650433 PMCID: PMC9847174 DOI: 10.1186/s12872-022-02971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 01/19/2023] Open
Abstract
Solid clinical and academic leadership remains necessary to ensure that healthcare based on digital technologies is relevant, meaningful, and stands on the best possible evidence. This compendium accompanying the "Digital Technologies in Cardiovascular Disorders" article collection in BMC Cardiovascular Disorders summarizes recent knowledge about robust and advanced digital tools for preventing, monitoring, diagnosing, and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Alexander Kharlamov
- Advanced Cardiovascular Imaging Lab, De Haar Research Foundation (DHRF), Tallinn, Estonia ,Innovation Lab, De Haar Research Task Force, Rotterdam, The Netherlands ,DHRF, Keurenplein 41, G9950, 1069 CD Amsterdam, The Netherlands
| | - Morten Lamberts
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark ,grid.411646.00000 0004 0646 7402Department of Cardiology, Herlev-Gentofte University Hospital, Herlev, Denmark
| |
Collapse
|
156
|
D'Ancona G, Massussi M, Savardi M, Signoroni A, Di Bacco L, Farina D, Metra M, Maroldi R, Muneretto C, Ince H, Costabile D, Murero M, Chizzola G, Curello S, Benussi S. Deep learning to detect significant coronary artery disease from plain chest radiographs AI4CAD. Int J Cardiol 2023; 370:435-441. [PMID: 36343794 DOI: 10.1016/j.ijcard.2022.10.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The predictive role of chest radiographs in patients with suspected coronary artery disease (CAD) is underestimated and may benefit from artificial intelligence (AI) applications. OBJECTIVES To train, test, and validate a deep learning (DL) solution for detecting significant CAD based on chest radiographs. METHODS Data of patients referred for angina and undergoing chest radiography and coronary angiography were analysed retrospectively. A deep convolutional neural network (DCNN) was designed to detect significant CAD from posteroanterior/anteroposterior chest radiographs. The DCNN was trained for severe CAD binary classification (absence/presence). Coronary angiography reports were the ground truth. Stenosis severity of ≥70% for non-left main vessels and ≥ 50% for left main defined severe CAD. RESULTS Information of 7728 patients was reviewed. Severe CAD was present in 4091 (53%). Patients were randomly divided for algorithm training (70%; n = 5454) and fine-tuning/model validation (10%; n = 773). Internal clinical validation (model testing) was performed with the remaining patients (20%; n = 1501). At binary logistic regression, DCNN prediction was the strongest severe CAD predictor (p < 0.0001; OR: 1.040; CI: 1.032-1.048). Using a high sensitivity operating cut-point, the DCNN had a sensitivity of 0.90 to detect significant CAD (specificity 0.31; AUC 0.73; 95% CI DeLong, 0.69-0.76). Adding to the AI chest radiograph interpretation angina status improved the prediction (AUC 0.77; 95% CI DeLong, 0.74-0.80). CONCLUSION AI-read chest radiographs could be used to pre-test significant CAD probability in patients referred for suspected angina. Further studies are required to externally validate our algorithm, develop a clinically applicable tool, and support CAD screening in broader settings.
Collapse
Affiliation(s)
- Giuseppe D'Ancona
- Department of Cardiology and Cardiovascular Clinical Research Unit, Vivantes Klinikum Urban and Neukölln, Berlin, Germany.
| | - Mauro Massussi
- Cardiac Catheterization Laboratory and Cardiology, ASST Spedali Civili and Department Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Mattia Savardi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, and Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Alberto Signoroni
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, and Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Lorenzo Di Bacco
- Department of Cardiac Surgery, Spedali Civili Brescia and University of Brescia, Brescia, Italy
| | - Davide Farina
- Radiology 2, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Marco Metra
- Cardiac Catheterization Laboratory and Cardiology, ASST Spedali Civili and Department Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Roberto Maroldi
- Radiology 2, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Claudio Muneretto
- Department of Cardiac Surgery, Spedali Civili Brescia and University of Brescia, Brescia, Italy
| | - Hüseyin Ince
- Department of Cardiology and Cardiovascular Clinical Research Unit, Vivantes Klinikum Urban and Neukölln, Berlin, Germany
| | - Davide Costabile
- Department of Information Technology Spedali Civili Brescia, Brescia, Italy
| | - Monica Murero
- AI4 Life and Society International Institute, Federico II University, Naples, Italy
| | - Giuliano Chizzola
- Cardiac Catheterization Laboratory and Cardiology, ASST Spedali Civili and Department Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Salvatore Curello
- Cardiac Catheterization Laboratory and Cardiology, ASST Spedali Civili and Department Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Italy
| | - Stefano Benussi
- Department of Cardiac Surgery, Spedali Civili Brescia and University of Brescia, Brescia, Italy
| |
Collapse
|
157
|
Motwani M, Williams MC, Nieman K, Choi AD. Great debates in cardiac computed tomography: OPINION: "Artificial intelligence is key to the future of CCTA - The great hope". J Cardiovasc Comput Tomogr 2023; 17:18-21. [PMID: 35945132 DOI: 10.1016/j.jcct.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/05/2022] [Accepted: 07/16/2022] [Indexed: 10/17/2022]
Affiliation(s)
- Manish Motwani
- Manchester Heart Institute, Manchester University NHS Foundation Trust, UK; Institute of Cardiovascular Science, University of Manchester, UK
| | - Michelle C Williams
- Center for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, UK
| | - Koen Nieman
- Departments of Cardiovascular Medicine and Radiology, Stanford University, Stanford, CA, USA
| | - Andrew D Choi
- Division of Cardiology and Department of Radiology, The George Washington University School of Medicine, Washington, DC, USA.
| |
Collapse
|
158
|
Walpot J, Van Herck P, Van de Heyning CM, Bosmans J, Massalha S, Malbrain ML, Heidbuchel H, Inácio JR. Computed tomography measured epicardial adipose tissue and psoas muscle attenuation: new biomarkers to predict major adverse cardiac events (MACE) and mortality in patients with heart disease and critically ill patients. Part I: Epicardial adipose tissue. Anaesthesiol Intensive Ther 2023; 55:141-157. [PMID: 37728441 PMCID: PMC10496106 DOI: 10.5114/ait.2023.130922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/28/2023] [Indexed: 09/21/2023] Open
Abstract
Over the last two decades, the potential role of epicardial adipocyte tissue (EAT) as a marker for major adverse cardiovascular events has been extensively studied. Unlike other visceral adipocyte tissues (VAT), EAT is not separated from the adjacent myocardium by a fascial layer and shares the same microcirculation with the myocardium. Adipocytokines, secreted by EAT, interact directly with the myocardium through paracrine and vasocrine pathways. The role of the Randle cycle, linking VAT accumulation to insulin resistance, and the relevance of blood flow and mitochondrial function of VAT, are briefly discussed. The three available imaging modalities for the assessment of EAT are discussed. The advantages of echocardiography, cardiac CT, and cardiac magnetic resonance (CMR) are compared. The last section summarises the current stage of knowledge on EAT as a clinical marker for major adverse cardiovascular events (MACE). The association between EAT volume and coronary artery disease (CAD) has robustly been validated. There is growing evidence that EAT volume is associated with computed tomography coronary angiography (CTCA) assessed high-risk plaque features. The EAT CT attenuation coefficient predicts coronary events. Many studies have established EAT volume as a predictor of atrial fibrillation after cardiac surgery. Moreover, EAT thickness has been independently associated with severe aortic stenosis and mitral annular calcification. Studies have demonstrated that EAT volume is associated with heart failure. Finally, we discuss the potential role of EAT in critically ill patients admitted to the intensive care unit. In conclusion, EAT seems to be a promising new biomarker to predict MACE.
Collapse
Affiliation(s)
| | - Paul Van Herck
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - Caroline M. Van de Heyning
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Bosmans
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Manu L.N.G. Malbrain
- International Fluid Academy, Lovenjoel, Belgium
- First Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, Lublin, Poland
| | - Hein Heidbuchel
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium
- Cardiovascular Sciences, University of Antwerp, Antwerp, Belgium
| | - João R. Inácio
- Centro Universitario Hospitalar Lisboa Norte, Faculdade de Medicina de Lisboa, UL, Portugal
| |
Collapse
|
159
|
Moreira A, Tovar M, Smith AM, Lee GC, Meunier JA, Cheema Z, Moreira A, Winter C, Mustafa SB, Seidner S, Findley T, Garcia JGN, Thébaud B, Kwinta P, Ahuja SK. Development of a peripheral blood transcriptomic gene signature to predict bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L76-L87. [PMID: 36472344 PMCID: PMC9829478 DOI: 10.1152/ajplung.00250.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung disease of extreme prematurity, yet mechanisms that associate with or identify neonates with increased susceptibility for BPD are largely unknown. Combining artificial intelligence with gene expression data is a novel approach that may assist in better understanding mechanisms underpinning chronic lung disease and in stratifying patients at greater risk for BPD. The objective of this study is to develop an early peripheral blood transcriptomic signature that can predict preterm neonates at risk for developing BPD. Secondary analysis of whole blood microarray data from 97 very low birth weight neonates on day of life 5 was performed. BPD was defined as positive pressure ventilation or oxygen requirement at 28 days of age. Participants were randomly assigned to a training (70%) and testing cohort (30%). Four gene-centric machine learning models were built, and their discriminatory abilities were compared with gestational age or birth weight. This study adheres to the transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD) statement. Neonates with BPD (n = 62 subjects) exhibited a lower median gestational age (26.0 wk vs. 30.0 wk, P < 0.01) and birth weight (800 g vs. 1,280 g, P < 0.01) compared with non-BPD neonates. From an initial pool (33,252 genes/patient), 4,523 genes exhibited a false discovery rate (FDR) <1%. The area under the receiver operating characteristic curve (AUC) for predicting BPD utilizing gestational age or birth weight was 87.8% and 87.2%, respectively. The machine learning models, using a combination of five genes, revealed AUCs ranging between 85.8% and 96.1%. Pathways integral to T cell development and differentiation were associated with BPD. A derived five-gene whole blood signature can accurately predict BPD in the first week of life.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Miriam Tovar
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Alisha M Smith
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Grace C Lee
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Justin A Meunier
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Zoya Cheema
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Axel Moreira
- Division of Critical Care, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Caitlyn Winter
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Shamimunisa B Mustafa
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Steven Seidner
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Tina Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, Texas
| | - Joe G N Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Przemko Kwinta
- Neonatal Intensive Care Unit, Department of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Sunil K Ahuja
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
160
|
Tomihama RT, Camara JR, Kiang SC. Machine learning analysis of confounding variables of a convolutional neural network specific for abdominal aortic aneurysms. JVS Vasc Sci 2023. [DOI: 10.1016/j.jvssci.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
161
|
Ihdayhid AR, Lan NSR, Williams M, Newby D, Flack J, Kwok S, Joyner J, Gera S, Dembo L, Adler B, Ko B, Chow BJW, Dwivedi G. Evaluation of an artificial intelligence coronary artery calcium scoring model from computed tomography. Eur Radiol 2023; 33:321-329. [PMID: 35986771 PMCID: PMC9755106 DOI: 10.1007/s00330-022-09028-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/07/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Coronary artery calcium (CAC) scores derived from computed tomography (CT) scans are used for cardiovascular risk stratification. Artificial intelligence (AI) can assist in CAC quantification and potentially reduce the time required for human analysis. This study aimed to develop and evaluate a fully automated model that identifies and quantifies CAC. METHODS Fully convolutional neural networks for automated CAC scoring were developed and trained on 2439 cardiac CT scans and validated using 771 scans. The model was tested on an independent set of 1849 cardiac CT scans. Agatston CAC scores were further categorised into five risk categories (0, 1-10, 11-100, 101-400, and > 400). Automated scores were compared to the manual reference standard (level 3 expert readers). RESULTS Of 1849 scans used for model testing (mean age 55.7 ± 10.5 years, 49% males), the automated model detected the presence of CAC in 867 (47%) scans compared with 815 (44%) by human readers (p = 0.09). CAC scores from the model correlated very strongly with the manual score (Spearman's r = 0.90, 95% confidence interval [CI] 0.89-0.91, p < 0.001 and intraclass correlation coefficient = 0.98, 95% CI 0.98-0.99, p < 0.001). The model classified 1646 (89%) into the same risk category as human observers. The Bland-Altman analysis demonstrated little difference (1.69, 95% limits of agreement: -41.22, 44.60) and there was almost excellent agreement (Cohen's κ = 0.90, 95% CI 0.88-0.91, p < 0.001). Model analysis time was 13.1 ± 3.2 s/scan. CONCLUSIONS This artificial intelligence-based fully automated CAC scoring model shows high accuracy and low analysis times. Its potential to optimise clinical workflow efficiency and patient outcomes requires evaluation. KEY POINTS • Coronary artery calcium (CAC) scores are traditionally assessed using cardiac computed tomography and require manual input by human operators to identify calcified lesions. • A novel artificial intelligence (AI)-based model for fully automated CAC scoring was developed and tested on an independent dataset of computed tomography scans, showing very high levels of correlation and agreement with manual measurements as a reference standard. • AI has the potential to assist in the identification and quantification of CAC, thereby reducing the time required for human analysis.
Collapse
Affiliation(s)
- Abdul Rahman Ihdayhid
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia.
- Harry Perkins Institute of Medical Research, Curtin University, Perth, Australia.
| | - Nick S R Lan
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Michelle Williams
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | - David Newby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, Scotland, UK
| | | | | | | | - Sahil Gera
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Lawrence Dembo
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia
- Envision Medical Imaging, Perth, Australia
| | | | - Brian Ko
- Monash Cardiovascular Research Centre, Monash University and MonashHeart, Monash Health, Melbourne, Australia
| | | | - Girish Dwivedi
- Department of Cardiology, Fiona Stanley Hospital, Perth, Australia.
- Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia.
| |
Collapse
|
162
|
Zhang Z, Wang Y, Zhang H, Samusak A, Rao H, Xiao C, Abula M, Cao Q, Dai Q. Artificial intelligence-assisted diagnosis of ocular surface diseases. Front Cell Dev Biol 2023; 11:1133680. [PMID: 36875760 PMCID: PMC9981656 DOI: 10.3389/fcell.2023.1133680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
With the rapid development of computer technology, the application of artificial intelligence (AI) in ophthalmology research has gained prominence in modern medicine. Artificial intelligence-related research in ophthalmology previously focused on the screening and diagnosis of fundus diseases, particularly diabetic retinopathy, age-related macular degeneration, and glaucoma. Since fundus images are relatively fixed, their standards are easy to unify. Artificial intelligence research related to ocular surface diseases has also increased. The main issue with research on ocular surface diseases is that the images involved are complex, with many modalities. Therefore, this review aims to summarize current artificial intelligence research and technologies used to diagnose ocular surface diseases such as pterygium, keratoconus, infectious keratitis, and dry eye to identify mature artificial intelligence models that are suitable for research of ocular surface diseases and potential algorithms that may be used in the future.
Collapse
Affiliation(s)
- Zuhui Zhang
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China.,National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying Wang
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Hongzhen Zhang
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Arzigul Samusak
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Huimin Rao
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Chun Xiao
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Muhetaer Abula
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China
| | - Qixin Cao
- Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou, China
| | - Qi Dai
- The First People's Hospital of Aksu District in Xinjiang, Aksu City, China.,National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
163
|
Lowe M, Nguyen L, Patel DJ. A Review of the Recent Advances of Cardiac Pacemaker Technology in Handling Complications. J Long Term Eff Med Implants 2023; 33:21-29. [PMID: 37522582 DOI: 10.1615/jlongtermeffmedimplants.2022039586] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The total number of annual pacemaker implantations continues to grow globally, and help patients with heart rhythm disorders with an improved quality of life and reduced mortality. The first implantable pacemakers appeared in 1965, characterized by their bulkiness, short battery life, and a single pacing mode. Innovation led to the modern pacemaker: a smaller system with improved battery life and capacity, and innovation in lead technology. Certain arrhythmia conditions may also qualify for leadless pacemaker implantation, thus eliminating the spectrum of complications that could occur with leads. Adverse events can be divided into acute (perforation, lead dislodgement, infection) and long-term (lead fractures, device infection, insulation failure). Traditional long-term complications with leads occur in 10% of patients, compared with device-related adverse effects observed in 6.7% of leadless pacemakers. Furthermore, cardiac pacemaker implantation results in quality of life improvements across all age groups. Large cardiac rehabilitation studies have demonstrated the effectiveness of exercise in reducing the physical complications involved with pacemaker implantation. Of the three randomized controlled trials examined, all of them reported some benefit of exercise in the intervention group compared with the control. The following review aims to discuss the multitude of pacemaker options potentially available for the clinician, complications, their course of management, and the path forward with innovations arising out of previous research within the field.
Collapse
Affiliation(s)
- Megan Lowe
- Department of Public Health, University of Toronto, Toronto, Ontario M5S 1A1 Canada
| | - Lily Nguyen
- Department of Public Health, University of Toronto, Toronto, Ontario M5S 1A1 Canada
| | - Dhiman J Patel
- Departments of Kinesiology and Medicine, McMaster University, 1280 Main Street West, Hamilton, L8S 4L9, Ontario, Canada
| |
Collapse
|
164
|
Pastore MC, Ilardi F, Stefanini A, Mandoli GE, Palermi S, Bandera F, Benfari G, Esposito R, Lisi M, Pasquini A, Santoro C, Valente S, D’Andrea A, Cameli M. Bedside Ultrasound for Hemodynamic Monitoring in Cardiac Intensive Care Unit. J Clin Med 2022; 11:jcm11247538. [PMID: 36556154 PMCID: PMC9785677 DOI: 10.3390/jcm11247538] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/03/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Thanks to the advances in medical therapy and assist devices, the management of patients hospitalized in cardiac intensive care unit (CICU) is becoming increasingly challenging. In fact, Patients in the cardiac intensive care unit are frequently characterized by dynamic and variable diseases, which may evolve into several clinical phenotypes based on underlying etiology and its complexity. Therefore, the use of noninvasive tools in order to provide a personalized approach to these patients, according to their phenotype, may help to optimize the therapeutic strategies towards the underlying etiology. Echocardiography is the most reliable and feasible bedside method to assess cardiac function repeatedly, assisting clinicians not only in characterizing hemodynamic disorders, but also in helping to guide interventions and monitor response to therapies. Beyond basic echocardiographic parameters, its application has been expanded with the introduction of new tools such as lung ultrasound (LUS), the Venous Excess UltraSound (VexUS) grading system, and the assessment of pulmonary hypertension, which is fundamental to guide oxygen therapy. The aim of this review is to provide an overview on the current knowledge about the pathophysiology and echocardiographic evaluation of perfusion and congestion in patients in CICU, and to provide practical indications for the use of echocardiography across clinical phenotypes and new applications in CICU.
Collapse
Affiliation(s)
- Maria Concetta Pastore
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy
- Correspondence: (M.C.P.); (M.C.); Tel.: +39-057-758-5377 (M.C.P.)
| | - Federica Ilardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80138 Naples, Italy
- Mediterranea Cardiocentro, 80122 Naples, Italy
| | - Andrea Stefanini
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy
| | - Giulia Elena Mandoli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy
| | - Stefano Palermi
- Public Health Department, University of Naples Federico II, 80131 Naples, Italy
| | - Francesco Bandera
- Cardiology University Department, Heart Failure Unit, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
- Department of Biomedical Sciences for Health, University of Milano, 20122 Milan, Italy
| | - Giovanni Benfari
- Section of Cardiology, Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Roberta Esposito
- Department of Clinical Medicine and Surgery, Federico II University Hospital, 80131 Naples, Italy
| | - Matteo Lisi
- Department of Cardiovascular Disease—AUSL Romagna, Division of Cardiology, Ospedale S. Maria delle Croci, Viale Randi 5, 48121 Ravenna, Italy
| | - Annalisa Pasquini
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 20123 Rome, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80138 Naples, Italy
| | - Serafina Valente
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy
| | - Antonello D’Andrea
- Department of Cardiology, Umberto I Hospital, 84014 Nocera Inferiore, Italy
| | - Matteo Cameli
- Department of Medical Biotechnologies, Division of Cardiology, University of Siena, 53100 Siena, Italy
- Correspondence: (M.C.P.); (M.C.); Tel.: +39-057-758-5377 (M.C.P.)
| |
Collapse
|
165
|
Varudo R, Gonzalez FA, Leote J, Martins C, Bacariza J, Fernandes A, Michard F. Machine learning for the real-time assessment of left ventricular ejection fraction in critically ill patients: a bedside evaluation by novices and experts in echocardiography. Crit Care 2022; 26:386. [PMID: 36517906 PMCID: PMC9749290 DOI: 10.1186/s13054-022-04269-6] [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: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Machine learning algorithms have recently been developed to enable the automatic and real-time echocardiographic assessment of left ventricular ejection fraction (LVEF) and have not been evaluated in critically ill patients. METHODS Real-time LVEF was prospectively measured in 95 ICU patients with a machine learning algorithm installed on a cart-based ultrasound system. Real-time measurements taken by novices (LVEFNov) and by experts (LVEFExp) were compared with LVEF reference measurements (LVEFRef) taken manually by echo experts. RESULTS LVEFRef ranged from 26 to 80% (mean 54 ± 12%), and the reproducibility of measurements was 9 ± 6%. Thirty patients (32%) had a LVEFRef < 50% (left ventricular systolic dysfunction). Real-time LVEFExp and LVEFNov measurements ranged from 31 to 68% (mean 54 ± 10%) and from 28 to 70% (mean 54 ± 9%), respectively. The reproducibility of measurements was comparable for LVEFExp (5 ± 4%) and for LVEFNov (6 ± 5%) and significantly better than for reference measurements (p < 0.001). We observed a strong relationship between LVEFRef and both real-time LVEFExp (r = 0.86, p < 0.001) and LVEFNov (r = 0.81, p < 0.001). The average difference (bias) between real time and reference measurements was 0 ± 6% for LVEFExp and 0 ± 7% for LVEFNov. The sensitivity to detect systolic dysfunction was 70% for real-time LVEFExp and 73% for LVEFNov. The specificity to detect systolic dysfunction was 98% both for LVEFExp and LVEFNov. CONCLUSION Machine learning-enabled real-time measurements of LVEF were strongly correlated with manual measurements obtained by experts. The accuracy of real-time LVEF measurements was excellent, and the precision was fair. The reproducibility of LVEF measurements was better with the machine learning system. The specificity to detect left ventricular dysfunction was excellent both for experts and for novices, whereas the sensitivity could be improved. TRIAL REGISTRATION NCT05336448. Retrospectively registered on April 19, 2022.
Collapse
Affiliation(s)
- Rita Varudo
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal
| | - Filipe A. Gonzalez
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal ,grid.9983.b0000 0001 2181 4263Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - João Leote
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal
| | - Cristina Martins
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal
| | - Jacobo Bacariza
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal
| | - Antero Fernandes
- grid.414708.e0000 0000 8563 4416Intensive Care Department, Hospital Garcia de Orta, Almada, Portugal ,grid.9983.b0000 0001 2181 4263Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal ,grid.7427.60000 0001 2220 7094Faculdade de Ciencias da Saude da Universidade da Beira Interior, Covilha, Portugal
| | | |
Collapse
|
166
|
Bazmpani MA, Nikolaidou C, Papanastasiou CA, Ziakas A, Karamitsos TD. Cardiovascular Magnetic Resonance Parametric Mapping Techniques for the Assessment of Chronic Coronary Syndromes. J Cardiovasc Dev Dis 2022; 9:jcdd9120443. [PMID: 36547440 PMCID: PMC9782163 DOI: 10.3390/jcdd9120443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The term chronic coronary syndromes encompasses a variety of clinical presentations of coronary artery disease (CAD), ranging from stable angina due to epicardial coronary artery disease to microvascular coronary dysfunction. Cardiac magnetic resonance (CMR) imaging has an established role in the diagnosis, prognostication and treatment planning of patients with CAD. Recent advances in parametric mapping CMR techniques have added value in the assessment of patients with chronic coronary syndromes, even without the need for gadolinium contrast administration. Furthermore, quantitative perfusion CMR techniques have enabled the non-invasive assessment of myocardial blood flow and myocardial perfusion reserve and can reliably identify multivessel coronary artery disease and microvascular dysfunction. This review summarizes the clinical applications and the prognostic value of the novel CMR parametric mapping techniques in the setting of chronic coronary syndromes and discusses their strengths, pitfalls and future directions.
Collapse
Affiliation(s)
- Maria Anna Bazmpani
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | | | - Christos A. Papanastasiou
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Antonios Ziakas
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros D. Karamitsos
- Department of First Cardiology, Aristotle University of Thessaloniki School of Medicine, AHEPA University Hospital, 54636 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-2310994832; Fax: +30-2310994673
| |
Collapse
|
167
|
Huang JY, Lin YH, Hung CL, Chen WP, Tamaki N, Bax JJ, Morris DA, Korosoglou G, Wu YW. Editorial: Atherosclerosis and functional imaging. Front Cardiovasc Med 2022; 9:1053100. [PMID: 36561766 PMCID: PMC9767462 DOI: 10.3389/fcvm.2022.1053100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jei-Yie Huang
- Department of Nuclear Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Hung Lin
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chung-Lieh Hung
- Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan,Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Wen-Pin Chen
- Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - Nagara Tamaki
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jeroen J. Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniel A. Morris
- Department of Internal Medicine and Cardiology, Charité University Hospital, Berlin, Germany
| | - Grigorios Korosoglou
- Department of Cardiology and Vascular Medicine, GRN Hospital Weinheim, Weinheim, Germany
| | - Yen-Wen Wu
- Department of Nuclear Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan,Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan,Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan,Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan,*Correspondence: Yen-Wen Wu
| |
Collapse
|
168
|
Jone PN, Gearhart A, Lei H, Xing F, Nahar J, Lopez-Jimenez F, Diller GP, Marelli A, Wilson L, Saidi A, Cho D, Chang AC. Artificial Intelligence in Congenital Heart Disease: Current State and Prospects. JACC. ADVANCES 2022; 1:100153. [PMID: 38939457 PMCID: PMC11198540 DOI: 10.1016/j.jacadv.2022.100153] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 06/29/2024]
Abstract
The current era of big data offers a wealth of new opportunities for clinicians to leverage artificial intelligence to optimize care for pediatric and adult patients with a congenital heart disease. At present, there is a significant underutilization of artificial intelligence in the clinical setting for the diagnosis, prognosis, and management of congenital heart disease patients. This document is a call to action and will describe the current state of artificial intelligence in congenital heart disease, review challenges, discuss opportunities, and focus on the top priorities of artificial intelligence-based deployment in congenital heart disease.
Collapse
Affiliation(s)
- Pei-Ni Jone
- Section of Pediatric Cardiology, Department of Pediatrics, Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Addison Gearhart
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Howard Lei
- Division of Pediatric Cardiology, Children’s Hospital of Orange County, Orange, California, USA
| | - Fuyong Xing
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jai Nahar
- Department of Cardiology, Children's National Hospital, Washington, DC, USA
| | | | - Gerhard-Paul Diller
- Department of Cardiology III-Adult Congenital and Valvular Heart Disease, University Hospital Muenster, Muenster, Germany
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton and Harefield National Health Service Foundation Trust, Imperial College London, London, UK
- National Register for Congenital Heart Defects, Berlin, Germany
| | - Ariane Marelli
- McGill Adult Unit for Congenital Heart Disease Excellence, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Laura Wilson
- Department of Pediatrics, University of Florida-Congenital Heart Center, Gainesville, Florida, USA
| | - Arwa Saidi
- Department of Pediatrics, University of Florida-Congenital Heart Center, Gainesville, Florida, USA
| | - David Cho
- Department of Cardiology, University of California at Los Angeles, Los Angeles, California, USA
| | - Anthony C. Chang
- Division of Pediatric Cardiology, Children’s Hospital of Orange County, Orange, California, USA
| |
Collapse
|
169
|
Liu H, Wu J, Shi L, Liu Y, Miller E, Sinusas A, Liu YH, Liu C. Post-reconstruction attenuation correction for SPECT myocardium perfusion imaging facilitated by deep learning-based attenuation map generation. J Nucl Cardiol 2022; 29:2881-2892. [PMID: 34671940 DOI: 10.1007/s12350-021-02817-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/09/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Attenuation correction can improve the quantitative accuracy of single-photon emission computed tomography (SPECT) images. Existing SPECT-only systems normally can only provide non-attenuation corrected (NC) images which are susceptible to attenuation artifacts. In this work, we developed a post-reconstruction attenuation correction (PRAC) approach facilitated by a deep learning-based attenuation map for myocardial perfusion SPECT imaging. METHODS In the PRAC method, new projection data were estimated via forwardly projecting the scanner-generated NC image. Then an attenuation map, generated from NC image using a pretrained deep learning (DL) convolutional neural network, was incorporated into an offline reconstruction algorithm to obtain the attenuation-corrected images from the forwardly projected projections. We evaluated the PRAC method using 30 subjects with a DL network trained with 40 subjects, using the vendor-generated AC images and CT-based attenuation maps as the ground truth. RESULTS The PRAC methods using DL-generated and CT-based attenuation maps were both highly consistent with the scanner-generated AC image. The globally normalized mean absolute errors were 1.1% ± .6% and .7% ± .4% and the localized absolute percentage errors were 8.9% ± 13.4% and 7.8% ± 11.4% in the left ventricular (LV) blood pool, respectively, and - 1.3% ± 8.0% and - 3.8% ± 4.5% in the LV myocardium for PRAC methods using DL-generated and CT-based attenuation maps, respectively. The summed stress scores after PRAC using both attenuation maps were more consistent with the ground truth than those of the NC images. CONCLUSION We developed a PRAC approach facilitated by deep learning-based attenuation maps for SPECT myocardial perfusion imaging. It may be feasible for this approach to provide AC images for SPECT-only scanner data.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA.
- Department of Engineering Physics, Tsinghua University, Beijing, 100084, China.
- Key Laboratory of Particle & Radiation Imaging (Tsinghua University), Ministry of Education, Beijing, China.
| | - Jing Wu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
- Center for Advanced Quantum Studies and Department of Physics, Beijing Normal University, Beijing, China
| | - Luyao Shi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yaqiang Liu
- Department of Engineering Physics, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Particle & Radiation Imaging (Tsinghua University), Ministry of Education, Beijing, China
| | - Edward Miller
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
- Department of Internal Medicine (Cardiology), Yale University, New Haven, CT, USA
| | - Albert Sinusas
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Internal Medicine (Cardiology), Yale University, New Haven, CT, USA
| | - Yi-Hwa Liu
- Department of Internal Medicine (Cardiology), Yale University, New Haven, CT, USA
- Department of Biomedical Imaging and Radiological Sciences, National Yangming Jiaotong University, Taipei, Taiwan
| | - Chi Liu
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, 06520, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
170
|
Artificial intelligence for prediction of response to cancer immunotherapy. Semin Cancer Biol 2022; 87:137-147. [PMID: 36372326 DOI: 10.1016/j.semcancer.2022.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Artificial intelligence (AI) indicates the application of machines to imitate intelligent behaviors for solving complex tasks with minimal human intervention, including machine learning and deep learning. The use of AI in medicine improves health-care systems in multiple areas such as diagnostic confirmation, risk stratification, analysis, prognosis prediction, treatment surveillance, and virtual health support, which has considerable potential to revolutionize and reshape medicine. In terms of immunotherapy, AI has been applied to unlock underlying immune signatures to associate with responses to immunotherapy indirectly as well as predict responses to immunotherapy responses directly. The AI-based analysis of high-throughput sequences and medical images can provide useful information for management of cancer immunotherapy considering the excellent abilities in selecting appropriate subjects, improving therapeutic regimens, and predicting individualized prognosis. In present review, we aim to evaluate a broad framework about AI-based computational approaches for prediction of response to cancer immunotherapy on both indirect and direct manners. Furthermore, we summarize our perspectives about challenges and opportunities of further AI applications on cancer immunotherapy relating to clinical practicability.
Collapse
|
171
|
Ikerionwu C, Ugwuishiwu C, Okpala I, James I, Okoronkwo M, Nnadi C, Orji U, Ebem D, Ike A. Application of machine and deep learning algorithms in optical microscopic detection of Plasmodium: A malaria diagnostic tool for the future. Photodiagnosis Photodyn Ther 2022; 40:103198. [PMID: 36379305 DOI: 10.1016/j.pdpdt.2022.103198] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Machine and deep learning techniques are prevalent in the medical discipline due to their high level of accuracy in disease diagnosis. One such disease is malaria caused by Plasmodium falciparum and transmitted by the female anopheles mosquito. According to the World Health Organisation (WHO), millions of people are infected annually, leading to inevitable deaths in the infected population. Statistical records show that early detection of malaria parasites could prevent deaths and machine learning (ML) has proved helpful in the early detection of malarial parasites. Human error is identified to be a major cause of inaccurate diagnostics in the traditional microscopy malaria diagnosis method. Therefore, the method would be more reliable if human expert dependency is restricted or entirely removed, and thus, the motivation of this paper. This study presents a systematic review to understand the prevalent machine learning algorithms applied to a low-cost, portable optical microscope in the automation of blood film interpretation for malaria parasite detection. Peer-reviewed papers were downloaded from selected reputable databases eg. Elsevier, IEEExplore, Pubmed, Scopus, Web of Science, etc. The extant literature suggests that convolutional neural network (CNN) and its variants (deep learning) account for 41.9% of the microscopy malaria diagnosis using machine learning with a prediction accuracy of 99.23%. Thus, the findings suggest that early detection of the malaria parasite has improved through the application of CNN and other ML algorithms on microscopic malaria parasite detection.
Collapse
Affiliation(s)
- Charles Ikerionwu
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Software Engineering, Federal University of Technology, Owerri, Imo State, Nigeria
| | - Chikodili Ugwuishiwu
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Computer Science, University of Nigeria, Nsukka, Enugu State, Nigeria.
| | - Izunna Okpala
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Information Technology, University of Cincinnati, USA
| | - Idara James
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Computer Science, Akwa Ibom State University, Nigeria
| | - Matthew Okoronkwo
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Computer Science, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Charles Nnadi
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Deprtment of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Ugochukwu Orji
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Computer Science, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Deborah Ebem
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Computer Science, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Anthony Ike
- Machine Learning on Disease Diagnosis Research Group, Nigeria; Department of Microbiology, University of Nigeria, Nsukka, Enugu State, Nigeria
| |
Collapse
|
172
|
Singh A, Miller RJH. Deep learning-based attenuation map generation and correction; could it be useful clinically? J Nucl Cardiol 2022; 29:2893-2895. [PMID: 34877640 DOI: 10.1007/s12350-021-02875-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 01/22/2023]
Affiliation(s)
- Ananya Singh
- Departments of Imaging, Medicine and Biomedical Sciences, Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Robert J H Miller
- Departments of Imaging, Medicine and Biomedical Sciences, Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Cardiac Sciences, University of Calgary, GAA08, 3230 Hospital Drive NW, Calgary, AB, T2N 2T9, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| |
Collapse
|
173
|
Bhavnani SP, Khedraki R, Cohoon TJ, Meine FJ, Stuckey TD, McMinn T, Depta JP, Bennett B, McGarry T, Carroll W, Suh D, Steuter JA, Roberts M, Gillins HR, Shadforth I, Lange E, Doomra A, Firouzi M, Fathieh F, Burton T, Khosousi A, Ramchandani S, Sanders WE, Smart F. Multicenter validation of a machine learning phase space electro-mechanical pulse wave analysis to predict elevated left ventricular end diastolic pressure at the point-of-care. PLoS One 2022; 17:e0277300. [PMID: 36378672 PMCID: PMC9665374 DOI: 10.1371/journal.pone.0277300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Phase space is a mechanical systems approach and large-scale data representation of an object in 3-dimensional space. Whether such techniques can be applied to predict left ventricular pressures non-invasively and at the point-of-care is unknown. OBJECTIVE This study prospectively validated a phase space machine-learned approach based on a novel electro-mechanical pulse wave method of data collection through orthogonal voltage gradient (OVG) and photoplethysmography (PPG) for the prediction of elevated left ventricular end diastolic pressure (LVEDP). METHODS Consecutive outpatients across 15 US-based healthcare centers with symptoms suggestive of coronary artery disease were enrolled at the time of elective cardiac catheterization and underwent OVG and PPG data acquisition immediately prior to angiography with signals paired with LVEDP (IDENTIFY; NCT #03864081). The primary objective was to validate a ML algorithm for prediction of elevated LVEDP using a definition of ≥25 mmHg (study cohort) and normal LVEDP ≤ 12 mmHg (control cohort), using AUC as the measure of diagnostic accuracy. Secondary objectives included performance of the ML predictor in a propensity matched cohort (age and gender) and performance for an elevated LVEDP across a spectrum of comparative LVEDP (<12 through 24 at 1 mmHg increments). Features were extracted from the OVG and PPG datasets and were analyzed using machine-learning approaches. RESULTS The study cohort consisted of 684 subjects stratified into three LVEDP categories, ≤12 mmHg (N = 258), LVEDP 13-24 mmHg (N = 347), and LVEDP ≥25 mmHg (N = 79). Testing of the ML predictor demonstrated an AUC of 0.81 (95% CI 0.76-0.86) for the prediction of an elevated LVEDP with a sensitivity of 82% and specificity of 68%, respectively. Among a propensity matched cohort (N = 79) the ML predictor demonstrated a similar result AUC 0.79 (95% CI: 0.72-0.8). Using a constant definition of elevated LVEDP and varying the lower threshold across LVEDP the ML predictor demonstrated and AUC ranging from 0.79-0.82. CONCLUSION The phase space ML analysis provides a robust prediction for an elevated LVEDP at the point-of-care. These data suggest a potential role for an OVG and PPG derived electro-mechanical pulse wave strategy to determine if LVEDP is elevated in patients with symptoms suggestive of cardiac disease.
Collapse
Affiliation(s)
- Sanjeev P. Bhavnani
- Division of Cardiovascular Medicine, Healthcare Innovation & Practice Transformation Laboratory, Scripps Clinic, San Diego, California, United States of America
- * E-mail:
| | - Rola Khedraki
- Division of Cardiology, Section Advanced Heart Failure, Scripps Clinic, San Diego, California, United States of America
| | - Travis J. Cohoon
- Division of Cardiovascular Medicine, Healthcare Innovation & Practice Transformation Laboratory, Scripps Clinic, San Diego, California, United States of America
| | - Frederick J. Meine
- Novant Health New Hanover Regional Medical Center, Wilmington, North Carolina, United States of America
| | - Thomas D. Stuckey
- Cone Health Heart and Vascular Center, Greensboro, North Carolina, United States of America
| | - Thomas McMinn
- Austin Heart, Austin, Texas, United States of America
| | - Jeremiah P. Depta
- Rochester General Hospital, Rochester, New York, United States of America
| | - Brett Bennett
- Jackson Heart Clinic, Jackson, Mississippi, United States of America
| | - Thomas McGarry
- Oklahoma Heart Hospital, Oklahoma City, Oklahoma, United States of America
| | - William Carroll
- Cardiology Associates of North Mississippi, Tupelo, Mississippi, United States of America
| | - David Suh
- Atlanta Heart Specialists, Atlanta, Georgia, United States of America
| | | | - Michael Roberts
- Lexington Medical Center, West Columbia, South Carolina, United States of America
| | | | - Ian Shadforth
- CorVista Health, Inc., Washington, DC, United States of America
| | - Emmanuel Lange
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Abhinav Doomra
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Mohammad Firouzi
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Farhad Fathieh
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Timothy Burton
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Ali Khosousi
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | - Shyam Ramchandani
- CorVista Health, Toronto, Ontario, Canada
- Analytics For Life Inc., d.b.a CorVista Health, Toronto, Canada
| | | | - Frank Smart
- LSU Health Science Center, New Orleans, Louisiana, United States of America
| |
Collapse
|
174
|
Szabo L, Raisi-Estabragh Z, Salih A, McCracken C, Ruiz Pujadas E, Gkontra P, Kiss M, Maurovich-Horvath P, Vago H, Merkely B, Lee AM, Lekadir K, Petersen SE. Clinician's guide to trustworthy and responsible artificial intelligence in cardiovascular imaging. Front Cardiovasc Med 2022; 9:1016032. [PMID: 36426221 PMCID: PMC9681217 DOI: 10.3389/fcvm.2022.1016032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/11/2022] [Indexed: 12/01/2023] Open
Abstract
A growing number of artificial intelligence (AI)-based systems are being proposed and developed in cardiology, driven by the increasing need to deal with the vast amount of clinical and imaging data with the ultimate aim of advancing patient care, diagnosis and prognostication. However, there is a critical gap between the development and clinical deployment of AI tools. A key consideration for implementing AI tools into real-life clinical practice is their "trustworthiness" by end-users. Namely, we must ensure that AI systems can be trusted and adopted by all parties involved, including clinicians and patients. Here we provide a summary of the concepts involved in developing a "trustworthy AI system." We describe the main risks of AI applications and potential mitigation techniques for the wider application of these promising techniques in the context of cardiovascular imaging. Finally, we show why trustworthy AI concepts are important governing forces of AI development.
Collapse
Affiliation(s)
- Liliana Szabo
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
- Semmelweis University Heart and Vascular Center, Budapest, Hungary
| | - Zahra Raisi-Estabragh
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Ahmed Salih
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Celeste McCracken
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, University of Oxford, Oxford, United Kingdom
| | - Esmeralda Ruiz Pujadas
- Departament de Matemàtiques i Informàtica, Artificial Intelligence in Medicine Lab (BCN-AIM), Universitat de Barcelona, Barcelona, Spain
| | - Polyxeni Gkontra
- Departament de Matemàtiques i Informàtica, Artificial Intelligence in Medicine Lab (BCN-AIM), Universitat de Barcelona, Barcelona, Spain
| | - Mate Kiss
- Siemens Healthcare Hungary, Budapest, Hungary
| | - Pal Maurovich-Horvath
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Hajnalka Vago
- Semmelweis University Heart and Vascular Center, Budapest, Hungary
| | - Bela Merkely
- Semmelweis University Heart and Vascular Center, Budapest, Hungary
| | - Aaron M. Lee
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Karim Lekadir
- Departament de Matemàtiques i Informàtica, Artificial Intelligence in Medicine Lab (BCN-AIM), Universitat de Barcelona, Barcelona, Spain
| | - Steffen E. Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
- Health Data Research UK, London, United Kingdom
- Alan Turing Institute, London, United Kingdom
| |
Collapse
|
175
|
Tang CX, Zhou Z, Zhang JY, Xu L, Lv B, Jiang Zhang L. Cardiovascular Imaging in China: Yesterday, Today, and Tomorrow. J Thorac Imaging 2022; 37:355-365. [PMID: 36162066 DOI: 10.1097/rti.0000000000000678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The high prevalence and mortality of cardiovascular diseases in China's large population has increased the use of cardiovascular imaging for the assessment of conditions in recent years. In this study, we review the past 20 years of cardiovascular imaging in China, the increasingly important role played by cardiovascular computed tomography in coronary artery disease and pulmonary embolism assessment, magnetic resonance imaging's use for cardiomyopathy assessment, the development and application of artificial intelligence in cardiovascular imaging, and the future of Chinese cardiovascular imaging.
Collapse
Affiliation(s)
- Chun Xiang Tang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province
| | - Zhen Zhou
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University
| | - Jia Yin Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Xu
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University
| | - Bin Lv
- Department of Radiology, Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences
- State Key Laboratory and National Center for Cardiovascular Diseases, Beijing
| | - Long Jiang Zhang
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province
| |
Collapse
|
176
|
Berz AM, Dromain C, Vietti-Violi N, Boughdad S, Duran R. Tumor response assessment on imaging following immunotherapy. Front Oncol 2022; 12:982983. [PMID: 36387133 PMCID: PMC9641095 DOI: 10.3389/fonc.2022.982983] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, various systemic immunotherapies have been developed for cancer treatment, such as monoclonal antibodies (mABs) directed against immune checkpoints (immune checkpoint inhibitors, ICIs), oncolytic viruses, cytokines, cancer vaccines, and adoptive cell transfer. While being estimated to be eligible in 38.5% of patients with metastatic solid or hematological tumors, ICIs, in particular, demonstrate durable disease control across many oncologic diseases (e.g., in melanoma, lung, bladder, renal, head, and neck cancers) and overall survival benefits. Due to their unique mechanisms of action based on T-cell activation, response to immunotherapies is characterized by different patterns, such as progression prior to treatment response (pseudoprogression), hyperprogression, and dissociated responses following treatment. Because these features are not encountered in the Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), which is the standard for response assessment in oncology, new criteria were defined for immunotherapies. The most important changes in these new morphologic criteria are, firstly, the requirement for confirmatory imaging examinations in case of progression, and secondly, the appearance of new lesions is not necessarily considered a progressive disease. Until today, five morphologic (immune-related response criteria (irRC), immune-related RECIST (irRECIST), immune RECIST (iRECIST), immune-modified RECIST (imRECIST), and intra-tumoral RECIST (itRECIST)) criteria have been developed to accurately assess changes in target lesion sizes, taking into account the specific response patterns after immunotherapy. In addition to morphologic response criteria, 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG-PET/CT) is a promising option for metabolic response assessment and four metabolic criteria are used (PET/CT Criteria for Early Prediction of Response to Immune Checkpoint Inhibitor Therapy (PECRIT), PET Response Evaluation Criteria for Immunotherapy (PERCIMT), immunotherapy-modified PET Response Criteria in Solid Tumors (imPERCIST5), and immune PERCIST (iPERCIST)). Besides, there is evidence that parameters on 18F-FDG-PET/CT, such as the standardized uptake value (SUV)max and several radiotracers, e.g., directed against PD-L1, may be potential imaging biomarkers of response. Moreover, the emerge of human intratumoral immunotherapy (HIT-IT), characterized by the direct injection of immunostimulatory agents into a tumor lesion, has given new importance to imaging assessment. This article reviews the specific imaging patterns of tumor response and progression and available imaging response criteria following immunotherapy.
Collapse
Affiliation(s)
- Antonia M. Berz
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
- Department of Radiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Naïk Vietti-Violi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Sarah Boughdad
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Rafael Duran
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
177
|
The Merits, Limitations, and Future Directions of Cost-Effectiveness Analysis in Cardiac MRI with a Focus on Coronary Artery Disease: A Literature Review. J Cardiovasc Dev Dis 2022; 9:jcdd9100357. [PMID: 36286309 PMCID: PMC9604922 DOI: 10.3390/jcdd9100357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiac magnetic resonance (CMR) imaging has a wide range of clinical applications with a high degree of accuracy for many myocardial pathologies. Recent literature has shown great utility of CMR in diagnosing many diseases, often changing the course of treatment. Despite this, it is often underutilized possibly due to perceived costs, limiting patient factors and comfort, and longer examination periods compared to other imaging modalities. In this regard, we conducted a literature review using keywords “Cost-Effectiveness” and “Cardiac MRI” and selected articles from the PubMed MEDLINE database that met our inclusion and exclusion criteria to examine the cost-effectiveness of CMR. Our search result yielded 17 articles included in our review. We found that CMR can be cost-effective in quality-adjusted life years (QALYs) in select patient populations with various cardiac pathologies. Specifically, the use of CMR in coronary artery disease (CAD) patients with a pretest probability below a certain threshold may be more cost-effective compared to patients with a higher pretest probability, although its use can be limited based on geographic location, professional society guidelines, and differing reimbursement patterns. In addition, a stepwise combination of different imaging modalities, with conjunction of AHA/ACC guidelines can further enhance the cost-effectiveness of CMR.
Collapse
|
178
|
Kunovac A, Hathaway QA, Burrage EN, Coblentz T, Kelley EE, Sengupta PP, Hollander JM, Chantler PD. Left Ventricular Segmental Strain Identifies Unique Myocardial Deformation Patterns After Intrinsic and Extrinsic Stressors in Mice. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2128-2138. [PMID: 35933241 PMCID: PMC9427680 DOI: 10.1016/j.ultrasmedbio.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
We used segmental strain analysis to evaluate whether intrinsic (diet-induced obesity [DIO]) and extrinsic (unpredictable chronic mild stress [UCMS]) stressors can alter deformational patterns of the left ventricle. Six-week-old male C57BL/6J mice were randomized into the lean or obese group (n = 24/group). Mice underwent 12 wk of DIO with a high-fat diet (HFD). At 18 wk, lean and obese mice were further randomized into UCMS and non-UCMS groups (UCMS, 7 h/d, 5 d/wk, for 8 wk). Echocardiography was performed at baseline (6 wk), post-HFD (18 wk) and post-UCMS (26 wk). Machine learning was applied to the DIO and UCMS groups. There was robust predictive accuracy (area under the receiver operating characteristic curve [AUC] = 0.921) when comparing obese with lean mice, with radial strain changes in the lateral (-64%, p ≤ 0.001) and anterior free (-53%, p < 0.001) walls being most informative. The ability to predict mice that underwent UCMS, irrespective of diet, was assessed (AUC = 0.886), revealing longitudinal strain rate of the anterior midwall and radial strain of the posterior septal wall as the top features. The wall segments indicate a predilection for changes in deformation patterns to the free wall (DIO) and septal wall (UCMS), indicating disease-specific alterations to the myocardium.
Collapse
Affiliation(s)
- Amina Kunovac
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA; Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University, Morgantown, West Virginia, USA
| | - Quincy A Hathaway
- Heart and Vascular Institute, West Virginia University, Morgantown, West Virginia, USA.
| | - Emily N Burrage
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Tyler Coblentz
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Partho P Sengupta
- Heart and Vascular Institute, West Virginia University, Morgantown, West Virginia, USA; Rutgers Robert Wood Johnson University Hospital, New Brunswick, New Jersey, USA
| | - John M Hollander
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA; Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University, Morgantown, West Virginia, USA
| | - Paul D Chantler
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA; Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University, Morgantown, West Virginia, USA; Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
179
|
Lin A, van Diemen PA, Motwani M, McElhinney P, Otaki Y, Han D, Kwan A, Tzolos E, Klein E, Kuronuma K, Grodecki K, Shou B, Rios R, Manral N, Cadet S, Danad I, Driessen RS, Berman DS, Nørgaard BL, Slomka PJ, Knaapen P, Dey D. Machine Learning From Quantitative Coronary Computed Tomography Angiography Predicts Fractional Flow Reserve-Defined Ischemia and Impaired Myocardial Blood Flow. Circ Cardiovasc Imaging 2022; 15:e014369. [PMID: 36252116 PMCID: PMC10085569 DOI: 10.1161/circimaging.122.014369] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/13/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND A pathophysiological interplay exists between plaque morphology and coronary physiology. Machine learning (ML) is increasingly being applied to coronary computed tomography angiography (CCTA) for cardiovascular risk stratification. We sought to assess the performance of a ML score integrating CCTA-based quantitative plaque features for predicting vessel-specific ischemia by invasive fractional flow reserve (FFR) and impaired myocardial blood flow (MBF) by positron emission tomography (PET). METHODS This post-hoc analysis of the PACIFIC trial (Prospective Comparison of Cardiac Positron Emission Tomography/Computed Tomography [CT]' Single Photon Emission Computed Tomography/CT Perfusion Imaging and CT Coronary Angiography with Invasive Coronary Angiography) included 208 patients with suspected coronary artery disease who prospectively underwent CCTA' [15O]H2O PET, and invasive FFR. Plaque quantification from CCTA was performed using semiautomated software. An ML algorithm trained on the prospective NXT trial (484 vessels) was used to develop a ML score for the prediction of ischemia (FFR≤0.80), which was then evaluated in 581 vessels from the PACIFIC trial. Thereafter, the ML score was applied for predicting impaired hyperemic MBF (≤2.30 mL/min per g) from corresponding PET scans. The performance of the ML score was compared with CCTA reads and noninvasive FFR derived from CCTA (FFRCT). RESULTS One hundred thirty-nine (23.9%) vessels had FFR-defined ischemia, and 195 (33.6%) vessels had impaired hyperemic MBF. For the prediction of FFR-defined ischemia, the ML score yielded an area under the receiver-operating characteristic curve of 0.92, which was significantly higher than that of visual stenosis grade (0.84; P<0.001) and comparable with that of FFRCT (0.93; P=0.34). Quantitative percent diameter stenosis and low-density noncalcified plaque volume had the greatest ML feature importance for predicting FFR-defined ischemia. When applied for impaired MBF prediction, the ML score exhibited an area under the receiver-operating characteristic curve of 0.80; significantly higher than visual stenosis grade (area under the receiver-operating characteristic curve 0.74; P=0.02) and comparable with FFRCT (area under the receiver-operating characteristic curve 0.77; P=0.16). CONCLUSIONS An externally validated ML score integrating CCTA-based quantitative plaque features accurately predicts FFR-defined ischemia and impaired MBF by PET, performing superiorly to standard CCTA stenosis evaluation and comparably to FFRCT.
Collapse
Affiliation(s)
- Andrew Lin
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pepijn A. van Diemen
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Manish Motwani
- Manchester Heart Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Priscilla McElhinney
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yuka Otaki
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Donghee Han
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alan Kwan
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Evangelos Tzolos
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
| | - Eyal Klein
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Keiichiro Kuronuma
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kajetan Grodecki
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Benjamin Shou
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard Rios
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nipun Manral
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sebastien Cadet
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ibrahim Danad
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Roel S. Driessen
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Daniel S. Berman
- Department of Imaging and Medicine and the Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bjarne L. Nørgaard
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Piotr J. Slomka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul Knaapen
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
180
|
Choi W, Key J, Youn I, Lee H, Han S. Cavitation-assisted sonothrombolysis by asymmetrical nanostars for accelerated thrombolysis. J Control Release 2022; 350:870-885. [PMID: 36096365 DOI: 10.1016/j.jconrel.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022]
Abstract
Sonothrombolysis with recombinant tissue plasminogen activator (rtPA) and microbubbles has been widely studied to enhance thrombolytic potential. Here, we report different sonothrombolysis strategy in nanoparticles using microbubbles cavitation. We found that different particles in shape exhibited different reactivity toward the cavitation, leading to a distinct sonothrombolytic potential. Two different gold nanoparticles in shape were functionalized with the rtPA: rtPA-functionalized gold nanospheres (NPt) and gold nanostars (NSt). NPt could not accelerate the thrombolytic potential with a sole acoustic stimulus. Importantly, NSt enhanced the potential with acoustic stimulus and microbubble-mediated cavitation, while NPt were not reactive to cavitation. Coadministration of NSt and microbubbles resulted in a dramatic reduction of the infarcts in a photothrombotic model and recovery in the cerebral blood flow. Given the synergistic effect and in vivo feasibility of this strategy, cavitation-assisted sonothrombolysis by asymmetrical NSt might be useful for treating acute ischemic stroke.
Collapse
Affiliation(s)
- Wonseok Choi
- Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Seongbuk-gu, Republic of Korea; Department of Biomedical Engineering, Yonsei University, Wonju 26493, Gangwon-do, Republic of Korea
| | - Jaehong Key
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Gangwon-do, Republic of Korea
| | - Inchan Youn
- Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Seongbuk-gu, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Seongbuk-gu, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Seongbuk-gu, Republic of Korea
| | - Hyojin Lee
- Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Seongbuk-gu, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Seongbuk-gu, Republic of Korea.
| | - Sungmin Han
- Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Seongbuk-gu, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Seongbuk-gu, Republic of Korea.
| |
Collapse
|
181
|
Li D, Hu J, Zhang L, Li L, Yin Q, Shi J, Guo H, Zhang Y, Zhuang P. Deep learning and machine intelligence: New computational modeling techniques for discovery of the combination rules and pharmacodynamic characteristics of Traditional Chinese Medicine. Eur J Pharmacol 2022; 933:175260. [PMID: 36116517 DOI: 10.1016/j.ejphar.2022.175260] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
It has been increasingly accepted that Multi-Ingredient-Based interventions provide advantages over single-target therapy for complex diseases. With the growing development of Traditional Chinese Medicine (TCM) and continually being refined of a holistic view, "multi-target" and "multi-pathway" integration characteristics of which are being accepted. However, its effector substances, efficacy targets, especially the combination rules and mechanisms remain unclear, and more powerful strategies to interpret the synergy are urgently needed. Artificial intelligence (AI) and computer vision lead to a rapidly expanding in many fields, including diagnosis and treatment of TCM. AI technology significantly improves the reliability and accuracy of diagnostics, target screening, and new drug research. While all AI techniques are capable of matching models to biological big data, the specific methods are complex and varied. Retrieves literature by the keywords such as "artificial intelligence", "machine learning", "deep learning", "traditional Chinese medicine" and "Chinese medicine". Search the application of computer algorithms of TCM between 2000 and 2021 in PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), Elsevier and Springer. This review concentrates on the application of computational in herb quality evaluation, drug target discovery, optimized compatibility and medical diagnoses of TCM. We describe the characteristics of biological data for which different AI techniques are applicable, and discuss some of the best data mining methods and the problems faced by deep learning and machine learning methods applied to Chinese medicine.
Collapse
Affiliation(s)
- Dongna Li
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jing Hu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lili Li
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qingsheng Yin
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China
| | - Hong Guo
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanjun Zhang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, China.
| | - Pengwei Zhuang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
182
|
Nguyen MB, Villemain O, Friedberg MK, Lovstakken L, Rusin CG, Mertens L. Artificial intelligence in the pediatric echocardiography laboratory: Automation, physiology, and outcomes. FRONTIERS IN RADIOLOGY 2022; 2:881777. [PMID: 37492680 PMCID: PMC10365116 DOI: 10.3389/fradi.2022.881777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/01/2022] [Indexed: 07/27/2023]
Abstract
Artificial intelligence (AI) is frequently used in non-medical fields to assist with automation and decision-making. The potential for AI in pediatric cardiology, especially in the echocardiography laboratory, is very high. There are multiple tasks AI is designed to do that could improve the quality, interpretation, and clinical application of echocardiographic data at the level of the sonographer, echocardiographer, and clinician. In this state-of-the-art review, we highlight the pertinent literature on machine learning in echocardiography and discuss its applications in the pediatric echocardiography lab with a focus on automation of the pediatric echocardiogram and the use of echo data to better understand physiology and outcomes in pediatric cardiology. We also discuss next steps in utilizing AI in pediatric echocardiography.
Collapse
Affiliation(s)
- Minh B. Nguyen
- Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Department of Pediatric Cardiology, Baylor College of Medicine, Houston, TX, United States
| | - Olivier Villemain
- Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Mark K. Friedberg
- Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Lasse Lovstakken
- Centre for Innovative Ultrasound Solutions and Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Craig G. Rusin
- Department of Pediatric Cardiology, Baylor College of Medicine, Houston, TX, United States
| | - Luc Mertens
- Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
183
|
Zheng C, Sun BC, Wu YL, Ferencik M, Lee MS, Redberg RF, Kawatkar AA, Musigdilok VV, Sharp AL. Automated interpretation of stress echocardiography reports using natural language processing. EUROPEAN HEART JOURNAL. DIGITAL HEALTH 2022; 3:626-637. [PMID: 36710893 PMCID: PMC9779789 DOI: 10.1093/ehjdh/ztac047] [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: 05/31/2022] [Revised: 08/08/2022] [Indexed: 02/01/2023]
Abstract
Aims Stress echocardiography (SE) findings and interpretations are commonly documented in free-text reports. Reusing SE results requires laborious manual reviews. This study aimed to develop and validate an automated method for abstracting SE reports in a large cohort. Methods and results This study included adult patients who had SE within 30 days of their emergency department visit for suspected acute coronary syndrome in a large integrated healthcare system. An automated natural language processing (NLP) algorithm was developed to abstract SE reports and classify overall SE results into normal, non-diagnostic, infarction, and ischaemia categories. Randomly selected reports (n = 140) were double-blindly reviewed by cardiologists to perform criterion validity of the NLP algorithm. Construct validity was tested on the entire cohort using abstracted SE data and additional clinical variables. The NLP algorithm abstracted 6346 consecutive SE reports. Cardiologists had good agreements on the overall SE results on the 140 reports: Kappa (0.83) and intraclass correlation coefficient (0.89). The NLP algorithm achieved 98.6% specificity and negative predictive value, 95.7% sensitivity, positive predictive value, and F-score on the overall SE results and near-perfect scores on ischaemia findings. The 30-day acute myocardial infarction or death outcomes were highest among patients with ischaemia (5.0%), followed by infarction (1.4%), non-diagnostic (0.8%), and normal (0.3%) results. We found substantial variations in the format and quality of SE reports, even within the same institution. Conclusions Natural language processing is an accurate and efficient method for abstracting unstructured SE reports. This approach creates new opportunities for research, public health measures, and care improvement.
Collapse
Affiliation(s)
- Chengyi Zheng
- Corresponding author. Tel: 1-626-376-7029, Fax: 626-564-3694,
| | - Benjamin C Sun
- Department of Emergency Medicine and Leonard Davis Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi-Lin Wu
- Research and Evaluation Department, Kaiser Permanente Southern California, 100 S Los Robles Ave, 2nd Floor, Pasadena, CA 91101, USA
| | - Maros Ferencik
- Oregon Health and Science University, Knight Cardiovascular Institute, Portland, OR 97239, USA
| | - Ming-Sum Lee
- Division of Cardiology, Kaiser Permanente Southern California, Los Angeles Medical Center, Los Angeles, CA 90027, USA
| | - Rita F Redberg
- Division of Cardiology, University of California, San Francisco, CA 94143, USA
| | - Aniket A Kawatkar
- Research and Evaluation Department, Kaiser Permanente Southern California, 100 S Los Robles Ave, 2nd Floor, Pasadena, CA 91101, USA
| | - Visanee V Musigdilok
- Research and Evaluation Department, Kaiser Permanente Southern California, 100 S Los Robles Ave, 2nd Floor, Pasadena, CA 91101, USA
| | - Adam L Sharp
- Research and Evaluation Department, Kaiser Permanente Southern California, 100 S Los Robles Ave, 2nd Floor, Pasadena, CA 91101, USA,Clinical Science Department, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA 91101, USA
| |
Collapse
|
184
|
Seetharam K, Balla S, Bianco C, Cheung J, Pachulski R, Asti D, Nalluri N, Tejpal A, Mir P, Shah J, Bhat P, Mir T, Hamirani Y. Applications of Machine Learning in Cardiology. Cardiol Ther 2022; 11:355-368. [PMID: 35829916 PMCID: PMC9381660 DOI: 10.1007/s40119-022-00273-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
In this digital era, artificial intelligence (AI) is establishing a strong foothold in commercial industry and the field of technology. These effects are trickling into the healthcare industry, especially in the clinical arena of cardiology. Machine learning (ML) algorithms are making substantial progress in various subspecialties of cardiology. This will have a positive impact on patient care and move the field towards precision medicine. In this review article, we explore the progress of ML in cardiovascular imaging, electrophysiology, heart failure, and interventional cardiology.
Collapse
Affiliation(s)
- Karthik Seetharam
- Medicine Heart and Vascular Institute, West Virginia University, 1 Medical Center Drive, Morgantown, WV, 26506, USA.
- Weil Cornell Medical Center, New York, NY, USA.
| | - Sudarshan Balla
- Medicine Heart and Vascular Institute, West Virginia University, 1 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Christopher Bianco
- Medicine Heart and Vascular Institute, West Virginia University, 1 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Jim Cheung
- Weil Cornell Medical Center, New York, NY, USA
| | - Roman Pachulski
- St. John's Episcopal Hospital-South Shore, New York, NY, USA
| | - Deepak Asti
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | | | - Astha Tejpal
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | - Parvez Mir
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | - Jilan Shah
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | - Premila Bhat
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | - Tanveer Mir
- Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | | |
Collapse
|
185
|
Case JA. Deep Learning and Artificial Intelligence: What Does the Cardiologist Really Need to Know? Circ Cardiovasc Imaging 2022; 15:e014744. [PMID: 36126127 DOI: 10.1161/circimaging.122.014744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- James A Case
- Cardiovascular Imaging Technologies, University of Missouri Kansas City
| |
Collapse
|
186
|
Asif A, Charters PFP, Thompson CAS, Komber HMEI, Hudson BJ, Rodrigues JCL. Artificial intelligence can detect left ventricular dilatation on contrast-enhanced thoracic computer tomography relative to cardiac magnetic resonance imaging. Br J Radiol 2022; 95:20210852. [PMID: 35286140 PMCID: PMC9815728 DOI: 10.1259/bjr.20210852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES To assess the diagnostic accuracy of an automated algorithm to detect left ventricular (LV) dilatation on non-ECG gated CT, using cardiac magnetic resonance (CMR) as reference standard. METHODS Consecutive patients with contrast-enhanced CT thorax and CMR within 31 days (2016-2020) were analysed (n = 84). LV dilatation was defined against age-, sex- and body surface area-specific values for CMR. CTs underwent automated artificial intelligence(AI)-derived analysis that segmented ventricular chambers, presenting maximal LV diameter and volume. Area under the receiver operator curve (AUC-ROC) analysis identified CT thresholds with ≥90% sensitivity and highest specificity and ≥90% specificity with highest sensitivity. Youden's Index was used to identify thresholds with optimised sensitivity and specificity. RESULTS Automated diameter analysis was feasible in 92% of cases (77/84; 45 men, age 61 ± 14 years, mean CT to CMR interval 10 ± 8 days). Relative to CMR as a reference standard, 45% had LV dilatation. In males, an automated LV diameter measurement of ≥55.5 mm was ≥90% specific for CMR-defined LV dilatation (positive predictive value (PPV) 85.7%, negative predictive value (NPV) 61.2%, accuracy 68.9%). In females, an LV diameter of ≥49.7 mm was ≥90% specific for CMR-defined LV dilatation (PPV 66.7%, NPV 73.1%, accuracy 71.9%). AI CT volumetry data did not significantly improve AUC performance. CONCLUSION Fully automated AI-derived analysis LV dilatation on routine unselected non-gated contrast-enhanced CT thorax studies is feasible. We have defined thresholds for the detection of LV dilatation on CT relative to CMR, which could be used to routinely screen for dilated cardiomyopathy at the time of CT. ADVANCES IN KNOWLEDGE We show, for the first time, that a fully-automated AI-derived analysis of maximal LV chamber axial diameter on non-ECG-gated thoracic CT is feasible in unselected real-world cases and that the derived measures can predict LV dilatation relative to cardiac magnetic resonance imaging, the non-invasive reference standard for determining cardiac chamber size. We have derived sex-specific cut-off values to screen for LV dilatation on routine contrast-enhanced thoracic CT. Future work should validate these thresholds and determine if technology can alter clinical outcomes in a cost-effective manner.
Collapse
Affiliation(s)
- Ashar Asif
- Medical School, University of Bristol, Bristol, UK
| | | | | | | | | | | |
Collapse
|
187
|
Li X, Liu X, Deng X, Fan Y. Interplay between Artificial Intelligence and Biomechanics Modeling in the Cardiovascular Disease Prediction. Biomedicines 2022; 10:2157. [PMID: 36140258 PMCID: PMC9495955 DOI: 10.3390/biomedicines10092157] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the most common cause of morbidity and mortality worldwide, and early accurate diagnosis is the key point for improving and optimizing the prognosis of CVD. Recent progress in artificial intelligence (AI), especially machine learning (ML) technology, makes it possible to predict CVD. In this review, we first briefly introduced the overview development of artificial intelligence. Then we summarized some ML applications in cardiovascular diseases, including ML-based models to directly predict CVD based on risk factors or medical imaging findings and the ML-based hemodynamics with vascular geometries, equations, and methods for indirect assessment of CVD. We also discussed case studies where ML could be used as the surrogate for computational fluid dynamics in data-driven models and physics-driven models. ML models could be a surrogate for computational fluid dynamics, accelerate the process of disease prediction, and reduce manual intervention. Lastly, we briefly summarized the research difficulties and prospected the future development of AI technology in cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaoyin Li
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xiao Liu
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xiaoyan Deng
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
188
|
Singh A, Kwiecinski J, Miller RJH, Otaki Y, Kavanagh PB, Van Kriekinge SD, Parekh T, Gransar H, Pieszko K, Killekar A, Tummala R, Liang JX, Di Carli M, Berman DS, Dey D, Slomka PJ. Deep Learning for Explainable Estimation of Mortality Risk From Myocardial Positron Emission Tomography Images. Circ Cardiovasc Imaging 2022; 15:e014526. [PMID: 36126124 PMCID: PMC10035936 DOI: 10.1161/circimaging.122.014526] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND We aim to develop an explainable deep learning (DL) network for the prediction of all-cause mortality directly from positron emission tomography myocardial perfusion imaging flow and perfusion polar map data and evaluate it using prospective testing. METHODS A total of 4735 consecutive patients referred for stress and rest 82Rb positron emission tomography between 2010 and 2018 were followed up for all-cause mortality for 4.15 (2.24-6.3) years. DL network utilized polar maps of stress and rest perfusion, myocardial blood flow, myocardial flow reserve, and spill-over fraction combined with cardiac volumes, singular indices, and sex. Patients scanned from 2010 to 2016 were used for training and validation. The network was tested in a set of 1135 patients scanned from 2017 to 2018 to simulate prospective clinical implementation. RESULTS In prospective testing, the area under the receiver operating characteristic curve for all-cause mortality prediction by DL (0.82 [95% CI, 0.77-0.86]) was higher than ischemia (0.60 [95% CI, 0.54-0.66]; P <0.001), myocardial flow reserve (0.70 [95% CI, 0.64-0.76], P <0.001) or a comprehensive logistic regression model (0.75 [95% CI, 0.69-0.80], P <0.05). The highest quartile of patients by DL had an annual all-cause mortality rate of 11.87% and had a 16.8 ([95% CI, 6.12%-46.3%]; P <0.001)-fold increase in the risk of death compared with the lowest quartile patients. DL showed a 21.6% overall reclassification improvement as compared with established measures of ischemia. CONCLUSIONS The DL model trained directly on polar maps allows improved patient risk stratification in comparison with established methods for positron emission tomography flow or perfusion assessments.
Collapse
Affiliation(s)
- Ananya Singh
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacek Kwiecinski
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, Warsaw, Poland
| | - Robert JH Miller
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Cardiac Sciences, University of Calgary, Calgary AB, Canada
| | - Yuka Otaki
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul B. Kavanagh
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Serge D. Van Kriekinge
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tejas Parekh
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Heidi Gransar
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Konrad Pieszko
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Interventional Cardiology and Cardiac Surgery, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | - Aditya Killekar
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ramyashree Tummala
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joanna X. Liang
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcelo Di Carli
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Daniel S. Berman
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Damini Dey
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Piotr J. Slomka
- Departments of Medicine (Division of Artificial Intelligence in Medicine), Imaging and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
189
|
Extra-coronary Calcification and Cardiovascular Events: What Do We Know and Where Are We Heading? Curr Atheroscler Rep 2022; 24:755-766. [PMID: 36040566 DOI: 10.1007/s11883-022-01051-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW The coronary artery calcium score is a guideline-endorsed aid for further risk stratification in the primary prevention of atherosclerotic cardiovascular disease. The non-contrast scan performed for detection of coronary artery calcium also gives an opportunity to visualize calcifications in the thoracic aorta and in the heart valves, at no additional cost or radiation exposure. The purpose of this review was to discuss the potential clinical value of measuring thoracic aortic calcification, aortic valve calcification, and mitral annulus calcification. RECENT FINDINGS After two decades of active research, all three calcifications have been extensively evaluated, across various cohorts. We discuss classic and recent studies, current knowledge gaps, and future directions in this space. The added value of these measurements has traditionally been considered modest at best, and they are not currently discussed in relevant primary prevention guidelines in North America and Europe. However, recent studies evaluating high thoracic calcification thresholds and younger populations have further enriched this space. Specifically, some studies suggest that detection of severe thoracic aortic calcification may be helpful in further risk assessment and that detection of aortic valve calcifications may have important prognostic implications in younger individuals. Although more research is needed, particularly in larger young-to-middle-aged cohorts, future guidelines might consider including these features as risk-enhancing factors.
Collapse
|
190
|
Vilas-Boas F, Ribeiro T, Afonso J, Cardoso H, Lopes S, Moutinho-Ribeiro P, Ferreira J, Mascarenhas-Saraiva M, Macedo G. Deep Learning for Automatic Differentiation of Mucinous versus Non-Mucinous Pancreatic Cystic Lesions: A Pilot Study. Diagnostics (Basel) 2022; 12:diagnostics12092041. [PMID: 36140443 PMCID: PMC9498252 DOI: 10.3390/diagnostics12092041] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
Endoscopic ultrasound (EUS) morphology can aid in the discrimination between mucinous and non-mucinous pancreatic cystic lesions (PCLs) but has several limitations that can be overcome by artificial intelligence. We developed a convolutional neural network (CNN) algorithm for the automatic diagnosis of mucinous PCLs. Images retrieved from videos of EUS examinations for PCL characterization were used for the development, training, and validation of a CNN for mucinous cyst diagnosis. The performance of the CNN was measured calculating the area under the receiving operator characteristic curve (AUC), sensitivity, specificity, and positive and negative predictive values. A total of 5505 images from 28 pancreatic cysts were used (3725 from mucinous lesions and 1780 from non-mucinous cysts). The model had an overall accuracy of 98.5%, sensitivity of 98.3%, specificity of 98.9% and AUC of 1. The image processing speed of the CNN was 7.2 ms per frame. We developed a deep learning algorithm that differentiated mucinous and non-mucinous cysts with high accuracy. The present CNN may constitute an important tool to help risk stratify PCLs.
Collapse
Affiliation(s)
- Filipe Vilas-Boas
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Tiago Ribeiro
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
| | - João Afonso
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
| | - Hélder Cardoso
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Susana Lopes
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Pedro Moutinho-Ribeiro
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - João Ferreira
- Department of Mechanical Engineering, Faculty of Engineering of the University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- INEGI—Institute of Science and Innovation in Mechanical and Industrial Engineering, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel Mascarenhas-Saraiva
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- Correspondence:
| | - Guilherme Macedo
- Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- World Gastroenterology Organisation Gastroenterology and Hepatology Training Center, 4200-427 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| |
Collapse
|
191
|
Li Y, Liu PJ, Zhang ZL, Wang YN. Cardiac imaging techniques for the assessment of immune checkpoint inhibitor-induced cardiotoxicity and their potential clinical applications. Am J Cancer Res 2022; 12:3548-3560. [PMID: 36119829 PMCID: PMC9442027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have encouraged a paradigm shift in the clinical management of patients with cancer. Despite the dramatically improved tumor response and patient prognosis, ICIs have been associated with ICI-related myocarditis, which has a high fatality rate. Cardiac imaging plays a critical role in the assessment of cardiac injury. Echocardiography, cardiac magnetic resonance imaging, and targeted tracer-based cardiac molecular imaging techniques alone or in combination reflect pathophysiology and depict different aspects of lesions at different clinical stages, i.e., they have potentially complementary value. Imaging techniques for identifying ICI-induced cardiotoxicity at the early stage may reduce the incidence of adverse cardiovascular events. Particularly in planned ICI therapy among patients with cancer, improved monitoring approaches to identify patients who are at the highest risk of ICI-related myocarditis may help in refining clinical decisions, allowing treatment to be more accurately targeted toward patients who are most likely to benefit. In this study, we systematically reviewed the studies on cardiac imaging techniques for assessing ICI-induced cardiotoxicity. We elaborated about the potential applications of cardiac imaging techniques for the optimized management of patients with ICI-related myocarditis, including risk stratification, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
- Medical Science Research Center (MRC), Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Pei-Jun Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Zhuo-Li Zhang
- Radiological Sciences, Chao Family Comprehensive Cancer Center, University of California (Irvine)USA
| | - Yi-Ning Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
192
|
Mehta C, Shah R, Yanamala N, Sengupta PP. Cardiovascular Imaging Databases: Building Machine Learning Algorithms for Regenerative Medicine. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00216-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
193
|
Chen H, Ouyang D, Baykaner T, Jamal F, Cheng P, Rhee JW. Artificial intelligence applications in cardio-oncology: Leveraging high dimensional cardiovascular data. Front Cardiovasc Med 2022; 9:941148. [PMID: 35958422 PMCID: PMC9360492 DOI: 10.3389/fcvm.2022.941148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Growing evidence suggests a wide spectrum of potential cardiovascular complications following cancer therapies, leading to an urgent need for better risk-stratifying and disease screening in patients undergoing oncological treatment. As many cancer patients undergo frequent surveillance through imaging as well as other diagnostic testing, there is a wealth of information that can be utilized to assess one's risk for cardiovascular complications of cancer therapies. Over the past decade, there have been remarkable advances in applying artificial intelligence (AI) to analyze cardiovascular data obtained from electrocardiograms, echocardiograms, computed tomography, and cardiac magnetic resonance imaging to detect early signs or future risk of cardiovascular diseases. Studies have shown AI-guided cardiovascular image analysis can accurately, reliably and inexpensively identify and quantify cardiovascular risk, leading to better detection of at-risk or disease features, which may open preventive and therapeutic opportunities in cardio-oncology. In this perspective, we discuss the potential for the use of AI in analyzing cardiovascular data to identify cancer patients at risk for cardiovascular complications early in treatment which would allow for rapid intervention to prevent adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Haidee Chen
- City of Hope National Medical Center, Duarte, CA, United States
| | - David Ouyang
- Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Tina Baykaner
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA, United States
| | - Faizi Jamal
- City of Hope National Medical Center, Duarte, CA, United States
| | - Paul Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Palo Alto, CA, United States
| | - June-Wha Rhee
- City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
194
|
Wellnhofer E. Real-World and Regulatory Perspectives of Artificial Intelligence in Cardiovascular Imaging. Front Cardiovasc Med 2022; 9:890809. [PMID: 35935648 PMCID: PMC9354141 DOI: 10.3389/fcvm.2022.890809] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/13/2022] [Indexed: 12/02/2022] Open
Abstract
Recent progress in digital health data recording, advances in computing power, and methodological approaches that extract information from data as artificial intelligence are expected to have a disruptive impact on technology in medicine. One of the potential benefits is the ability to extract new and essential insights from the vast amount of data generated during health care delivery every day. Cardiovascular imaging is boosted by new intelligent automatic methods to manage, process, segment, and analyze petabytes of image data exceeding historical manual capacities. Algorithms that learn from data raise new challenges for regulatory bodies. Partially autonomous behavior and adaptive modifications and a lack of transparency in deriving evidence from complex data pose considerable problems. Controlling new technologies requires new controlling techniques and ongoing regulatory research. All stakeholders must participate in the quest to find a fair balance between innovation and regulation. The regulatory approach to artificial intelligence must be risk-based and resilient. A focus on unknown emerging risks demands continuous surveillance and clinical evaluation during the total product life cycle. Since learning algorithms are data-driven, high-quality data is fundamental for good machine learning practice. Mining, processing, validation, governance, and data control must account for bias, error, inappropriate use, drifts, and shifts, particularly in real-world data. Regulators worldwide are tackling twenty-first century challenges raised by "learning" medical devices. Ethical concerns and regulatory approaches are presented. The paper concludes with a discussion on the future of responsible artificial intelligence.
Collapse
Affiliation(s)
- Ernst Wellnhofer
- Institute of Computer-Assisted Cardiovascular Medicine, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
195
|
Rios R, Miller RJH, Hu LH, Otaki Y, Singh A, Diniz M, Sharir T, Einstein AJ, Fish MB, Ruddy TD, Kaufmann PA, Sinusas AJ, Miller EJ, Bateman TM, Dorbala S, DiCarli M, Van Kriekinge S, Kavanagh P, Parekh T, Liang JX, Dey D, Berman DS, Slomka P. Determining a minimum set of variables for machine learning cardiovascular event prediction: results from REFINE SPECT registry. Cardiovasc Res 2022; 118:2152-2164. [PMID: 34259870 PMCID: PMC9302886 DOI: 10.1093/cvr/cvab236] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS Optimal risk stratification with machine learning (ML) from myocardial perfusion imaging (MPI) includes both clinical and imaging data. While most imaging variables can be derived automatically, clinical variables require manual collection, which is time-consuming and prone to error. We determined the fewest manually input and imaging variables required to maintain the prognostic accuracy for major adverse cardiac events (MACE) in patients undergoing a single-photon emission computed tomography (SPECT) MPI. METHODS AND RESULTS This study included 20 414 patients from the multicentre REFINE SPECT registry and 2984 from the University of Calgary for training and external testing of the ML models, respectively. ML models were trained using all variables (ML-All) and all image-derived variables (including age and sex, ML-Image). Next, ML models were sequentially trained by incrementally adding manually input and imaging variables to baseline ML models based on their importance ranking. The fewest variables were determined as the ML models (ML-Reduced, ML-Minimum, and ML-Image-Reduced) that achieved comparable prognostic performance to ML-All and ML-Image. Prognostic accuracy of the ML models was compared with visual diagnosis, stress total perfusion deficit (TPD), and traditional multivariable models using area under the receiver-operating characteristic curve (AUC). ML-Minimum (AUC 0.798) obtained comparable prognostic accuracy to ML-All (AUC 0.799, P = 0.19) by including 12 of 40 manually input variables and 11 of 58 imaging variables. ML-Reduced achieved comparable accuracy (AUC 0.796) with a reduced set of manually input variables and all imaging variables. In external validation, the ML models also obtained comparable or higher prognostic accuracy than traditional multivariable models. CONCLUSION Reduced ML models, including a minimum set of manually collected or imaging variables, achieved slightly lower accuracy compared to a full ML model but outperformed standard interpretation methods and risk models. ML models with fewer collected variables may be more practical for clinical implementation.
Collapse
Affiliation(s)
- Richard Rios
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Robert J H Miller
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - Lien Hsin Hu
- Department of Nuclear Medicine, Taipei, Veterans General Hospital, Taipei, Taiwan
| | - Yuka Otaki
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ananya Singh
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marcio Diniz
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tali Sharir
- Department of Nuclear Cardiology, Assuta Medical Center, Tel Aviv, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Andrew J Einstein
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
- Department of Radiology, Columbia University Irving Medical Center and New York-Presbyterian Hospital, New York, NY, USA
| | - Mathews B Fish
- Department of Nuclear Medicine, Oregon Heart and Vascular Institute, Sacred Heart Medical Center, Springfield, OR, USA
| | - Terrence D Ruddy
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa ON, Canada
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, Cardiac Imaging, University Hospital Zurich, Zurich, Switzerland
| | - Albert J Sinusas
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Edward J Miller
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Sharmila Dorbala
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Marcelo DiCarli
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Serge Van Kriekinge
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul Kavanagh
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tejas Parekh
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joanna X Liang
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Damini Dey
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Daniel S Berman
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Piotr Slomka
- Department of Imaging, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
196
|
Human vs Artificial Intelligence-Based Echocardiography Analysis as Predictor of Outcomes: An analysis from the World Alliance Societies of Echocardiography COVID study. J Am Soc Echocardiogr 2022; 35:1226-1237.e7. [PMID: 35863542 PMCID: PMC9293371 DOI: 10.1016/j.echo.2022.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 05/15/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022]
Abstract
Background Transthoracic echocardiography is the leading cardiac imaging modality for patients admitted with COVID-19, a condition of high short-term mortality. The aim of this study was to test the hypothesis that artificial intelligence (AI)–based analysis of echocardiographic images could predict mortality more accurately than conventional analysis by a human expert. Methods Patients admitted to 13 hospitals for acute COVID-19 who underwent transthoracic echocardiography were included. Left ventricular ejection fraction (LVEF) and left ventricular longitudinal strain (LVLS) were obtained manually by multiple expert readers and by automated AI software. The ability of the manual and AI analyses to predict all-cause mortality was compared. Results In total, 870 patients were enrolled. The mortality rate was 27.4% after a mean follow-up period of 230 ± 115 days. AI analysis had lower variability than manual analysis for both LVEF (P = .003) and LVLS (P = .005). AI-derived LVEF and LVLS were predictors of mortality in univariable and multivariable regression analysis (odds ratio, 0.974 [95% CI, 0.956-0.991; P = .003] for LVEF; odds ratio, 1.060 [95% CI, 1.019-1.105; P = .004] for LVLS), but LVEF and LVLS obtained by manual analysis were not. Direct comparison of the predictive value of AI versus manual measurements of LVEF and LVLS showed that AI was significantly better (P = .005 and P = .003, respectively). In addition, AI-derived LVEF and LVLS had more significant and stronger correlations to other objective biomarkers of acute disease than manual reads. Conclusions AI-based analysis of LVEF and LVLS had similar feasibility as manual analysis, minimized variability, and consequently increased the statistical power to predict mortality. AI-based, but not manual, analyses were a significant predictor of in-hospital and follow-up mortality.
Collapse
|
197
|
Dell'Angela L, Nicolosi GL. Artificial intelligence applied to cardiovascular imaging, a critical focus on echocardiography: The point-of-view from "the other side of the coin". JOURNAL OF CLINICAL ULTRASOUND : JCU 2022; 50:772-780. [PMID: 35466409 DOI: 10.1002/jcu.23215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular imaging has achieved a crucial role in the management of cardiovascular diseases. In this field, echocardiography advantages include wide availability, portability, and affordability, at a relatively low cost. However, echocardiographic assessment requires highly trained operators, and implies high observer variability, as compared with the other cardiac imaging modalities. Hence, artificial intelligence might be extremely helpful. From the point-of-view of the peripheral "Spoke" Hospital potential user ("the other side of the coin"), artificial intelligence development appears very slow in the clinical arena. Many limitations are still present, and require full involvement, cooperation, and coordination of professional operators into Hub-and-Spoke network.
Collapse
Affiliation(s)
- Luca Dell'Angela
- Emergency Department, Cardiology Division, Gorizia & Monfalcone Hospital, ASUGI, Gorizia, Italy
| | | |
Collapse
|
198
|
Huang Z, Xiao J, Wang X, Li Z, Guo N, Hu Y, Li X, Wang X. Clinical Evaluation of the Automatic Coronary Artery Disease Reporting and Data System (CAD-RADS) in Coronary Computed Tomography Angiography Using Convolutional Neural Networks. Acad Radiol 2022; 30:698-706. [PMID: 35753936 DOI: 10.1016/j.acra.2022.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/01/2022]
Abstract
RATIONALE AND OBJECTIVES The coronary artery disease reporting and data system (CAD-RADS™) was recently introduced to standardise reporting. We aimed to evaluate the utility of an automatic postprocessing and reporting system based on CAD-RADS™ in suspected coronary artery disease (CAD) patients. MATERIALS AND METHODS Clinical evaluation was performed in 346 patients who underwent coronary computed tomography angiography (CCTA). We compared deep learning (DL)-based CCTA with human readers for evaluation of CAD-RADS™ with commercially-available automated segmentation and manual postprocessing in a retrospective validation cohort. RESULTS Compared with invasive coronary angiography, the sensitivity, specificity, positive predictive value, negative predictive value, and accuracy of the DL model for diagnosis of CAD were 79.02%, 86.52%, 89.50%, 73.94%, and 82.08%, respectively. There was no significant difference between the DL-based and the reader-based CAD-RADS™ grading of CCTA results. Consistency testing showed that the Kappa value between the model and the readers was 0.775 (95% confidence interval [CI]: 0.728-0.823, p < 0.001), 0.802 (95% CI: 0.756-0.847, p < 0.001), and 0.796 (95% CI: 0.750-0.843, p < 0.001), respectively. This system reduces the time taken from 14.97 ± 1.80 min to 5.02 ± 0.8 min (p < 0.001). CONCLUSION The standardised reporting of DL-based CAD-RADS™ in CCTA can accurately and rapidly evaluate suspected CAD patients, and has good consistency with grading by radiologists.
Collapse
Affiliation(s)
- Zengfa Huang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianwei Xiao
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Wang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuoqin Li
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Guo
- Shukun (Beijing) Technology Co., Ltd., Jinhui Building, Qiyang Road, 100102 Beijing, China
| | - Yun Hu
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Li
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Wang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
199
|
Prediction of All-Cause Mortality Following Percutaneous Coronary Intervention in Bifurcation Lesions Using Machine Learning Algorithms. J Pers Med 2022; 12:jpm12060990. [PMID: 35743777 PMCID: PMC9224705 DOI: 10.3390/jpm12060990] [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: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Stratifying prognosis following coronary bifurcation percutaneous coronary intervention (PCI) is an unmet clinical need that may be fulfilled through the adoption of machine learning (ML) algorithms to refine outcome predictions. We sought to develop an ML-based risk stratification model built on clinical, anatomical, and procedural features to predict all-cause mortality following contemporary bifurcation PCI. Multiple ML models to predict all-cause mortality were tested on a cohort of 2393 patients (training, n = 1795; internal validation, n = 598) undergoing bifurcation PCI with contemporary stents from the real-world RAIN registry. Twenty-five commonly available patient-/lesion-related features were selected to train ML models. The best model was validated in an external cohort of 1701 patients undergoing bifurcation PCI from the DUTCH PEERS and BIO-RESORT trial cohorts. At ROC curves, the AUC for the prediction of 2-year mortality was 0.79 (0.74–0.83) in the overall population, 0.74 (0.62–0.85) at internal validation and 0.71 (0.62–0.79) at external validation. Performance at risk ranking analysis, k-center cross-validation, and continual learning confirmed the generalizability of the models, also available as an online interface. The RAIN-ML prediction model represents the first tool combining clinical, anatomical, and procedural features to predict all-cause mortality among patients undergoing contemporary bifurcation PCI with reliable performance.
Collapse
|
200
|
Functional Evaluation of Intermediate Coronary Lesions with Integrated Computed Tomography Angiography and Invasive Angiography in Patients with Stable Coronary Artery Disease. J Transl Int Med 2022; 10:255-263. [PMID: 36776233 PMCID: PMC9901557 DOI: 10.2478/jtim-2022-0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background and objectives The hemodynamic evaluation of coronary stenoses undergoes a transition from wire-based invasive measurements to image-based computational assessments. However, fractional flow reserve (FFR) values derived from coronary CT angiography (CCTA) and angiography-based quantitative flow ratio have certain limitations in accuracy and efficiency, preventing their widespread use in routine practice. Hence, we aimed to investigate the diagnostic performance of FFR derived from the integration of CCTA and invasive angiography (FFRCT-angio) with artificial intelligence assistance in patients with stable coronary artery disease (CAD). Methods Forty stable CAD patients with 67 target vessels (50%-90% diameter stenosis) were included in this single-center retrospective study. All patients underwent CCTA followed by coronary angiography with FFR measurement within 30 days. Both CCTA and angiographic images were combined to generate a three-dimensional reconstruction of the coronary arteries using artificial intelligence. Subsequently, functional assessment was performed through a deep learning algorithm. FFR was used as the reference. Results FFRCT-angio values were significantly correlated with FFR values (r = 0.81, P < 0.001, Spearman analysis). Per-vessel diagnostic accuracy of FFRCT-angio was 92.54%. Sensitivity and specificity in identifying ischemic lesions were 100% and 88.10%, respectively. Positive predictive value and negative predictive value were 83.33% and 100%, respectively. Moreover, the diagnostic performance of FFRCT-angio was satisfactory in different target vessels and different segment lesions. Conclusions FFRCT-angio exhibits excellent diagnostic performance of identifying ischemic lesions in patients with stable CAD. Combining CCTA and angiographic imaging, FFRCT-angio may represent an effective and practical alternative to invasive FFR in selected patients.
Collapse
|