1
|
Glass M, Ji Z, Davis R, Pavlisko EN, DiBernardo L, Carney J, Fishbein G, Luthringer D, Miller D, Mitchell R, Larsen B, Butt Y, Bois M, Maleszewski J, Halushka M, Seidman M, Lin CY, Buja M, Stone J, Dov D, Carin L, Glass C. A machine learning algorithm improves the diagnostic accuracy of the histologic component of antibody mediated rejection (AMR-H) in cardiac transplant endomyocardial biopsies. Cardiovasc Pathol 2024; 72:107646. [PMID: 38677634 DOI: 10.1016/j.carpath.2024.107646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Pathologic antibody mediated rejection (pAMR) remains a major driver of graft failure in cardiac transplant patients. The endomyocardial biopsy remains the primary diagnostic tool but presents with challenges, particularly in distinguishing the histologic component (pAMR-H) defined by 1) intravascular macrophage accumulation in capillaries and 2) activated endothelial cells that expand the cytoplasm to narrow or occlude the vascular lumen. Frequently, pAMR-H is difficult to distinguish from acute cellular rejection (ACR) and healing injury. With the advent of digital slide scanning and advances in machine deep learning, artificial intelligence technology is widely under investigation in the areas of oncologic pathology, but in its infancy in transplant pathology. For the first time, we determined if a machine learning algorithm could distinguish pAMR-H from normal myocardium, healing injury and ACR. MATERIALS AND METHODS A total of 4,212 annotations (1,053 regions of normal, 1,053 pAMR-H, 1,053 healing injury and 1,053 ACR) were completed from 300 hematoxylin and eosin slides scanned using a Leica Aperio GT450 digital whole slide scanner at 40X magnification. All regions of pAMR-H were annotated from patients confirmed with a previous diagnosis of pAMR2 (>50% positive C4d immunofluorescence and/or >10% CD68 positive intravascular macrophages). Annotations were imported into a Python 3.7 development environment using the OpenSlide™ package and a convolutional neural network approach utilizing transfer learning was performed. RESULTS The machine learning algorithm showed 98% overall validation accuracy and pAMR-H was correctly distinguished from specific categories with the following accuracies: normal myocardium (99.2%), healing injury (99.5%) and ACR (99.5%). CONCLUSION Our novel deep learning algorithm can reach acceptable, and possibly surpass, performance of current diagnostic standards of identifying pAMR-H. Such a tool may serve as an adjunct diagnostic aid for improving the pathologist's accuracy and reproducibility, especially in difficult cases with high inter-observer variability. This is one of the first studies that provides evidence that an artificial intelligence machine learning algorithm can be trained and validated to diagnose pAMR-H in cardiac transplant patients. Ongoing studies include multi-institutional verification testing to ensure generalizability.
Collapse
Affiliation(s)
- Matthew Glass
- Duke Division of Artificial Intelligence and Computational Pathology, Duke University Medical Center, Durham NC, USA; Department of Anesthesiology, Duke University Medical Center, Durham NC, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke School of Medicine, Durham NC, USA
| | - Richard Davis
- Department of Pathology, Duke University Medical Center, Durham NC, USA
| | - Elizabeth N Pavlisko
- Duke Division of Artificial Intelligence and Computational Pathology, Duke University Medical Center, Durham NC, USA; Department of Pathology, Duke University Medical Center, Durham NC, USA
| | - Louis DiBernardo
- Department of Pathology, Duke University Medical Center, Durham NC, USA
| | - John Carney
- Department of Pathology, Duke University Medical Center, Durham NC, USA
| | - Gregory Fishbein
- Department of Pathology, University of California at Los Angeles, Los Angeles CA, USA
| | - Daniel Luthringer
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles CA, USA
| | - Dylan Miller
- Department of Pathology, Intermountain Healthcare, Salt Lake City UT, USA
| | - Richard Mitchell
- Department of Pathology, Brigham and Women's Hospital, Boston MA, USA
| | - Brandon Larsen
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Phoenix AZ, USA
| | - Yasmeen Butt
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Phoenix AZ, USA
| | - Melanie Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN, USA
| | - Joseph Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN, USA
| | - Marc Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Michael Seidman
- Department of Pathology, University Health Network, Toronto ON, CA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University, St. Louis MO, USA
| | - Maximilian Buja
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston TX, USA
| | - James Stone
- Department of Pathology, Massachusetts General Hospital, Boston MA, USA
| | - David Dov
- Duke Division of Artificial Intelligence and Computational Pathology, Duke University Medical Center, Durham NC, USA; Pratt School of Engineering, Department of Electrical and Computer Engineering, Duke University, Durham NC, USA
| | - Lawrence Carin
- Duke Division of Artificial Intelligence and Computational Pathology, Duke University Medical Center, Durham NC, USA; Pratt School of Engineering, Department of Electrical and Computer Engineering, Duke University, Durham NC, USA
| | - Carolyn Glass
- Duke Division of Artificial Intelligence and Computational Pathology, Duke University Medical Center, Durham NC, USA; Department of Pathology, Duke University Medical Center, Durham NC, USA.
| |
Collapse
|
2
|
Lyu X, Dong L, Fan Z, Sun Y, Zhang X, Liu N, Wang D. Artificial intelligence-based graded training of pulmonary nodules for junior radiology residents and medical imaging students. BMC MEDICAL EDUCATION 2024; 24:740. [PMID: 38982410 PMCID: PMC11234785 DOI: 10.1186/s12909-024-05723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND To evaluate the efficiency of artificial intelligence (AI)-assisted diagnosis system in the pulmonary nodule detection and diagnosis training of junior radiology residents and medical imaging students. METHODS The participants were divided into three groups. Medical imaging students of Grade 2020 in the Jinzhou Medical University were randomly divided into Groups 1 and 2; Group 3 comprised junior radiology residents. Group 1 used the traditional case-based teaching mode; Groups 2 and 3 used the 'AI intelligent assisted diagnosis system' teaching mode. All participants performed localisation, grading and qualitative diagnosed of 1,057 lung nodules in 420 cases for seven rounds of testing after training. The sensitivity and number of false positive nodules in different densities (solid, pure ground glass, mixed ground glass and calcification), sizes (less than 5 mm, 5-10 mm and over 10 mm) and positions (subpleural, peripheral and central) of the pulmonary nodules in the three groups were detected. The pathological results and diagnostic opinions of radiologists formed the criteria. The detection rate, diagnostic compliance rate, false positive number/case, and kappa scores of the three groups were compared. RESULTS There was no statistical difference in baseline test scores between Groups 1 and 2, and there were statistical differences with Group 3 (P = 0.036 and 0.011). The detection rate of solid, pure ground glass and calcified nodules; small-, medium-, and large-diameter nodules; and peripheral nodules were significantly different among the three groups (P<0.05). After seven rounds of training, the diagnostic compliance rate increased in all three groups, with the largest increase in Group 2. The average kappa score increased from 0.508 to 0.704. The average kappa score for Rounds 1-4 and 5-7 were 0.595 and 0.714, respectively. The average kappa scores of Groups 1,2 and 3 increased from 0.478 to 0.658, 0.417 to 0.757, and 0.638 to 0.791, respectively. CONCLUSION The AI assisted diagnosis system is a valuable tool for training junior radiology residents and medical imaging students to perform pulmonary nodules detection and diagnosis.
Collapse
Affiliation(s)
- Xiaohong Lyu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liang Dong
- School of Electrical Engineering, Liaoning University of Technology, Jinzhou, China
| | - Zhongkai Fan
- Office of Educational Administration, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yu Sun
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xianglin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ning Liu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| | - Dongdong Wang
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
3
|
Pavlisko EN, Adam BA, Berry GJ, Calabrese F, Cortes-Santiago N, Glass CH, Goddard M, Greenland JR, Kreisel D, Levine DJ, Martinu T, Verleden SE, Weigt SS, Roux A. The 2022 Banff Meeting Lung Report. Am J Transplant 2024; 24:542-548. [PMID: 37931751 DOI: 10.1016/j.ajt.2023.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
The Lung Session of the 2022 16th Banff Foundation for Allograft Pathology Conference-held in Banff, Alberta-focused on non-rejection lung allograft pathology and novel technologies for the detection of allograft injury. A multidisciplinary panel reviewed the state-of-the-art of current histopathologic entities, serologic studies, and molecular practices, as well as novel applications of digital pathology with artificial intelligence, gene expression analysis, and quantitative image analysis of chest computerized tomography. Current states of need as well as prospective integration of the aforementioned tools and technologies for complete assessment of allograft injury and its impact on lung transplant outcomes were discussed. Key conclusions from the discussion were: (1) recognition of limitations in current standard of care assessment of lung allograft dysfunction; (2) agreement on the need for a consensus regarding the standardized approach to the collection and assessment of pathologic data, inclusive of all lesions associated with graft outcome (eg, non-rejection pathology); and (3) optimism regarding promising novel diagnostic modalities, especially minimally invasive, which should be integrated into large, prospective multicenter studies to further evaluate their utility in clinical practice for directing personalized therapies to improve graft outcomes.
Collapse
Affiliation(s)
- Elizabeth N Pavlisko
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA.
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Gerald J Berry
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Padova, Italy
| | - Nahir Cortes-Santiago
- Department of Pathology and Immunology, Texas Children's Hospital, Houston, Texas, USA
| | - Carolyn H Glass
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Martin Goddard
- Pathology Department, Royal Papworth Hospital, NHS Trust, Papworth Everard, Cambridge, UK
| | - John R Greenland
- Department of Medicine, University of California, San Francisco, USA; Veterans Affairs Health Care System, San Francisco, California, USA
| | - Daniel Kreisel
- Department of Surgery, Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Deborah J Levine
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, California, USA
| | - Tereza Martinu
- Division of Respirology, Department of Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada; Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Stijn E Verleden
- Lung Transplant Unit, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium; Department of ASTARC, University of Antwerp, Wilrijk, Belgium
| | - S Sam Weigt
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Antoine Roux
- Department of Respiratory Medicine, Foch Hospital, Suresnes, France
| |
Collapse
|
4
|
Kveton M, Hudec L, Vykopal I, Halinkovic M, Laco M, Felsoova A, Benesova W, Fabian O. Digital pathology in cardiac transplant diagnostics: from biopsies to algorithms. Cardiovasc Pathol 2024; 68:107587. [PMID: 37926351 DOI: 10.1016/j.carpath.2023.107587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023] Open
Abstract
In the field of heart transplantation, the ability to accurately and promptly diagnose cardiac allograft rejection is crucial. This comprehensive review explores the transformative role of digital pathology and computational pathology, especially through machine learning, in this critical domain. These methodologies harness large datasets to extract subtle patterns and valuable information that extend beyond human perceptual capabilities, potentially enhancing diagnostic outcomes. Current research indicates that these computer-based systems could offer accuracy and performance matching, or even exceeding, that of expert pathologists, thereby introducing more objectivity and reducing observer variability. Despite promising results, several challenges such as limited sample sizes, diverse data sources, and the absence of standardized protocols pose significant barriers to the widespread adoption of these techniques. The future of digital pathology in heart transplantation diagnostics depends on utilizing larger, more diverse patient cohorts, standardizing data collection, processing, and evaluation protocols, and fostering collaborative research efforts. The integration of various data types, including clinical, demographic, and imaging information, could further refine diagnostic precision. As researchers address these challenges and promote collaborative efforts, digital pathology has the potential to become an integral part of clinical practice, ultimately improving patient care in heart transplantation.
Collapse
Affiliation(s)
- Martin Kveton
- Third Faculty of Medicine, Charles University, Prague, Czech Republic; Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Lukas Hudec
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Ivan Vykopal
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Matej Halinkovic
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Miroslav Laco
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Andrea Felsoova
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Histology and Embryology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Wanda Benesova
- Faculty of Informatics and Information Technologies, Slovak University of Technology, Bratislava, Slovakia
| | - Ondrej Fabian
- Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic; Department of Pathology and Molecular Medicine, Third Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| |
Collapse
|
5
|
Jacquemyn X, Kutty S, Manlhiot C. The Lifelong Impact of Artificial Intelligence and Clinical Prediction Models on Patients With Tetralogy of Fallot. CJC PEDIATRIC AND CONGENITAL HEART DISEASE 2023; 2:440-452. [PMID: 38161675 PMCID: PMC10755786 DOI: 10.1016/j.cjcpc.2023.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/24/2023] [Indexed: 01/03/2024]
Abstract
Medical advancements in the diagnosis, surgical techniques, perioperative care, and continued care throughout childhood have transformed the outlook for individuals with tetralogy of Fallot (TOF), improving survival and shifting the perspective towards lifelong care. However, with a growing population of survivors, longstanding challenges have been accentuated, and new challenges have surfaced, necessitating a re-evaluation of TOF care. Availability of prenatal diagnostics, insufficient information from traditional imaging techniques, previously unforeseen medical complications, and debates surrounding optimal timing and indications for reintervention are among the emerging issues. To address these challenges, the integration of artificial intelligence and machine learning holds great promise as they have the potential to revolutionize patient management and positively impact lifelong outcomes for individuals with TOF. Innovative applications of artificial intelligence and machine learning have spanned across multiple domains of TOF care, including screening and diagnosis, automated image processing and interpretation, clinical risk stratification, and planning and performing cardiac interventions. By embracing these advancements and incorporating them into routine clinical practice, personalized medicine could be delivered, leading to the best possible outcomes for patients. In this review, we provide an overview of these evolving applications and emphasize the challenges, limitations, and future potential for integrating them into clinical care.
Collapse
Affiliation(s)
- Xander Jacquemyn
- Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Shelby Kutty
- Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Cedric Manlhiot
- Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Kawaguchi N, Nakanishi T. Animal Disease Models and Patient-iPS-Cell-Derived In Vitro Disease Models for Cardiovascular Biology-How Close to Disease? BIOLOGY 2023; 12:468. [PMID: 36979160 PMCID: PMC10045735 DOI: 10.3390/biology12030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023]
Abstract
Currently, zebrafish, rodents, canines, and pigs are the primary disease models used in cardiovascular research. In general, larger animals have more physiological similarities to humans, making better disease models. However, they can have restricted or limited use because they are difficult to handle and maintain. Moreover, animal welfare laws regulate the use of experimental animals. Different species have different mechanisms of disease onset. Organs in each animal species have different characteristics depending on their evolutionary history and living environment. For example, mice have higher heart rates than humans. Nonetheless, preclinical studies have used animals to evaluate the safety and efficacy of human drugs because no other complementary method exists. Hence, we need to evaluate the similarities and differences in disease mechanisms between humans and experimental animals. The translation of animal data to humans contributes to eliminating the gap between these two. In vitro disease models have been used as another alternative for human disease models since the discovery of induced pluripotent stem cells (iPSCs). Human cardiomyocytes have been generated from patient-derived iPSCs, which are genetically identical to the derived patients. Researchers have attempted to develop in vivo mimicking 3D culture systems. In this review, we explore the possible uses of animal disease models, iPSC-derived in vitro disease models, humanized animals, and the recent challenges of machine learning. The combination of these methods will make disease models more similar to human disease.
Collapse
Affiliation(s)
- Nanako Kawaguchi
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Tokyo Women’s Medical University, Tokyo 162-8666, Japan;
| | | |
Collapse
|
7
|
Abstract
Machine learning and artificial intelligence approaches have revolutionized multiple disciplines, including toxicology. This review summarizes representative recent applications of machine learning and artificial intelligence approaches in different areas of toxicology, including physiologically based pharmacokinetic (PBPK) modeling, quantitative structure-activity relationship modeling for toxicity prediction, adverse outcome pathway analysis, high-throughput screening, toxicogenomics, big data and toxicological databases. By leveraging machine learning and artificial intelligence approaches, now it is possible to develop PBPK models for hundreds of chemicals efficiently, to create in silico models to predict toxicity for a large number of chemicals with similar accuracies compared to in vivo animal experiments, and to analyze a large amount of different types of data (toxicogenomics, high-content image data, etc.) to generate new insights into toxicity mechanisms rapidly, which was impossible by manual approaches in the past. To continue advancing the field of toxicological sciences, several challenges should be considered: (1) not all machine learning models are equally useful for a particular type of toxicology data, and thus it is important to test different methods to determine the optimal approach; (2) current toxicity prediction is mainly on bioactivity classification (yes/no), so additional studies are needed to predict the intensity of effect or dose-response relationship; (3) as more data become available, it is crucial to perform rigorous data quality check and develop infrastructure to store, share, analyze, evaluate, and manage big data; and (4) it is important to convert machine learning models to user-friendly interfaces to facilitate their applications by both computational and bench scientists.
Collapse
Affiliation(s)
- Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL, 32608, USA
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, 32610, USA.,Center for Environmental and Human Toxicology, University of Florida, FL, 32608, USA
| |
Collapse
|
8
|
Nattel S. Digital Technologies: Revolutionizing Cardiovascular Medicine and Reshaping the World. Can J Cardiol 2021; 38:142-144. [PMID: 34954008 DOI: 10.1016/j.cjca.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France.
| |
Collapse
|