1
|
Lei C, Sun W, Wang K, Weng R, Kan X, Li R. Artificial intelligence-assisted diagnosis of early gastric cancer: present practice and future prospects. Ann Med 2025; 57:2461679. [PMID: 39928093 PMCID: PMC11812113 DOI: 10.1080/07853890.2025.2461679] [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: 10/08/2024] [Revised: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Gastric cancer (GC) occupies the first few places in the world among tumors in terms of incidence and mortality, causing serious harm to human health, and at the same time, its treatment greatly consumes the health care resources of all countries in the world. The diagnosis of GC is usually based on histopathologic examination, and it is very important to be able to detect and identify cancerous lesions at an early stage, but some endoscopists' lack of diagnostic experience and fatigue at work lead to a certain rate of under diagnosis. The rapid and striking development of Artificial intelligence (AI) has helped to enhance the ability to extract abnormal information from endoscopic images to some extent, and more and more researchers are applying AI technology to the diagnosis of GC. This initiative has not only improved the detection rate of early gastric cancer (EGC), but also significantly improved the survival rate of patients after treatment. This article reviews the results of various AI-assisted diagnoses of EGC in recent years, including the identification of EGC, the determination of differentiation type and invasion depth, and the identification of borders. Although AI has a better application prospect in the early diagnosis of ECG, there are still major challenges, and the prospects and limitations of AI application need to be further discussed.
Collapse
Affiliation(s)
- Changda Lei
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Wenqiang Sun
- Suzhou Medical College, Soochow University, Suzhou, China
- Department of Neonatology, Children’s Hospital of Soochow University, Suzhou, China
| | - Kun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Ruixia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Xiuji Kan
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Kemper EHM, Erenstein H, Boverhof BJ, Redekop K, Andreychenko AE, Dietzel M, Groot Lipman KBW, Huisman M, Klontzas ME, Vos F, IJzerman M, Starmans MPA, Visser JJ. ESR Essentials: how to get to valuable radiology AI: the role of early health technology assessment-practice recommendations by the European Society of Medical Imaging Informatics. Eur Radiol 2025; 35:3432-3441. [PMID: 39636421 PMCID: PMC12081502 DOI: 10.1007/s00330-024-11188-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/30/2024] [Accepted: 09/16/2024] [Indexed: 12/07/2024]
Abstract
AI tools in radiology are revolutionising the diagnosis, evaluation, and management of patients. However, there is a major gap between the large number of developed AI tools and those translated into daily clinical practice, which can be primarily attributed to limited usefulness and trust in current AI tools. Instead of technically driven development, little effort has been put into value-based development to ensure AI tools will have a clinically relevant impact on patient care. An iterative comprehensive value evaluation process covering the complete AI tool lifecycle should be part of radiology AI development. For value assessment of health technologies, health technology assessment (HTA) is an extensively used and comprehensive method. While most aspects of value covered by HTA apply to radiology AI, additional aspects, including transparency, explainability, and robustness, are unique to radiology AI and crucial in its value assessment. Additionally, value assessment should already be included early in the design stage to determine the potential impact and subsequent requirements of the AI tool. Such early assessment should be systematic, transparent, and practical to ensure all stakeholders and value aspects are considered. Hence, early value-based development by incorporating early HTA will lead to more valuable AI tools and thus facilitate translation to clinical practice. CLINICAL RELEVANCE STATEMENT: This paper advocates for the use of early value-based assessments. These assessments promote a comprehensive evaluation on how an AI tool in development can provide value in clinical practice and thus help improve the quality of these tools and the clinical process they support. KEY POINTS: Value in radiology AI should be perceived as a comprehensive term including health technology assessment domains and AI-specific domains. Incorporation of an early health technology assessment for radiology AI during development will lead to more valuable radiology AI tools. Comprehensive and transparent value assessment of radiology AI tools is essential for their widespread adoption.
Collapse
Affiliation(s)
- Erik H M Kemper
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hendrik Erenstein
- Department of Medical Imaging and Radiation Therapy, The Hanze University of Applied Sciences, Groningen, The Netherlands
- Department of Radiotherapy, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Research Group Healthy Ageing, Allied Health Care and Nursing, The Hanze University of Applied Sciences, Groningen, The Netherlands
| | - Bart-Jan Boverhof
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Ken Redekop
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | | | - Matthias Dietzel
- Department of Radiology, University Hospital Erlangen, Erlangen, Germany
| | - Kevin B W Groot Lipman
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Merel Huisman
- Radboud University Medical Center, Department of Radiology and Nuclear Medicine, Nijmegen, The Netherlands
| | - Michail E Klontzas
- Department of Radiology, School of Medicine, University of Crete, Heraklion, Greece
- Division of Radiology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Computational Biomedicine Laboratory, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece
| | - Frans Vos
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Maarten IJzerman
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Martijn P A Starmans
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pathology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jacob J Visser
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Strand F. AI image analysis as the basis for risk-stratified screening. Jpn J Radiol 2025; 43:927-933. [PMID: 39794661 DOI: 10.1007/s11604-025-01734-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Artificial intelligence (AI) has emerged as a transformative tool in breast cancer screening, with two distinct applications: computer-aided cancer detection (CAD) and risk prediction. While AI CAD systems are slowly finding its way into clinical practice to assist radiologists or make independent reads, this review focuses on AI risk models, which aim to predict a patient's likelihood of being diagnosed with breast cancer within a few years after negative screening. Unlike AI CAD systems, AI risk models are mainly explored in research settings without widespread clinical adoption. This review synthesizes advances in AI-driven risk prediction models, from traditional imaging biomarkers to cutting-edge deep learning methodologies and multimodal approaches. Contributions by leading researchers are explored with critical appraisal of their methods and findings. Ethical, practical, and clinical challenges in implementing AI models are also discussed, with an emphasis on real-world applications. This review concludes by proposing future directions to optimize the adoption of AI tools in breast cancer screening and improve equity and outcomes for diverse populations.
Collapse
Affiliation(s)
- Fredrik Strand
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden.
- Medical Diagnostics Karolinska, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
4
|
Loaiza-Bonilla A, Thaker N, Chung C, Parikh RB, Stapleton S, Borkowski P. Driving Knowledge to Action: Building a Better Future With Artificial Intelligence-Enabled Multidisciplinary Oncology. Am Soc Clin Oncol Educ Book 2025; 45:e100048. [PMID: 40315375 DOI: 10.1200/edbk-25-100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Artificial intelligence (AI) is transforming multidisciplinary oncology at an unprecedented pace, redefining how clinicians detect, classify, and treat cancer. From earlier and more accurate diagnoses to personalized treatment planning, AI's impact is evident across radiology, pathology, radiation oncology, and medical oncology. By leveraging vast and diverse data-including imaging, genomic, clinical, and real-world evidence-AI algorithms can uncover complex patterns, accelerate drug discovery, and help identify optimal treatment regimens for each patient. However, realizing the full potential of AI also necessitates addressing concerns regarding data quality, algorithmic bias, explainability, privacy, and regulatory oversight-especially in low- and middle-income countries (LMICs), where disparities in cancer care are particularly pronounced. This study provides a comprehensive overview of how AI is reshaping cancer care, reviews its benefits and challenges, and outlines ethical and policy implications in line with ASCO's 2025 theme, Driving Knowledge to Action. We offer concrete calls to action for clinicians, researchers, industry stakeholders, and policymakers to ensure that AI-driven, patient-centric oncology is accessible, equitable, and sustainable worldwide.
Collapse
Affiliation(s)
- Arturo Loaiza-Bonilla
- St Luke's University Health Network, Bethlehem, PA
- Massive Bio, Inc, New York, NY
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | - Caroline Chung
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Shawn Stapleton
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
5
|
Pignolo RJ, Connell JJ, Briggs W, Kelly CJ, Tromans C, Sultana N, Brady JM. Opportunistic assessment of osteoporosis using hip and pelvic X-rays with OsteoSight™: validation of an AI-based tool in a US population. Osteoporos Int 2025; 36:1053-1060. [PMID: 40263144 PMCID: PMC12122585 DOI: 10.1007/s00198-025-07487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Identifying patients at risk of low bone mineral density (BMD) from X-rays presents an attractive approach to increase case finding. This paper showed the diagnostic accuracy, reproducibility, and robustness of a new technology: OsteoSight™. OsteoSight could increase diagnosis and preventive treatment rates for patients with low BMD. PURPOSE This study aimed to evaluate the diagnostic accuracy, reproducibility, and robustness of OsteoSight™, an automated image analysis tool designed to identify low bone mineral density (BMD) from routine hip and pelvic X-rays. Given the global rise in osteoporosis-related fractures and the limitations of current diagnostic paradigms, OsteoSight offers a scalable solution that integrates into existing clinical workflows. METHODS Performance of the technology was tested across three key areas: (1) diagnostic accuracy in identifying low BMD as compared to dual-energy X-ray absorptiometry (DXA), the clinical gold standard; (2) reproducibility, through analysis of two images from the same patient; and (3) robustness, by evaluating the tool's performance across different patient demographics and X-ray scanner hardware. RESULTS The diagnostic accuracy of OsteoSight for identifying patients at risk of low BMD was area under the receiver operating characteristic curve (AUROC) 0.834 [0.789-0.880], with consistent results across subgroups of clinical confounders and X-ray scanner hardware. Specificity 0.852 [0.783-0.930] and sensitivity 0.628 [0.538-0.743] met pre-specified acceptance criteria. The pre-processing pipeline successfully excluded unsuitable cases including incorrect body parts, metalwork, and unacceptable femur positioning. CONCLUSION The results demonstrate that OsteoSight is accurate in identifying patients with low BMD. This suggests its utility as an opportunistic assessment tool, especially in settings where DXA accessibility is limited or not recently performed. The tool's reproducibility and robust performance across various clinical confounders further supports its integration into routine orthopedic and medical practices, potentially broadening the reach of osteoporosis assessment and enabling earlier intervention for at-risk patients.
Collapse
Affiliation(s)
| | | | - Will Briggs
- Naitive Technologies Ltd, London, EC1N 2SW, UK
| | | | | | | | | |
Collapse
|
6
|
Brancato B, Magni V, Saieva C, Risso GG, Buti F, Catarzi S, Ciuffi F, Peruzzi F, Regini F, Ambrogetti D, Alabiso G, Cruciani A, Doronzio V, Frati S, Giannetti GP, Guerra C, Valente P, Vignoli C, Atzori S, Carrera V, D'Agostino G, Fazzini G, Picano E, Turini FM, Vani V, Fantozzi F, Vietro DD, Cavallero D, Vietro FD, Plataroti D, Schiaffino S, Cozzi A. AI-supported approaches for mammography single and double reading: A controlled multireader study. Eur J Radiol 2025; 187:112101. [PMID: 40262459 DOI: 10.1016/j.ejrad.2025.112101] [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: 02/16/2025] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE To assess the impact of artificial intelligence (AI) on the diagnostic performance of radiologists with varying experience levels in mammography reading, considering single and simulated double reading approaches. METHODS In this retrospective study, 150 mammography examinations (30 with pathology-confirmed malignancies, 120 without malignancies [confirmed by 2-year follow-up]) were reviewed according to five approaches: A) human single reading by 26 radiologists of varying experience; B) AI single reading (Lunit INSIGHT MMG; C) human single reading with simultaneous AI support; D) simulated human-human double reading; E) simulated human-AI double reading, with AI as second independent reader flagging cases with a cancer probability ≥10 %. Sensitivity and specificity were calculated and compared using McNemar's test, univariate and multivariable logistic regression. RESULTS Compared to single reading without AI support, single reading with simultaneous AI support improved mean sensitivity from 69.2 % (standard deviation [SD] 15.6) to 84.5 % (SD 8.1, p < 0.001), providing comparable mean specificity (91.8 % versus 90.8 %, p = 0.06). The sensitivity increase provided by the AI-supported single reading was largest in the group of radiologists with a sensitivity below the median in the non-supported single reading, from 56.7 % (SD 12.1) to 79.7 % (SD 10.2, p < 0.001). In the simulated human-AI double reading approach, sensitivity further increased to 91.8 % (SD 3.4), surpassing that of the human-human simulated double reading (87.4 %, SD 8.8, p = 0.016), with comparable mean specificity (from 84.0 % to 83.0 %, p = 0.17). CONCLUSION AI support significantly enhanced sensitivity across all reading approaches, particularly benefiting worse performing radiologists. In the simulated double reading approaches, AI incorporation as independent second reader significantly increased sensitivity without compromising specificity.
Collapse
Affiliation(s)
- Beniamino Brancato
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Veronica Magni
- Postgraduate School in Radiodiagnostics, Università degli Studi di Milano, Milan, Italy.
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Gabriella Gemma Risso
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Francesca Buti
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Sandra Catarzi
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Fiorella Ciuffi
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Francesca Peruzzi
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Francesco Regini
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | - Daniela Ambrogetti
- Unit of Breast Imaging, Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Florence, Italy.
| | | | | | | | - Sara Frati
- Azienda USL Toscana Centro, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | - Vanina Vani
- Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.
| | | | - Dario De Vietro
- Postgraduate School in Radiodiagnostics, Università degli Studi di Firenze, Florence, Italy.
| | - Diletta Cavallero
- Postgraduate School in Radiodiagnostics, Università degli Studi di Pisa, Pisa, Italy.
| | - Fabrizio De Vietro
- Postgraduate School in Radiodiagnostics, Università degli Studi di Pisa, Pisa, Italy.
| | - Dario Plataroti
- Postgraduate School in Radiodiagnostics, Università degli Studi di Pisa, Pisa, Italy.
| | - Simone Schiaffino
- Imaging Institute of Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Andrea Cozzi
- Imaging Institute of Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland.
| |
Collapse
|
7
|
Taib AG, James JJ, Partridge GJW, Chen Y. Keeping AI on Track: Regular monitoring of algorithmic updates in mammography. Eur J Radiol 2025; 187:112100. [PMID: 40252280 DOI: 10.1016/j.ejrad.2025.112100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025]
Abstract
PURPOSE To demonstrate a method of benchmarking the performance of two consecutive software releases of the same commercial artificial intelligence (AI) product to trained human readers using the Personal Performance in Mammographic Screening scheme (PERFORMS) external quality assurance scheme. METHODS In this retrospective study, ten PERFORMS test sets, each consisting of 60 challenging cases, were evaluated by human readers between 2012 and 2023 and were evaluated by Version 1 (V1) and Version 2 (V2) of the same AI model in 2022 and 2023 respectively. Both AI and humans considered each breast independently. Both AI and humans considered the highest suspicion of malignancy score per breast for non-malignant cases and per lesion for breasts with malignancy. Sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) were calculated for comparison, with the study powered to detect a medium-sized effect (odds ratio, 3.5 or 0.29) for sensitivity. RESULTS The study included 1,254 human readers, with a total of 328 malignant lesions, 823 normal, and 55 benign breasts analysed. No significant difference was found between the AUCs for AI V1 (0.93) and V2 (0.94) (p = 0.13). In terms of sensitivity, no difference was observed between human readers and AI V1 (83.2 % vs 87.5 % respectively, p = 0.12), however V2 outperformed humans (88.7 %, p = 0.04). Specificity was higher for AI V1 (87.4 %) and V2 (88.2 %) compared to human readers (79.0 %, p < 0.01 respectively). CONCLUSION The upgraded AI model showed no significant difference in diagnostic performance compared to its predecessor when evaluating mammograms from PERFORMS test sets.
Collapse
Affiliation(s)
- Adnan G Taib
- Translational Medical Sciences, School of Medicine, University of Nottingham, Clinical Sciences Building, Nottingham City Hospital, Nottingham NG5 1PB, United Kingdom
| | - Jonathan J James
- Nottingham Breast Institute, Nottingham University Hospitals NHS Trust, Nottingham NG5 1PB England, United Kingdom
| | - George J W Partridge
- Translational Medical Sciences, School of Medicine, University of Nottingham, Clinical Sciences Building, Nottingham City Hospital, Nottingham NG5 1PB, United Kingdom
| | - Yan Chen
- Translational Medical Sciences, School of Medicine, University of Nottingham, Clinical Sciences Building, Nottingham City Hospital, Nottingham NG5 1PB, United Kingdom.
| |
Collapse
|
8
|
Peters J, van Leeuwen MM, Moriakov N, van Dijck JAAM, Mann RM, Teuwen J, Lips EH, van den Belt-Dusebout AW, Wesseling J, Penning de Vries BBL, Verboom S, Karssemeijer N, Elias SG, Broeders MJM. Development of radiomics-based models on mammograms with mass lesions to predict prognostically relevant characteristics of invasive breast cancer in a screening cohort. Br J Cancer 2025; 132:1040-1049. [PMID: 40188293 PMCID: PMC12120084 DOI: 10.1038/s41416-025-02995-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 01/17/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Optimizing breast-screening performance involves minimizing overdiagnosis of prognostically favorable invasive breast cancer (IBC) that does not need immediate recall and underdiagnosis of prognostically unfavorable IBC that is not recalled timely. We investigated whether mammographic features of masses predict prognostically relevant IBC characteristics. METHODS In a screening cohort, we obtained pathological information of 1587 IBCs presenting as a mass through the nationwide cancer registry and pathology databank. We developed models based on mammographic tumor appearance to predict whether IBC was prognostically favorable (T1N0M0 luminal A-like) or unfavorable. Models were based on 1095 positive screening mammograms (possible overdiagnosis), or on 603 last negative mammograms with in retrospect visible masses (possible underdiagnosis). We calculated performance metrics using cross-validation. RESULTS 23.5% of masses were prognostically favorable IBC. Using 1095 positive mammograms, the model's predictions to have prognostically favorable IBC (10th-90th percentile range 8.7-47.0%) yielded AUC 0.75 (SD across repeats 0.01), slope 1.16 (SD 0.07). Performance in 603 last negative screening mammograms with masses was poor: AUC 0.60 (SD 0.02), slope 0.85 (SD 0.28). CONCLUSIONS Mammography-based models from masses representing IBC at time of recall (possible overdiagnosis) predict prognostically relevant characteristics of IBC. Models based on in retrospect visible masses (possible underdiagnosis) performed poorly.
Collapse
Affiliation(s)
- Jim Peters
- Department IQ Health, Radboud University Medical Center, Nijmegen, Netherlands.
| | - Merle M van Leeuwen
- Division of Molecular Pathology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
| | - Nikita Moriakov
- Department of Radiation Oncology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
- Department of Radiology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
| | - Jos A A M van Dijck
- Department IQ Health, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ritse M Mann
- Department of Radiology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jonas Teuwen
- Department of Radiation Oncology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
| | | | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
- Department of Pathology, Netherlands Cancer Institute (NKI), Amsterdam, Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Bas B L Penning de Vries
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, University Utrecht, Utrecht, Netherlands
| | - Sarah Verboom
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nico Karssemeijer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sjoerd G Elias
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, University Utrecht, Utrecht, Netherlands
| | - Mireille J M Broeders
- Department IQ Health, Radboud University Medical Center, Nijmegen, Netherlands
- Dutch Expert Centre for Screening, Nijmegen, Netherlands
| |
Collapse
|
9
|
Tan H, Wu Q, Wu Y, Zheng B, Wang B, Chen Y, Du L, Zhou J, Fu F, Guo H, Fu C, Ma L, Dong P, Xue Z, Shen D, Wang M. Mammography-based artificial intelligence for breast cancer detection, diagnosis, and BI-RADS categorization using multi-view and multi-level convolutional neural networks. Insights Imaging 2025; 16:109. [PMID: 40397242 PMCID: PMC12095762 DOI: 10.1186/s13244-025-01983-x] [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: 03/08/2024] [Accepted: 04/27/2025] [Indexed: 05/22/2025] Open
Abstract
PURPOSE We developed an artificial intelligence system (AIS) using multi-view multi-level convolutional neural networks for breast cancer detection, diagnosis, and BI-RADS categorization support in mammography. METHODS Twenty-four thousand eight hundred sixty-six breasts from 12,433 Asian women between August 2012 and December 2018 were enrolled. The study consisted of three parts: (1) evaluation of AIS performance in malignancy diagnosis; (2) stratified analysis of BI-RADS 3-4 subgroups with AIS; and (3) reassessment of BI-RADS 0 breasts with AIS assistance. We further evaluate AIS by conducting a counterbalance-designed AI-assisted study, where ten radiologists read 1302 cases with/without AIS assistance. The area under the receiver operating characteristic curve (AUC), sensitivity, specificity, accuracy, and F1 score were measured. RESULTS The AIS yielded AUC values of 0.995, 0.933, and 0.947 for malignancy diagnosis in the validation set, testing set 1, and testing set 2, respectively. Within BI-RADS 3-4 subgroups with pathological results, AIS downgraded 83.1% of false-positives into benign groups, and upgraded 54.1% of false-negatives into malignant groups. AIS also successfully assisted radiologists in identifying 7 out of 43 malignancies initially diagnosed with BI-RADS 0, with a specificity of 96.7%. In the counterbalance-designed AI-assisted study, the average AUC across ten readers significantly improved with AIS assistance (p = 0.001). CONCLUSION AIS can accurately detect and diagnose breast cancer on mammography and further serve as a supportive tool for BI-RADS categorization. CRITICAL RELEVANCE STATEMENT An AI risk assessment tool employing deep learning algorithms was developed and validated for enhancing breast cancer diagnosis from mammograms, to improve risk stratification accuracy, particularly in patients with dense breasts, and serve as a decision support aid for radiologists. KEY POINTS The false positive and negative rates of mammography diagnosis remain high. The AIS can yield a high AUC for malignancy diagnosis. The AIS is important in stratifying BI-RADS categorization.
Collapse
Affiliation(s)
- Hongna Tan
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Qingxia Wu
- Beijing United Imaging Research Institute of Intelligent Imaging, Beijing, China
- United Imaging Intelligence (Beijing) Co. Ltd., Beijing, China
| | - Yaping Wu
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Bingjie Zheng
- Department of Radiology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University Zhengzhou, Henan, China
| | - Bo Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Chen
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijuan Du
- Department of Radiology, Zhengzhou Central Hospital, Zhengzhou, China
| | - Jing Zhou
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Fangfang Fu
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Huihui Guo
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Cong Fu
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China
| | - Lun Ma
- Department of Radiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Pei Dong
- Beijing United Imaging Research Institute of Intelligent Imaging, Beijing, China
- United Imaging Intelligence (Beijing) Co. Ltd., Beijing, China
| | - Zhong Xue
- Shanghai United Imaging Intelligence Co. Ltd, Shanghai, China
| | - Dinggang Shen
- Shanghai United Imaging Intelligence Co. Ltd, Shanghai, China
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, China.
- Imaging Diagnosis of Neurological Diseases and Research Laboratory of Henan Province, Zhengzhou, China.
| |
Collapse
|
10
|
Horton S, Wilson ML, Cheung ANY, DeStigter K, Kohli M, Sayed S, Schroeder LF, Sullivan R, Tan BS, Alooh M, Dahn B, Donoso-Bach L, Garcia PJ, Hussain S, Kao K, Looi LM, Pai M, Plebani M, Tebeje YK, Umutesi G, Walia K, Fleming KA. Moving the dial on diagnostics: an update from the Lancet Commission on diagnostics. Lancet 2025:S0140-6736(25)00804-9. [PMID: 40409332 DOI: 10.1016/s0140-6736(25)00804-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 05/25/2025]
Abstract
The Lancet Commission on diagnostics made recommendations for ten topics: national strategy (including national essential diagnostics lists), access in primary care, workforce, regulatory framework, national financing, affordability, appropriate use of technology, needs in conflict or fragile situations, advocacy, and an international alliance with oversight capabilities. Since 2021, progress in these areas has benefitted greatly from the adoption of a World Health Assembly resolution on diagnostics and the work of a broad coalition, as assessed by literature surveys by subject matter experts, quantitative findings (where feasible), and an anonymous survey of knowledgeable and engaged individuals. Greater progress was observed where there was political will and the production of diagnostics coincided with industrial policy goals, also in areas where changing the legal and health policy frameworks was involved. Progress was slower on recommendations with substantial resource implications (eg, labour force, affordability, and diagnostics for conflict situations). It is expected that the Global Diagnostics Coalition will consolidate and accelerate progress.
Collapse
Affiliation(s)
- Susan Horton
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada.
| | | | - Annie N Y Cheung
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | | | | - Lee F Schroeder
- University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Richard Sullivan
- Institute of Cancer Policy Centre for Conflict & Health Research, King's College London, London, UK
| | | | | | | | - Lluis Donoso-Bach
- Department of Medical Imaging, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Spain
| | - Patricia J Garcia
- School of Public Health, Universidad Peruana Cayetano Heredia, San Martín de Porres, Peru
| | - Sarwat Hussain
- University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Lai-Meng Looi
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Madhukar Pai
- School of Population and Global Health, McGill University, Montreal, QC, Canada
| | - Mario Plebani
- School of Medicine, University of Padova, Padova, Italy
| | - Yenew Kebede Tebeje
- Division of Laboratory Systems, Africa Centres for Disease Control and Prevention, Addis Ababa, Ethiopia
| | - Grace Umutesi
- Health and Life Sciences, Gates Ventures, Seattle, WA, USA
| | - Kamini Walia
- Indian Council of Medical Research, New Delhi, India
| | | |
Collapse
|
11
|
Bosma JS, Dercksen K, Builtjes L, André R, Roest C, Fransen SJ, Noordman CR, Navarro-Padilla M, Lefkes J, Alves N, de Grauw MJJ, van Eekelen L, Spronck JMA, Schuurmans M, de Wilde B, Hendrix W, Aswolinskiy W, Saha A, Twilt JJ, Geijs D, Veltman J, Yakar D, de Rooij M, Ciompi F, Hering A, Geerdink J, Huisman H. The DRAGON benchmark for clinical NLP. NPJ Digit Med 2025; 8:289. [PMID: 40379835 DOI: 10.1038/s41746-025-01626-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/09/2025] [Indexed: 05/19/2025] Open
Abstract
Artificial Intelligence can mitigate the global shortage of medical diagnostic personnel but requires large-scale annotated datasets to train clinical algorithms. Natural Language Processing (NLP), including Large Language Models (LLMs), shows great potential for annotating clinical data to facilitate algorithm development but remains underexplored due to a lack of public benchmarks. This study introduces the DRAGON challenge, a benchmark for clinical NLP with 28 tasks and 28,824 annotated medical reports from five Dutch care centers. It facilitates automated, large-scale, cost-effective data annotation. Foundational LLMs were pretrained using four million clinical reports from a sixth Dutch care center. Evaluations showed the superiority of domain-specific pretraining (DRAGON 2025 test score of 0.770) and mixed-domain pretraining (0.756), compared to general-domain pretraining (0.734, p < 0.005). While strong performance was achieved on 18/28 tasks, performance was subpar on 10/28 tasks, uncovering where innovations are needed. Benchmark, code, and foundational LLMs are publicly available.
Collapse
Affiliation(s)
- Joeran S Bosma
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Health & Information Technology, Ziekenhuisgroep Twente, Almelo, The Netherlands.
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Koen Dercksen
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Luc Builtjes
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Romain André
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Roest
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Stefan J Fransen
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Constant R Noordman
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Minimally Invasive Image-Guided Intervention Center, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mar Navarro-Padilla
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Judith Lefkes
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Natália Alves
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Max J J de Grauw
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leander van Eekelen
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joey M A Spronck
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Megan Schuurmans
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bram de Wilde
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ward Hendrix
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Witali Aswolinskiy
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anindo Saha
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
- Minimally Invasive Image-Guided Intervention Center, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jasper J Twilt
- Minimally Invasive Image-Guided Intervention Center, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daan Geijs
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen Veltman
- Department of Radiology, Ziekenhuisgroep Twente, Almelo, The Netherlands
| | - Derya Yakar
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten de Rooij
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Francesco Ciompi
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alessa Hering
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen Geerdink
- Department of Health & Information Technology, Ziekenhuisgroep Twente, Almelo, The Netherlands
| | - Henkjan Huisman
- Diagnostic Image Analysis Group, Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Mukherjee P, Beheshti A, Kumar SA, Wallace G, Merrett N, Clark J, Kos S, Rawstron E, Yang J, Grieve S, Shetty A, Singer S. Traffic Light Coding System for Engaging With AI in Surgery. ANZ J Surg 2025. [PMID: 40372391 DOI: 10.1111/ans.70172] [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: 02/13/2025] [Revised: 03/29/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Artificial Intelligence (AI) is generally defined as the development of computer systems or machines that can perform tasks typically requiring human intelligence and is increasingly being used in modern healthcare. While, various AI systems have existed for decades, its scale in healthcare has been escalated by global crises such as the COVID-19 pandemic and military conflicts, which has demanded rapid implementation of health system processes that improve efficiency in resource constrained environments. As AI-enabled technologies gain prominence, it is vital for surgeons to understand the various types of AI systems and their applications in medical practice.
Collapse
Affiliation(s)
- Payal Mukherjee
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
- Royal Prince Alfred Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, Australia
- Department of Head and Neck Surgery, Chris O'brien Lifehouse, Sydney, Australia
| | - Amin Beheshti
- Centre for Applied Artificial Intelligence, Macquarie University, Sydney, Australia
| | | | - Gordon Wallace
- Australian Institute for Innovative Materials, Intelligent Polymer Research Institute, Wollongong, Australia
| | - Neil Merrett
- Discipline of Surgery, School of Medicine, Western Sydney University, Sydney, Australia
| | - Jonathan Clark
- Royal Prince Alfred Institute of Academic Surgery, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, Australia
- Department of Head and Neck Surgery, Chris O'brien Lifehouse, Sydney, Australia
| | - Simon Kos
- Microsoft Ltd Australia, Auckland, New Zealand
| | - Ellen Rawstron
- Agency for Clinical Innovation, NSW Health, Sydney, Australia
| | - Jian Yang
- Centre for Applied Artificial Intelligence, Macquarie University, Sydney, Australia
| | - Stuart Grieve
- School of Health Sciences, University of Sydney, Sydney, Australia
| | - Amith Shetty
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney, Sydney, Australia
- System Sustainability and Performance, NSW Government, Sydney, Australia
| | - Simon Singer
- Australian Government Department of Health and Aged Care, Canberra, Australia
| |
Collapse
|
13
|
Oberije CJG, Currie R, Leaver A, Redman A, Teh W, Sharma N, Fox G, Glocker B, Khara G, Nash J, Ng AY, Kecskemethy PD. Assessing artificial intelligence in breast screening with stratified results on 306 839 mammograms across geographic regions, age, breast density and ethnicity: A Retrospective Investigation Evaluating Screening (ARIES) study. BMJ Health Care Inform 2025; 32:e101318. [PMID: 40374196 DOI: 10.1136/bmjhci-2024-101318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/27/2025] [Indexed: 05/17/2025] Open
Abstract
OBJECTIVES Evaluate an Artificial Intelligence (AI) system in breast screening through stratified results across age, breast density, ethnicity and screening centres, from different UK regions. METHODS A large-scale retrospective study evaluating two variations of using AI as an independent second reader in double reading was executed. Stratifications were conducted for clinical and operational metrics. Data from 306 839 mammography cases screened between 2017 and 2021 were used and included three different UK regions.The impact on safety and effectiveness was assessed using clinical metrics: cancer detection rate and positive predictive value, stratified according to age, breast density and ethnicity. Operational impact was assessed through reading workload and recall rate, measured overall and per centre.Non-inferiority was tested for AI workflows compared with human double reading, and when passed, superiority was tested. AI interval cancer (IC) flag rate was assessed to estimate additional cancer detection opportunity with AI that cannot be assessed retrospectively. RESULTS The AI workflows passed non-inferiority or superiority tests for every metric across all subgroups, with workload savings between 38.3% and 43.7%. The AI standalone flagged 41.2% of ICs overall, ranging between 33.3% and 46.8% across subgroups, with the highest detection rate for dense breasts. DISCUSSION Human double reading and AI workflows showed the same performance disparities across subgroups. The AI integrations maintained or improved performance at all metrics for all subgroups while achieving significant workload reduction. Moreover, complementing these integrations with AI as an additional reader can improve cancer detection. CONCLUSION The granularity of assessment showed that screening with the AI-system integrations was as safe as standard double reading across heterogeneous populations.
Collapse
Affiliation(s)
- Cary J G Oberije
- Kheiron Medical Technologies Limited, London, Greater London, UK
| | - Rachel Currie
- Breast Screening Programme, Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, UK
| | - Alice Leaver
- Breast Screening Unit, Gateshead Health NHS Foundation Trust, Gateshead, Gateshead, UK
| | - Alan Redman
- Breast Screening Unit, Gateshead Health NHS Foundation Trust, Gateshead, Gateshead, UK
| | - William Teh
- Breast Screening Programme, Royal Free London NHS Foundation Trust, London, UK
| | - Nisha Sharma
- Breast Screening Programme, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Georgia Fox
- Kheiron Medical Technologies Limited, London, Greater London, UK
| | - Ben Glocker
- Department of Computing, Imperial College London, London, UK
- DeepHealth, Somerville, MA, USA
| | - Galvin Khara
- Kheiron Medical Technologies Limited, London, Greater London, UK
| | - Jonathan Nash
- Kheiron Medical Technologies Limited, London, Greater London, UK
| | | | | |
Collapse
|
14
|
Schurz H, Solander K, Åström D, Cossío F, Choi T, Dustler M, Lundström C, Gustafsson H, Zackrisson S, Strand F. Simulating mismatch between calibration and target population in AI for mammography the retrospective VAIB study. NPJ Digit Med 2025; 8:259. [PMID: 40341801 PMCID: PMC12062211 DOI: 10.1038/s41746-025-01623-0] [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: 08/27/2024] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
AI cancer detection models require calibration to attain the desired balance between cancer detection rate (CDR) and false positive rate. In this study, we simulate the impact of six types of mismatches between the calibration population and the clinical target population, by creating purposefully non-representative datasets to calibrate AI for clinical settings. Mismatching the acquisition year between healthy and cancer-diagnosed screening participants led to a distortion in CDR between -3% to +19%. Mismatching age led to a distortion in CDR between -0.2% to +27%. Mismatching breast density distribution led to a distortion in CDR between +1% to 16%. Mismatching mammography vendors lead to a distortion in CDR between -32% to + 33%. Mismatches between calibration population and target clinical population lead to clinically important deviations. It is vital for safe clinical AI integration to ensure that important aspects of the calibration population are representative of the target population.
Collapse
Affiliation(s)
- Haiko Schurz
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden.
| | - Klara Solander
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | - Davida Åström
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | - Fernando Cossío
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
- Medical Diagnostics Karolinska, Karolinska University Hospital, Solna, Sweden
| | - Taeyang Choi
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
| | - Magnus Dustler
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Malmö, Sweden
| | - Claes Lundström
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Sectra AB, Linköping, Sweden
| | - Håkan Gustafsson
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
- Department of Medical Radiation Physics, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Sophia Zackrisson
- Department of Translational Medicine, Diagnostic Radiology, Lund University, Malmö, Sweden
- Department of Imaging and Physiology, Skåne University Hospital Malmö, Malmö, Sweden
| | - Fredrik Strand
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden.
- Medical Diagnostics Karolinska, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
15
|
Rosenqvist S, Dustler M, Brännmark J. Health Technologies and Impermissible Delays: The Case of Digital Breast Tomosynthesis. SCIENCE AND ENGINEERING ETHICS 2025; 31:13. [PMID: 40332720 PMCID: PMC12058816 DOI: 10.1007/s11948-025-00535-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/27/2025] [Indexed: 05/08/2025]
Abstract
This paper argues that we have a moral obligation to implement certain health technologies even if we have limited or incomplete evidence of their effectiveness. The focus is on technologies used in non-emergency settings, as opposed to "exceptional cases" such as compassionate use and emergency approvals during public health emergencies. A broadly plausible moral principle - the Ecumenical Principle - is introduced and applied to a test case: the use of Digital Breast Tomosynthesis in mammographic screening. The paper concludes by exploring the implications of the Ecumenical Principle for the adoption of other new health technologies.
Collapse
Affiliation(s)
- Simon Rosenqvist
- Department of Philosophy, Linguistics and Theory of Science, University of Gothenburg, Box 200, Göteborg, 405 30, Sweden.
| | - Magnus Dustler
- Diagnostic Radiology, Department of Translational Medicine, Lund University, Malmö, 205 02, Sweden
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Malmö, 205 02, Sweden
| | - Johan Brännmark
- Department of Philosophy, Stockholm University, Universitetsvägen 10D, Stockholm, 106 91, Sweden
| |
Collapse
|
16
|
Yu Y, Ren W, Mao L, Ouyang W, Hu Q, Yao Q, Tan Y, He Z, Ban X, Hu H, Lin R, Wang Z, Chen Y, Wu Z, Chen K, Ouyang J, Li T, Zhang Z, Liu G, Chen X, Li Z, Duan X, Wang J, Yao H. MRI-based multimodal AI model enables prediction of recurrence risk and adjuvant therapy in breast cancer. Pharmacol Res 2025; 216:107765. [PMID: 40345352 DOI: 10.1016/j.phrs.2025.107765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/06/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Timely intervention and improved prognosis for breast cancer patients rely on early metastasis risk detection and accurate treatment predictions. This study introduces an advanced multimodal MRI and AI-driven 3D deep learning model, termed the 3D-MMR-model, designed to predict recurrence risk in non-metastatic breast cancer patients. We conducted a multicenter study involving 1199 non-metastatic breast cancer patients from four institutions in China, with comprehensive MRI and clinical data retrospectively collected. Our model employed multimodal-data fusion, utilizing contrast-enhanced T1-weighted imaging (T1 + C) and T2-weighted imaging (T2WI) volumes, processed through a modified 3D-UNet for tumor segmentation and a DenseNet121-based architecture for disease-free survival (DFS) prediction. Additionally, we performed RNA-seq analysis to delve further into the relationship between concentrated hotspots within the tumor region and the tumor microenvironment. The 3D-MR-model demonstrated superior predictive performance, with time-dependent ROC analysis yielding AUC values of 0.90, 0.89, and 0.88 for 2-, 3-, and 4-year DFS predictions, respectively, in the training cohort. External validation cohorts corroborated these findings, highlighting the model's robustness across diverse clinical settings. Integration of clinicopathological features further enhanced the model's accuracy, with a multimodal approach significantly improving risk stratification and decision-making in clinical practice. Visualization techniques provided insights into the decision-making process, correlating predictions with tumor microenvironment characteristics. In summary, the 3D-MMR-model represents a significant advancement in breast cancer prognosis, combining cutting-edge AI technology with multimodal imaging to deliver precise and clinically relevant predictions of recurrence risk. This innovative approach holds promise for enhancing patient outcomes and guiding individualized treatment plans in breast cancer care.
Collapse
Affiliation(s)
- Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Joint Big Data Laboratory, Department of Medical Oncology, Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei, China; Institute for AI in Medicine and faculty of Medicine, Macau University of Science and Technology, Taipa, Macao, China; Department of Breast Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wei Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Luhui Mao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiugen Hu
- Department of Radiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Qinyue Yao
- Cells Vision (Guangzhou) Medical Technology Inc., Guangzhou, China
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zifan He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohua Ban
- Imaging Diagnostic and Interventional Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huijun Hu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruichong Lin
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China
| | - Zehua Wang
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China; UMedEVO and UMedREVO Artificial Intelligence Technology (Guangzhou) Co., Ltd
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Zhuo Wu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Ouyang
- Department of Breast Surgery, Tungwah Hospital, Dongguan, China
| | - Tang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zebang Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guoying Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuxing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuo Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jin Wang
- Cells Vision (Guangzhou) Medical Technology Inc., Guangzhou, China.
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
17
|
Sorin V, Sklair-Levy M, Glicksberg BS, Konen E, Nadkarni GN, Klang E. Deep Learning for Contrast Enhanced Mammography - A Systematic Review. Acad Radiol 2025; 32:2497-2508. [PMID: 39643464 DOI: 10.1016/j.acra.2024.11.035] [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: 08/14/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND/AIM Contrast-enhanced mammography (CEM) is a relatively novel imaging technique that enables both anatomical and functional breast imaging, with improved diagnostic performance compared to standard 2D mammography. The aim of this study is to systematically review the literature on deep learning (DL) applications for CEM, exploring how these models can further enhance CEM diagnostic potential. METHODS This systematic review was reported according to the PRISMA guidelines. We searched for studies published up to April 2024. MEDLINE, Scopus and Google Scholar were used as search databases. Two reviewers independently implemented the search strategy. We included all types of original studies published in English that evaluated DL algorithms for automatic analysis of contrast-enhanced mammography CEM images. The quality of the studies was independently evaluated by two reviewers based on the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) criteria. RESULTS Sixteen relevant studies published between 2018 and 2024 were identified. All but one used convolutional neural network models (CNN) models. All studies evaluated DL algorithms for lesion classification, while six studies also assessed lesion detection or segmentation. Segmentation was performed manually in three studies, both manually and automatically in two studies and automatically in ten studies. For lesion classification on retrospective datasets, CNN models reported varied areas under the curve (AUCs) ranging from 0.53 to 0.99. Models incorporating attention mechanism achieved accuracies of 88.1% and 89.1%. Prospective studies reported AUC values of 0.89 and 0.91. Some studies demonstrated that combining DL models with radiomics featured improved classification. Integrating DL algorithms with radiologists' assessments enhanced diagnostic performance. CONCLUSION While still at an early research stage, DL can improve CEM diagnostic precision. However, there is a relatively small number of studies evaluating different DL algorithms, and most studies are retrospective. Further prospective testing to assess performance of applications at actual clinical setting is warranted.
Collapse
Affiliation(s)
- Vera Sorin
- Department of Radiology, Mayo Clinic, Rochester, MN (V.S.).
| | - Miri Sklair-Levy
- Department of Diagnostic Imaging, Chaim Sheba Medical Center, affiliated to the Sackler School of Medicine, Tel-Aviv University, Israel (M.S-L., E.K.)
| | - Benjamin S Glicksberg
- Division of Data-Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY (B.S.G., G.N.N., E.K.)
| | - Eli Konen
- Department of Diagnostic Imaging, Chaim Sheba Medical Center, affiliated to the Sackler School of Medicine, Tel-Aviv University, Israel (M.S-L., E.K.)
| | - Girish N Nadkarni
- Division of Data-Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY (B.S.G., G.N.N., E.K.); The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY (G.N.N., E.K.)
| | - Eyal Klang
- Division of Data-Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY (B.S.G., G.N.N., E.K.); The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY (G.N.N., E.K.)
| |
Collapse
|
18
|
Graham-Knight JB, Liang P, Lin W, Wright Q, Shen H, Mar C, Sam J, Rajapakshe R. External Testing of a Commercial AI Algorithm for Breast Cancer Detection at Screening Mammography. Radiol Artif Intell 2025; 7:e240287. [PMID: 40072215 DOI: 10.1148/ryai.240287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Purpose To test a commercial artificial intelligence (AI) system for breast cancer detection at the BC Cancer Breast Screening Program. Materials and Methods In this retrospective study of 136 700 female individuals (mean age, 58.8 years ± 9.4 [SD]; median, 59.0 years; IQR = 14.0) who underwent digital mammography screening in British Columbia, Canada, between February 2019 and January 2020, breast cancer detection performance of a commercial AI algorithm was stratified by demographic, clinical, and imaging features and evaluated using the area under the receiver operating characteristic curve (AUC), and AI performance was compared with radiologists, using sensitivity and specificity. Results At 1-year follow-up, the AUC of the AI algorithm was 0.93 (95% CI: 0.92, 0.94) for breast cancer detection. Statistically significant differences were found for mammograms across radiologist-assigned Breast Imaging Reporting and Data System breast densities: category A, AUC of 0.96 (95% CI: 0.94, 0.99); category B, AUC of 0.94 (95% CI: 0.92, 0.95); category C, AUC of 0.93 (95% CI: 0.91, 0.95), and category D, AUC of 0.84 (95% CI: 0.76, 0.91) (AAUC > DAUC, P = .002; BAUC > DAUC, P = .009; CAUC > DAUC, P = .02). The AI showed higher performance for mammograms with architectural distortion (0.96 [95% CI: 0.94, 0.98]) versus without (0.92 [95% CI: 0.90, 0.93], P = .003) and lower performance for mammograms with calcification (0.87 [95% CI: 0.85, 0.90]) versus without (0.92 [95% CI: 0.91, 0.94], P < .001). Sensitivity of radiologists (92.6% ± 1.0) exceeded the AI algorithm (89.4% ± 1.1, P = .01), but there was no evidence of difference at 2-year follow-up (83.5% ± 1.2 vs 84.3% ± 1.2, P = .69). Conclusion The tested commercial AI algorithm is generalizable for a large external breast cancer screening cohort from Canada but showed different performance for some subgroups, including those with architectural distortion or calcification in the image. Keywords: Mammography, QA/QC, Screening, Technology Assessment, Screening Mammography, Artificial Intelligence, Breast Cancer, Model Testing, Bias and Fairness Supplemental material is available for this article. Published under a CC BY 4.0 license. See also commentary by Milch and Lee in this issue.
Collapse
Affiliation(s)
| | - Pengkun Liang
- Department of Medical Physics, BC Cancer-Kelowna, 399 Royal Ave, Kelowna, BC, Canada V1Y 5L3
| | - Wenna Lin
- Department of Medical Physics, BC Cancer-Kelowna, 399 Royal Ave, Kelowna, BC, Canada V1Y 5L3
| | - Quinn Wright
- Department of Medical Physics, BC Cancer-Kelowna, 399 Royal Ave, Kelowna, BC, Canada V1Y 5L3
| | - Hua Shen
- Department of Mathematics and Statistics, University of Calgary, Calgary, Canada
| | - Colin Mar
- BC Cancer Breast Screening Program, Vancouver, Canada
| | - Janette Sam
- BC Cancer Breast Screening Program, Vancouver, Canada
| | - Rasika Rajapakshe
- Department of Medical Physics, BC Cancer-Kelowna, 399 Royal Ave, Kelowna, BC, Canada V1Y 5L3
- BC Cancer Breast Screening Program, Vancouver, Canada
| |
Collapse
|
19
|
Friedewald SM, Sieniek M, Jansen S, Mahvar F, Kohlberger T, Schacht D, Bhole S, Gupta D, Prabhakara S, McKinney SM, Caron S, Melnick D, Etemadi M, Winter S, Saensuksopa T, Maciel A, Speroni L, Sevenich M, Agharwal A, Zhang R, Duggan G, Kadowaki S, Kiraly AP, Yang J, Mustafa B, Matias Y, Corrado GS, Tse D, Eswaran K, Shetty S. Triaging mammography with artificial intelligence: an implementation study. Breast Cancer Res Treat 2025; 211:1-10. [PMID: 39881074 PMCID: PMC11953103 DOI: 10.1007/s10549-025-07616-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
PURPOSE Many breast centers are unable to provide immediate results at the time of screening mammography which results in delayed patient care. Implementing artificial intelligence (AI) could identify patients who may have breast cancer and accelerate the time to diagnostic imaging and biopsy diagnosis. METHODS In this prospective randomized, unblinded, controlled implementation study we enrolled 1000 screening participants between March 2021 and May 2022. The experimental group used an AI system to prioritize a subset of cases for same-visit radiologist evaluation, and same-visit diagnostic workup if necessary. The control group followed the standard of care. The primary operational endpoints were time to additional imaging (TA) and time to biopsy diagnosis (TB). RESULTS The final cohort included 463 experimental and 392 control participants. The one-sided Mann-Whitney U test was employed for analysis of TA and TB. In the control group, the TA was 25.6 days [95% CI 22.0-29.9] and TB was 55.9 days [95% CI 45.5-69.6]. In comparison, the experimental group's mean TA was reduced by 25% (6.4 fewer days [one-sided 95% CI > 0.3], p<0.001) and mean TB was reduced by 30% (16.8 fewer days; 95% CI > 5.1], p=0.003). The time reduction was more pronounced for AI-prioritized participants in the experimental group. All participants eventually diagnosed with breast cancer were prioritized by the AI. CONCLUSIONS Implementing AI prioritization can accelerate care timelines for patients requiring additional workup, while maintaining the efficiency of delayed interpretation for most participants. Reducing diagnostic delays could contribute to improved patient adherence, decreased anxiety and addressing disparities in access to timely care.
Collapse
Affiliation(s)
- Sarah M Friedewald
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA.
- Lynn Sage Comprehensive Breast Center, Room 4-2304 250 E. Superior St., Chicago, IL, 60657, USA.
| | - Marcin Sieniek
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Sunny Jansen
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Fereshteh Mahvar
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Timo Kohlberger
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - David Schacht
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Sonya Bhole
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Dipti Gupta
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | | | | | - Stacey Caron
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - David Melnick
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Mozziyar Etemadi
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Samantha Winter
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | | | - Alejandra Maciel
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Luca Speroni
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Martha Sevenich
- Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL, 60611, USA
| | - Arnav Agharwal
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Rubin Zhang
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Gavin Duggan
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Shiro Kadowaki
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Atilla P Kiraly
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Jie Yang
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Basil Mustafa
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Yossi Matias
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Greg S Corrado
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Daniel Tse
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Krish Eswaran
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| | - Shravya Shetty
- Google Health, 1600 Amphitheatre Pkwy, Mountain View, CA, 94043, USA
| |
Collapse
|
20
|
Milch HS, Lee CI. Establishing the Evidence Needed for AI-driven Mammography Screening. Radiol Artif Intell 2025; 7:e250152. [PMID: 40172323 DOI: 10.1148/ryai.250152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Affiliation(s)
- Hannah S Milch
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, Calif
| | - Christoph I Lee
- Department of Radiology, University of Washington School of Medicine; Department of Health Systems & Population Health, University of Washington School of Public Health; Fred Hutchinson Cancer Center, 1144 Eastlake Ave E, LG-212, Seattle, WA 98109
| |
Collapse
|
21
|
Thomassin-Naggara I, Kilburn-Toppin F, Athanasiou A, Forrai G, Ispas M, Lesaru M, Giannotti E, Pinker-Domenig K, Van Ongeval C, Gilbert F, Mann RM, Pediconi F. Misdiagnosis in breast imaging: a statement paper from European Society Breast Imaging (EUSOBI)-Part 1: The role of common errors in radiology in missed breast cancer and implications of misdiagnosis. Eur Radiol 2025; 35:2387-2396. [PMID: 39545978 DOI: 10.1007/s00330-024-11128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 11/17/2024]
Abstract
IMPORTANCE Misdiagnosis in breast imaging can have significant implications for patients, healthcare providers, and the healthcare system as a whole. OBSERVATIONS Some of the potential implications of misdiagnosis in breast imaging include delayed diagnosis or false reassurance, which can result in a delay in treatment and potentially a worse prognosis. Misdiagnosis can also lead to unnecessary procedures, which can cause physical discomfort, anxiety, and emotional distress for patients, as well as increased healthcare costs. All these events can erode patient trust in the healthcare system and in individual healthcare providers. This can have negative implications for patient compliance with screening and treatment recommendations, as well as overall health outcomes. Moreover, misdiagnosis can also result in legal consequences for healthcare providers, including medical malpractice lawsuits and disciplinary action by licensing boards. CONCLUSION AND RELEVANCE To minimize the risk of misdiagnosis in breast imaging, it is important for healthcare providers to use appropriate imaging techniques and interpret images accurately and consistently. This requires ongoing training and education for radiologists and other healthcare providers, as well as collaboration and communication among healthcare providers to ensure that patients receive appropriate and timely care. If a misdiagnosis does occur, it is important for healthcare providers to communicate with patients and provide appropriate follow-up care to minimize the potential implications of the misdiagnosis. This may include repeat imaging, additional biopsies or other procedures, and referral to specialists for further evaluation and management. KEY POINTS Question What factors most contribute to and what implications stem from misdiagnosis in breast imaging? Findings Ongoing training and education for radiologists and other healthcare providers, as well as interdisciplinary collaboration and communication, is paramount. Clinical relevance Misdiagnosis in breast imaging can have significant implications for patients, healthcare providers, and the entire healthcare system.
Collapse
Affiliation(s)
- Isabelle Thomassin-Naggara
- Sorbonne Université, Paris, France.
- APHP Hopital Tenon, service d'Imageries Radiologiques et Interventionnelles Spécialisées (IRIS), Paris, France.
| | - Fleur Kilburn-Toppin
- Radiology Department, University of Cambridge, Hospital NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | | | - Gabor Forrai
- Duna Medical Center, GE-RAD Kft, Budapest, Hungary
| | - Miruna Ispas
- Department of Radiology, Imaging and Interventional Radiology Fundeni Clinical Institute, Bucharest, Romania
| | - Mihai Lesaru
- Department of Radiology, Imaging and Interventional Radiology Fundeni Clinical Institute, Bucharest, Romania
| | - Elisabetta Giannotti
- Radiology Department, University of Cambridge, Hospital NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Katja Pinker-Domenig
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna/Vienna General Hospital, Vienna, Austria
- Department of Breast Radiology, MSKCC, New York, NY, 10065, USA
| | - Chantal Van Ongeval
- Department of Radiology, Universitair Ziekenhuis Leuven, KU Leuven, Leuven, Belgium
| | - Fiona Gilbert
- Radiology Department, University of Cambridge, Hospital NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Ritse M Mann
- Department of Medical Imaging, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Amsterdam, The Netherlands
| | - Federica Pediconi
- Department of Radiological, Pathological and Oncological Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
22
|
Martiniussen MA, Larsen M, Hovda T, Kristiansen MU, Dahl FA, Eikvil L, Brautaset O, Bjørnerud A, Kristensen V, Bergan MB, Hofvind S. Performance of Two Deep Learning-based AI Models for Breast Cancer Detection and Localization on Screening Mammograms from BreastScreen Norway. Radiol Artif Intell 2025; 7:e240039. [PMID: 39907587 DOI: 10.1148/ryai.240039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Purpose To evaluate cancer detection and marker placement accuracy of two artificial intelligence (AI) models developed for interpretation of screening mammograms. Materials and Methods This retrospective study included data from 129 434 screening examinations (all female patients; mean age, 59.2 years ± 5.8 [SD]) performed between January 2008 and December 2018 in BreastScreen Norway. Model A was commercially available and model B was an in-house model. Area under the receiver operating characteristic curve (AUC) with 95% CIs were calculated. The study defined 3.2% and 11.1% of the examinations with the highest AI scores as positive, threshold 1 and 2, respectively. A radiologic review assessed location of AI markings and classified interval cancers as true or false negative. Results The AUC value was 0.93 (95% CI: 0.92, 0.94) for model A and B when including screen-detected and interval cancers. Model A identified 82.5% (611 of 741) of the screen-detected cancers at threshold 1 and 92.4% (685 of 741) at threshold 2. Model B identified 81.8% (606 of 741) at threshold 1 and 93.7% (694 of 741) at threshold 2. The AI markings were correctly localized for all screen-detected cancers identified by both models and 82% (56 of 68) of the interval cancers for model A and 79% (54 of 68) for model B. At the review, 21.6% (45 of 208) of the interval cancers were identified at the preceding screening by either or both models, correctly localized and classified as false negative (n = 17) or with minimal signs of malignancy (n = 28). Conclusion Both AI models showed promising performance for cancer detection on screening mammograms. The AI markings corresponded well to the true cancer locations. Keywords: Breast, Mammography, Screening, Computed-aided Diagnosis Supplemental material is available for this article. © RSNA, 2025 See also commentary by Cadrin-Chênevert in this issue.
Collapse
Affiliation(s)
- Marit A Martiniussen
- Department of Radiology, Østfold Hospital Trust, Kalnes, Norway
- University of Oslo, Institute of Clinical Medicine, Oslo, Norway
| | - Marthe Larsen
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, PO Box 5313, 0304, Oslo, Norway
| | - Tone Hovda
- Department of Radiology, Vestre Viken Hospital Trust, Drammen, Norway
| | | | | | | | | | - Atle Bjørnerud
- Computational Radiology & Artificial Intelligence (CRAI) Unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | | | - Marie B Bergan
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, PO Box 5313, 0304, Oslo, Norway
| | - Solveig Hofvind
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, PO Box 5313, 0304, Oslo, Norway
- Department of Health and Care Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
23
|
Cadrin-Chênevert A. Beyond Double Reading: Multiple Deep Learning Models Enhancing Radiologist-led Breast Screening. Radiol Artif Intell 2025; 7:e250125. [PMID: 40237597 DOI: 10.1148/ryai.250125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Affiliation(s)
- Alexandre Cadrin-Chênevert
- Department of Medical Imaging, CISSS Lanaudière, 1000 Blvd St-Anne, Saint-Charles-Borromée, QC, Canada J6E 6J2
| |
Collapse
|
24
|
Artificial intelligence improves breast cancer detection in mammography screening. Nat Med 2025; 31:1422-1423. [PMID: 40346277 DOI: 10.1038/s41591-025-03714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
|
25
|
Ciurescu S, Cerbu S, Dima CN, Borozan F, Pârvănescu R, Ilaș DG, Cîtu C, Vernic C, Sas I. AI in 2D Mammography: Improving Breast Cancer Screening Accuracy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:809. [PMID: 40428767 PMCID: PMC12113060 DOI: 10.3390/medicina61050809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Breast cancer is a leading global health challenge, where early detection is essential for improving survival outcomes. Two-dimensional (2D) mammography is the established standard for breast cancer screening; however, its diagnostic accuracy is limited by factors such as breast density and inter-reader variability. Recent advances in artificial intelligence (AI) have shown promise in enhancing radiological interpretation. This study aimed to assess the utility of AI in improving lesion detection and classification in 2D mammography. Materials and Methods: A retrospective analysis was performed on a dataset of 578 mammographic images obtained from a single radiology center. The dataset consisted of 36% pathologic and 64% normal cases, and was partitioned into training (403 images), validation (87 images), and test (88 images) sets. Image preprocessing involved grayscale conversion, contrast-limited adaptive histogram equalization (CLAHE), noise reduction, and sharpening. A convolutional neural network (CNN) model was developed using transfer learning with ResNet50. Model performance was evaluated using sensitivity, specificity, accuracy, and area under the receiver operating characteristic (AUC-ROC) curve. Results: The AI model achieved an overall classification accuracy of 88.5% and an AUC-ROC of 0.93, demonstrating strong discriminative capability between normal and pathologic cases. Notably, the model exhibited a high specificity of 92.7%, contributing to a reduction in false positives and improved screening efficiency. Conclusions: AI-assisted 2D mammography holds potential to enhance breast cancer detection by improving lesion classification and reducing false-positive findings. Although the model achieved high specificity, further optimization is required to minimize false negatives. Future efforts should aim to improve model sensitivity, incorporate multimodal imaging techniques, and validate results across larger, multicenter prospective cohorts to ensure effective integration into clinical radiology workflows.
Collapse
Affiliation(s)
- Sebastian Ciurescu
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (F.B.); (R.P.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (C.C.); (I.S.)
| | - Simona Cerbu
- Department XV of Orthopaedics, Traumatology, Urology and Medical Imaging, Discipline of Radiology and Medical Imaging, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Ciprian Nicușor Dima
- Division of Cardiovascular Surgery, Department VI Cardiology, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Florina Borozan
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (F.B.); (R.P.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (C.C.); (I.S.)
| | - Raluca Pârvănescu
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (F.B.); (R.P.)
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (C.C.); (I.S.)
| | - Diana-Gabriela Ilaș
- Department of Medical Semiology, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Cosmin Cîtu
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (C.C.); (I.S.)
| | - Corina Vernic
- Doctoral School in Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania; (S.C.); (F.B.); (R.P.)
- Department III—Functional Science, Discipline of Medical Informatics and Biostatistics, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Ioan Sas
- Department of Obstetrics and Gynecology, Victor Babeş University of Medicine and Pharmacy, 300041 Timișoara, Romania; (C.C.); (I.S.)
| |
Collapse
|
26
|
Xu X, Xi L, Zhu J, Feng C, Zhou P, Liu K, Shang Z, Shao Z. Intelligent Diagnosis of Cervical Lymph Node Metastasis Using a CNN Model. J Dent Res 2025:220345251322508. [PMID: 40271993 DOI: 10.1177/00220345251322508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
Lymph node (LN) metastasis is a prevalent cause of recurrence in oral squamous cell carcinoma (OSCC). However, accurately identifying metastatic LNs (LNs+) remains challenging. This prospective clinical study aims to test the effectiveness of our convolutional neural network (CNN) model for identifying OSCC cervical LN+ in contrast-enhanced computed tomography (CECT) in clinical practice. A CNN model was developed and trained using a dataset of 8,380 CECT images from previous OSCC patients. It was then prospectively validated on 17,777 preoperative CECT images from 354 OSCC patients between October 17, 2023, and August 31, 2024. The model's predicted LN results were provided to the surgical team without influencing surgical or treatment plans. During surgery, the predicted LN+ were identified and sent for separate pathological examination. The accuracy of the model's predictions was compared with those of human experts and verified against pathology reports. The capacity of the model to assist radiologists in LN+ diagnosis was also assessed. The CNN model was trained over 40 epochs and successfully validated after each. Compared with human experts (2 radiologists, 2 surgeons, and 2 students), the CNN model achieved higher sensitivity (81.89% vs. 81.48%, 46.91%, 50.62%), specificity (99.31% vs. 99.15%, 98.36%, 96.27%), LN+ accuracy (76.19% vs. 75.43%, P = 0.854; 40.64%, P < 0.001; 37.44%, P < 0.001), and clinical accuracy (86.16% vs. 83%, 61%, 56%). With the model's assistance, the radiologists surpassed both the previous predictive results without the model's support and the model's performance alone. The CNN model demonstrated an accuracy comparable to that of radiologists in identifying, locating, and predicting cervical LN+ in OSCC patients. Furthermore, the model has the potential to assist radiologists in making more accurate diagnoses.
Collapse
Affiliation(s)
- X Xu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Day Surgery Center, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L Xi
- School of Computer Science, Wuhan University, Wuhan, China
| | - J Zhu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Geriatric Dentistry, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - C Feng
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - P Zhou
- Department of Radiology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - K Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Head Neck Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Z Shang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Z Shao
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Day Surgery Center, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
27
|
van Nederpelt DR, Mendelsohn ZC, Bos L, Mattiesing RM, Ciccarelli O, Sastre-Garriga J, Carrasco FP, Kuijer JPA, Vrenken H, Killestein J, Schoonheim MM, Moraal B, Yousry T, Pontillo G, Rovira À, Strijbis EMM, Jasperse B, Barkhof F. User requirements for quantitative radiological reports in multiple sclerosis. Eur Radiol 2025:10.1007/s00330-025-11544-x. [PMID: 40240557 DOI: 10.1007/s00330-025-11544-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/30/2025] [Accepted: 02/20/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES Quantitative radiological reports (QReports) can enhance clinical management of multiple sclerosis (MS) by including quantitative data from MRI scans. However, the lack of consensus on the specific information to include, on and clinicians' preferences, hinders the adoption of these imaging analysis tools. This study aims to facilitate the clinical implementation of QReports by determining clinicians' requirements regarding their use in MS management. MATERIALS AND METHODS A four-phase Delphi panel approach was employed, involving neurologists and (neuro)radiologists across Europe. Initial interviews with experts helped develop a questionnaire addressing various QReport aspects. This questionnaire underwent refinement based on feedback and was distributed through the MAGNIMS network. A second questionnaire, incorporating additional questions, was circulated following a plenary discussion at the MAGNIMS workshop in Milan in November 2023. Responses from both questionnaire iterations were collected and analyzed, with adjustments made based on participant feedback. RESULTS The study achieved a 49.6% response rate, involving 78 respondents. Key preferences and barriers to QReport adoption were identified, highlighting the importance of integration into clinical workflows, cost-effectiveness, educational support for interpretation, and validation standards. Strong consensus emerged on including detailed lesion information and specific brain and spinal cord volume measurements. Concerns regarding report generation time, data protection, and reliability were also raised. CONCLUSION While QReports show potential for improving MS management, incorporation of the key metrics and addressing the identified barriers related to cost, validation, integration, and clinician education is crucial for practical implementation. These recommendations for developers to refine QReports could enhance their utility and adoption in clinical practice. KEY POINTS Question A lack of consensus on essential features for quantitative magnetic resonance imaging reports limits their integration into multiple sclerosis management. Findings This study identified key preferences, including detailed lesion information, specific brain and spinal cord measurements, and rigorous validation for effective quantitative reports. Clinical relevance This study identified essential features and barriers for implementing quantitative radiological reports in multiple sclerosis management, aiming to enhance clinical workflows, improve disease monitoring, and ultimately provide better, data-driven care for patients through tailored imaging solutions.
Collapse
Affiliation(s)
- David R van Nederpelt
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
| | - Zoe C Mendelsohn
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Radiology, Charité School of Medicine and University Hospital Berlin, Berlin, Germany
| | - Lonneke Bos
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Rozemarijn M Mattiesing
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Olga Ciccarelli
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Science, University College of London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | - Jaume Sastre-Garriga
- Department of Neurology, Multiple Sclerosis Centre of Catalonia, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ferran Prados Carrasco
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- e-Health Center, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Joost P A Kuijer
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Hugo Vrenken
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Joep Killestein
- MS Center Amsterdam, Neurology, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- MS Center Amsterdam, Anatomy and Neuroscience, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Bastiaan Moraal
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Tarek Yousry
- Lysholm Department of Neuroradiology and the Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, University College London Hospitals NHS Foundation Trust National Hospital for Neurology and Neurosurgery, London, UK
| | - Giuseppe Pontillo
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Science, University College of London, London, UK
- Departments of Advanced Biomedical Sciences and Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Àlex Rovira
- Section of Neuroradiology, Department of Radiology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eva M M Strijbis
- Department of Neurology, Multiple Sclerosis Centre of Catalonia, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Bas Jasperse
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frederik Barkhof
- MS Center Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Science, University College of London, London, UK
| |
Collapse
|
28
|
Kondo H, Ikawa F, Hara T, Kuwabara M, Ishii D, Tomimoto H, Horie N. Questionnaire Survey on the Current Use of Brain Docks and Their Compliance with Guidelines in Japan. Neurol Med Chir (Tokyo) 2025; 65:203-210. [PMID: 40128999 PMCID: PMC12061558 DOI: 10.2176/jns-nmc.2024-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025] Open
Abstract
Brain dock is used for the early diagnosis of intracranial lesions, prevention of cerebrovascular disorders, and early detection of cognitive decline. However, its application varies per facility. This study evaluated the use of brain dock and compliance with its guidelines via a questionnaire survey on the members of the Japan Society of Ningen Dock and Preventive Medical Care and the Japan Brain Dock Society. The questionnaire included information on the respondents, facility characteristics, and brain dock implementation. The number of responses was 288 (response rate: 10.3%). Brain dock was predominantly used in combination with other diagnostic methods. In addition to magnetic resonance imaging, the other examinations performed included the assessment of stroke risk factors and dementia. Radiographic image interpretation was frequently performed by more than one person, often by a neurosurgeon or radiologist. Artificial intelligence was used less frequently. In several facilities, the results were explained to all patients in person and to those who requested the findings in other facilities. Meanwhile, 10% of centers sent the results to the patients. Neurosurgeons were the most common professionals who provided explanations to the patients, followed by outpatient physicians who used the interpretation result as a reference. Only 24% of professionals were aware of the brain dock certification program. By solving the related problems, brain docks can play a greater role in improving medical issues in Japan, where the aging society is projected to increase.
Collapse
Affiliation(s)
- Hiroshi Kondo
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Fusao Ikawa
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
- Department of Neurosurgery, Shimane Prefectural Central Hospital
| | - Takeshi Hara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Masashi Kuwabara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Daizo Ishii
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine Faculty of Medicine
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
29
|
Yates J, Van Allen EM. New horizons at the interface of artificial intelligence and translational cancer research. Cancer Cell 2025; 43:708-727. [PMID: 40233719 PMCID: PMC12007700 DOI: 10.1016/j.ccell.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland; ETH AI Center, ETH Zurich, Zurich, Switzerland; Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard University, Boston, MA, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
30
|
Ha SM, Lee JM, Jang MJ, Kim HK, Chang JM. Breast Cancer Detection with Standalone AI versus Radiologist Interpretation of Unilateral Surveillance Mammography after Mastectomy. Radiology 2025; 315:e242955. [PMID: 40197097 DOI: 10.1148/radiol.242955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Limited data are available regarding the accuracy of artificial intelligence (AI) algorithms trained on bilateral mammograms for second breast cancer surveillance in patients with a personal history of breast cancer treated with unilateral mastectomy. Purpose To compare the performance of standalone AI for second breast cancer surveillance on unilateral mammograms with that of radiologists reading mammograms without AI assistance. Materials and Methods In this retrospective institutional database study, patients who were diagnosed with breast cancer between January 2001 and December 2018 and underwent postmastectomy surveillance mammography from January 2011 to March 2023 were included. Radiologists' mammogram interpretations without AI assistance were collected from these records and compared with AI interpretations of the same mammograms. The reference standards were histologic examination and 1-year follow-up data. The cancer detection rate per 1000 screening examinations, sensitivity, and specificity of standalone AI and the radiologists' interpretations without AI were compared using the McNemar test. Results Among the 4184 asymptomatic female patients (mean age, 52 years), 111 (2.7%) had contralateral second breast cancer. The cancer detection rate (17.4 per 1000 examinations [73 of 4184]; 95% CI: 13.7, 21.9) and sensitivity (65.8% [73 of 111]; 95% CI: 56.2, 74.5) were greater for standalone AI than for radiologists (14.6 per 1000 examinations [61 of 4184]; 95% CI: 11.2, 18.7; P = .01; 55.0% [61 of 111]; 95% CI: 45.2, 64.4; P = .01). The specificity was lower for standalone AI than for radiologists (91.5% [3725 of 4073]; 95% CI: 90.6, 92.3 vs 98.1% [3996 of 4073]; 95% CI: 97.6, 98.5; P < .001). AI detected 16 of 50 (32%) cancers missed by radiologists; however, 34 of 111 (30.6%) breast cancers were missed by both radiologists and AI. Conclusion Standalone AI for surveillance mammography showed higher sensitivity with lower specificity for contralateral breast cancer detection in patients treated with unilateral mastectomy than radiologists without AI assistance. © RSNA, 2025 Supplemental material is available for this article. See also the editorial by Philpotts in this issue.
Collapse
Affiliation(s)
- Su Min Ha
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Janie M Lee
- Department of Radiology, University of Washington School of Medicine, Seattle, Wash
| | - Myoung-Jin Jang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hong-Kyu Kim
- Department of Surgery, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung Min Chang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Brodsky V, Ullah E, Bychkov A, Song AH, Walk EE, Louis P, Rasool G, Singh RS, Mahmood F, Bui MM, Parwani AV. Generative Artificial Intelligence in Anatomic Pathology. Arch Pathol Lab Med 2025; 149:298-318. [PMID: 39836377 DOI: 10.5858/arpa.2024-0215-ra] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
CONTEXT.— Generative artificial intelligence (AI) has emerged as a transformative force in various fields, including anatomic pathology, where it offers the potential to significantly enhance diagnostic accuracy, workflow efficiency, and research capabilities. OBJECTIVE.— To explore the applications, benefits, and challenges of generative AI in anatomic pathology, with a focus on its impact on diagnostic processes, workflow efficiency, education, and research. DATA SOURCES.— A comprehensive review of current literature and recent advancements in the application of generative AI within anatomic pathology, categorized into unimodal and multimodal applications, and evaluated for clinical utility, ethical considerations, and future potential. CONCLUSIONS.— Generative AI demonstrates significant promise in various domains of anatomic pathology, including diagnostic accuracy enhanced through AI-driven image analysis, virtual staining, and synthetic data generation; workflow efficiency, with potential for improvement by automating routine tasks, quality control, and reflex testing; education and research, facilitated by AI-generated educational content, synthetic histology images, and advanced data analysis methods; and clinical integration, with preliminary surveys indicating cautious optimism for nondiagnostic AI tasks and growing engagement in academic settings. Ethical and practical challenges require rigorous validation, prompt engineering, federated learning, and synthetic data generation to help ensure trustworthy, reliable, and unbiased AI applications. Generative AI can potentially revolutionize anatomic pathology, enhancing diagnostic accuracy, improving workflow efficiency, and advancing education and research. Successful integration into clinical practice will require continued interdisciplinary collaboration, careful validation, and adherence to ethical standards to ensure the benefits of AI are realized while maintaining the highest standards of patient care.
Collapse
Affiliation(s)
- Victor Brodsky
- From the Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri (Brodsky)
| | - Ehsan Ullah
- the Department of Surgery, Health New Zealand, Counties Manukau, New Zealand (Ullah)
| | - Andrey Bychkov
- the Department of Pathology, Kameda Medical Center, Kamogawa City, Chiba Prefecture, Japan (Bychkov)
- the Department of Pathology, Nagasaki University, Nagasaki, Japan (Bychkov)
| | - Andrew H Song
- the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Song, Mahmood)
| | - Eric E Walk
- Office of the Chief Medical Officer, PathAI, Boston, Massachusetts (Walk)
| | - Peter Louis
- the Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey (Louis)
| | - Ghulam Rasool
- the Department of Oncologic Sciences, Morsani College of Medicine and Department of Electrical Engineering, University of South Florida, Tampa (Rasool)
- the Department of Machine Learning, Moffitt Cancer Center and Research Institute, Tampa, Florida (Rasool)
- Department of Machine Learning, Neuro-Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida (Rasool)
| | - Rajendra S Singh
- Dermatopathology and Digital Pathology, Summit Health, Berkley Heights, New Jersey (Singh)
| | - Faisal Mahmood
- the Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Song, Mahmood)
| | - Marilyn M Bui
- Department of Machine Learning, Pathology, Moffitt Cancer Center and Research Institute, Tampa, Florida (Bui)
| | - Anil V Parwani
- the Department of Pathology, The Ohio State University, Columbus (Parwani)
| |
Collapse
|
32
|
Boge F, Mosig A. Causality and scientific explanation of artificial intelligence systems in biomedicine. Pflugers Arch 2025; 477:543-554. [PMID: 39470762 PMCID: PMC11958387 DOI: 10.1007/s00424-024-03033-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024]
Abstract
With rapid advances of deep neural networks over the past decade, artificial intelligence (AI) systems are now commonplace in many applications in biomedicine. These systems often achieve high predictive accuracy in clinical studies, and increasingly in clinical practice. Yet, despite their commonly high predictive accuracy, the trustworthiness of AI systems needs to be questioned when it comes to decision-making that affects the well-being of patients or the fairness towards patients or other stakeholders affected by AI-based decisions. To address this, the field of explainable artificial intelligence, or XAI for short, has emerged, seeking to provide means by which AI-based decisions can be explained to experts, users, or other stakeholders. While it is commonly claimed that explanations of artificial intelligence (AI) establish the trustworthiness of AI-based decisions, it remains unclear what traits of explanations cause them to foster trustworthiness. Building on historical cases of scientific explanation in medicine, we here propagate our perspective that, in order to foster trustworthiness, explanations in biomedical AI should meet the criteria of being scientific explanations. To further undermine our approach, we discuss its relation to the concepts of causality and randomized intervention. In our perspective, we combine aspects from the three disciplines of biomedicine, machine learning, and philosophy. From this interdisciplinary angle, we shed light on how the explanation and trustworthiness of artificial intelligence relate to the concepts of causality and robustness. To connect our perspective with AI research practice, we review recent cases of AI-based studies in pathology and, finally, provide guidelines on how to connect AI in biomedicine with scientific explanation.
Collapse
Affiliation(s)
- Florian Boge
- Institute for Philosophy and Political Science, Technical University Dortmund, Emil-Figge-Str. 50, 44227, Dortmund, Germany
| | - Axel Mosig
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum (RUB), Gesundheitscampus 4, 44801, Bochum, NRW, Germany.
- Center for Protein Diagnostics, Ruhr University Bochum, Gesundheitscampus 4, 44801, Bochum, Germany.
| |
Collapse
|
33
|
Eisenstein M. How AI is helping to boost cancer screening. Nature 2025; 640:S62-S64. [PMID: 40269287 DOI: 10.1038/d41586-025-01153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
|
34
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [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: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
35
|
Walger L, Bauer T, Kügler D, Schmitz MH, Schuch F, Arendt C, Baumgartner T, Birkenheier J, Borger V, Endler C, Grau F, Immanuel C, Kölle M, Kupczyk P, Lakghomi A, Mackert S, Neuhaus E, Nordsiek J, Odenthal AM, Dague KO, Ostermann L, Pukropski J, Racz A, von der Ropp K, Schmeel FC, Schrader F, Sitter A, Unruh-Pinheiro A, Voigt M, Vychopen M, von Wedel P, von Wrede R, Attenberger U, Vatter H, Philipsen A, Becker A, Reuter M, Hattingen E, Sander JW, Radbruch A, Surges R, Rüber T. A Quantitative Comparison Between Human and Artificial Intelligence in the Detection of Focal Cortical Dysplasia. Invest Radiol 2025; 60:253-259. [PMID: 39437019 DOI: 10.1097/rli.0000000000001125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Artificial intelligence (AI) is thought to improve lesion detection. However, a lack of knowledge about human performance prevents a comparative evaluation of AI and an accurate assessment of its impact on clinical decision-making. The objective of this work is to quantitatively evaluate the ability of humans to detect focal cortical dysplasia (FCD), compare it to state-of-the-art AI, and determine how it may aid diagnostics. MATERIALS AND METHODS We prospectively recorded the performance of readers in detecting FCDs using single points and 3-dimensional bounding boxes. We acquired predictions of 3 AI models for the same dataset and compared these to readers. Finally, we analyzed pairwise combinations of readers and models. RESULTS Twenty-eight readers, including 20 nonexpert and 5 expert physicians, reviewed 180 cases: 146 subjects with FCD (median age: 25, interquartile range: 18) and 34 healthy control subjects (median age: 43, interquartile range: 19). Nonexpert readers detected 47% (95% confidence interval [CI]: 46, 49) of FCDs, whereas experts detected 68% (95% CI: 65, 71). The 3 AI models detected 32%, 51%, and 72% of FCDs, respectively. The latter, however, also predicted more than 13 false-positive clusters per subject on average. Human performance was improved in the presence of a transmantle sign ( P < 0.001) and cortical thickening ( P < 0.001). In contrast, AI models were sensitive to abnormal gyration ( P < 0.01) or gray-white matter blurring ( P < 0.01). Compared with single experts, expert-expert pairs detected 13% (95% CI: 9, 18) more FCDs ( P < 0.001). All AI models increased expert detection rates by up to 19% (95% CI: 15, 24) ( P < 0.001). Nonexpert+AI pairs could still outperform single experts by up to 13% (95% CI: 10, 17). CONCLUSIONS This study pioneers the comparative evaluation of humans and AI for FCD lesion detection. It shows that AI and human predictions differ, especially for certain MRI features of FCD, and, thus, how AI may complement the diagnostic workup.
Collapse
Affiliation(s)
- Lennart Walger
- From the Department of Neuroradiology, University Hospital Bonn, Bonn, Germany (L.W., T. Bauer, M.H.S., F.G., A.L., F.C.S., A. Radbruch, T.R.); Department of Epileptology, University Hospital Bonn, Bonn, Germany (L.W., T. Bauer, M.H.S., F. Schuch, T. Baumgartner, K.O.D., L.O., J.P., A. Racz, K.v.d.R., A.U.-P., P.v.W., R.v.W., R.S., T.R.); German Center for Neurodegenerative Diseases, Bonn, Germany (D.K., M.R., A. Radbruch); Department of Neuroradiology, Goethe University Frankfurt, Frankfurt, Germany (C.A., E.N., E.H.); Department of Neurology, University Hospital Bonn, Bonn, Germany (J.B., J.N.); Department of Neurosurgery, University Hospital Bonn, Bonn, Germany (V.B., M. Vychopen, H.V.); Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany (C.E., C.I., P.K., A.L., A.-M.O., M. Voigt, U.A.); Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany (M.K., S.M., F. Schrader, A.S., A.P.); Chair of Economic & Social Policy, WHU-Otto Beisheim School of Management, Vallendar, Germany (P.v.W.); Department of Neuropathology, University Hospital Bonn, Bonn, Germany (A.B.); A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA (M.R.); Department of Radiology, Harvard Medical School, Boston, MA (M.R.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (J.W.S.); Chalfont Centre for Epilepsy, Chalfont St Peter, United Kingdom (J.W.S.); Stichting Epilepsie Instellingen Nederland, Heemstede, the Netherland (J.W.S.); Department of Neurology, West China Hospital, Sichuan University, Chengdu, China (J.W.S.); and Center for Medical Data Usability and Translation, University of Bonn, Bonn, Germany (A. Radbruch, T.R.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hovda T, Larsen M, Bergan MB, Gjesvik J, Akslen LA, Hofvind S. Retrospective evaluation of a CE-marked AI system, including 1,017,208 mammography screening examinations. Eur Radiol 2025:10.1007/s00330-025-11521-4. [PMID: 40140078 DOI: 10.1007/s00330-025-11521-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVES To retrospectively evaluate the performance of a CE-marked AI system for identifying breast cancer on screening mammograms. Evidence from large retrospective studies is crucial for planning prospective studies and to further ensure safe implementation. MATERIALS AND METHODS We used data from screening examinations performed from 2004 to 2021 at ten breast centers in BreastScreen Norway. In the standard independent double reading setting, each radiologist scored each breast from 1 (negative) to 5 (high probability of cancer). The AI system assigned each examination an NT and an SN score; the NT score aimed to classify examinations as negative with minimal misclassification while the SN score aimed to classify examinations as positive with high confidence. N70 was defined as being among the 70% with the lowest NT score and P3 was defined as being among the 3% with the highest SN score. RESULTS A total of 1,017,208 screening examinations were included in the study sample. At N70, 1.8% (107/5977) of the screen-detected and 34.5% (625/1812) of the interval cancers were defined as negative. Using P3 to define cases as positive, 81.5% (4871/5977) of the screen-detected and 19.0% (344/1812) of the interval cancers were defined as positive. Among the screen-detected cancers in N70, 11.2% (12/107) had an interpretation score > 2 by both radiologists. CONCLUSION The AI system performed well according to identifying negative cases and cancer cases. Thus, the AI system can be used to reduce workload for the radiologists and potentially increase the sensitivity of mammography. KEY POINTS Question Results from large mammography screening samples not used in training AI algorithms are important to consider when planning prospective studies and implementation. Findings More than 80% of the screening-detected cancers were classified as positive by AI when considering 3% of the examinations with the highest AI risk score as positive. Clinical relevance A lack of radiologists is a challenge in mammographic screening. Our findings support other studies that suggest the use of AI to reduce screen-reading workload.
Collapse
Affiliation(s)
- Tone Hovda
- Department of Radiology, Vestre Viken Hospital Trust, Drammen, Norway
| | - Marthe Larsen
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Marie Burns Bergan
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Jonas Gjesvik
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Solveig Hofvind
- Section for Breast Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway.
- Department of Health and Care Sciences, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
37
|
Vasilev Y, Rumyantsev D, Vladzymyrskyy A, Omelyanskaya O, Pestrenin L, Shulkin I, Nikitin E, Kapninskiy A, Arzamasov K. Evolution of an Artificial Intelligence-Powered Application for Mammography. Diagnostics (Basel) 2025; 15:822. [PMID: 40218172 PMCID: PMC11988740 DOI: 10.3390/diagnostics15070822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background: The implementation of radiological artificial intelligence (AI) solutions remains challenging due to limitations in existing testing methodologies. This study assesses the efficacy of a comprehensive methodology for performance testing and monitoring of commercial-grade mammographic AI models. Methods: We utilized a combination of retrospective and prospective multicenter approaches to evaluate a neural network based on the Faster R-CNN architecture with a ResNet-50 backbone, trained on a dataset of 3641 mammograms. The methodology encompassed functional and calibration testing, coupled with routine technical and clinical monitoring. Feedback from testers and radiologists was relayed to the developers, who made updates to the AI model. The test dataset comprised 112 medical organizations, representing 10 manufacturers of mammography equipment and encompassing 593,365 studies. The evaluation metrics included the area under the curve (AUC), accuracy, sensitivity, specificity, technical defects, and clinical assessment scores. Results: The results demonstrated significant enhancement in the AI model's performance through collaborative efforts among developers, testers, and radiologists. Notable improvements included functionality, diagnostic accuracy, and technical stability. Specifically, the AUC rose by 24.7% (from 0.73 to 0.91), the accuracy improved by 15.6% (from 0.77 to 0.89), sensitivity grew by 37.1% (from 0.62 to 0.85), and specificity increased by 10.7% (from 0.84 to 0.93). The average proportion of technical defects declined from 9.0% to 1.0%, while the clinical assessment score improved from 63.4 to 72.0. Following 2 years and 9 months of testing, the AI solution was integrated into the compulsory health insurance system. Conclusions: The multi-stage, lifecycle-based testing methodology demonstrated substantial potential in software enhancement and integration into clinical practice. Key elements of this methodology include robust functional and diagnostic requirements, continuous testing and updates, systematic feedback collection from testers and radiologists, and prospective monitoring.
Collapse
Affiliation(s)
- Yuriy Vasilev
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
| | - Denis Rumyantsev
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
| | - Anton Vladzymyrskyy
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
- Department of Information Technology and Medical Data Processing, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Olga Omelyanskaya
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
| | - Lev Pestrenin
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
| | - Igor Shulkin
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
| | - Evgeniy Nikitin
- Celsus (Medical Screening Systems), Viktorenko St., Bldg. 11, Room 21N, 125167 Moscow, Russia; (E.N.); (A.K.)
| | - Artem Kapninskiy
- Celsus (Medical Screening Systems), Viktorenko St., Bldg. 11, Room 21N, 125167 Moscow, Russia; (E.N.); (A.K.)
| | - Kirill Arzamasov
- Research and Practical Clinical Center for Diagnostics and Telemedicine Technologies of the Moscow Health Care Department, 127051 Moscow, Russia; (Y.V.); (A.V.); (O.O.); (L.P.); (I.S.); (K.A.)
- Department of Artificial Intelligence Technologies, MIREA—Russian Technological University, 119454 Moscow, Russia
| |
Collapse
|
38
|
Wekenborg MK, Gilbert S, Kather JN. Examining human-AI interaction in real-world healthcare beyond the laboratory. NPJ Digit Med 2025; 8:169. [PMID: 40108434 PMCID: PMC11923224 DOI: 10.1038/s41746-025-01559-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Artificial Intelligence (AI) is revolutionizing healthcare, but its true impact depends on seamless human interaction. While most research focuses on technical metrics, we lack frameworks to measure the compatibility or synergy of real-world human-AI interactions in healthcare settings. We propose a multimodal toolkit combining ecological momentary assessment, quantitative observations, and baseline measurements to optimize AI implementation.
Collapse
Affiliation(s)
- Magdalena Katharina Wekenborg
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Stephen Gilbert
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
39
|
Beresford-Wylie B, Ashcroft S, Gaglani B, Green A, Smillie RW, Tucker K, Bakhai A, Esdaile B, Palamaras I. Integrating artificial intelligence into skin cancer pathways: the opportunities and obstacles. Br J Dermatol 2025; 192:738-739. [PMID: 39661653 DOI: 10.1093/bjd/ljae480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Artificial intelligence (AI) is a possible paradigm shift in our ability to provide dermatology services. Our article endeavours to outline the opportunities and obstacles faced by our specialty with integrating AI into our departments.
Collapse
Affiliation(s)
- Buket Beresford-Wylie
- The Dermatology Centre, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester, UK
| | | | | | - Alice Green
- Royal Free London NHS Foundation Trust, London, UK
| | | | - Katie Tucker
- Royal Free London NHS Foundation Trust, London, UK
| | - Ameet Bakhai
- Royal Free London NHS Foundation Trust, London, UK
| | | | | |
Collapse
|
40
|
Nishikawa RM, Sumkin A. Stop Training Artificial Intelligence Algorithms Now. Start Prospective Trials! JOURNAL OF BREAST IMAGING 2025; 7:165-167. [PMID: 39611808 DOI: 10.1093/jbi/wbae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Indexed: 11/30/2024]
Affiliation(s)
| | - Alisa Sumkin
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Ahsen ME, Ayvaci MUS, Mookerjee R, Stolovitzky G. Economics of AI and human task sharing for decision making in screening mammography. Nat Commun 2025; 16:2289. [PMID: 40055356 PMCID: PMC11889172 DOI: 10.1038/s41467-025-57409-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/12/2025] [Indexed: 05/13/2025] Open
Abstract
The rising global incidence of breast cancer and the persistent shortage of specialized radiologists have heightened the demand for innovative solutions in mammography screening. Artificial intelligence (AI) has emerged as a promising tool to bridge this demand-supply gap, with potential applications ranging from full automation to integrated AI-human decision-making. This study evaluates the economic feasibility of incorporating artificial intelligence (AI) into mammography screening within healthcare settings, considering full or partial integration. To evaluate the economic viability, we employ an optimization model specifically designed to minimize mammography screening costs. This model considers three distinct approaches when interpreting mammograms: automation strategy utilizing AI exclusively, delegation strategy involving the selective allocation of tasks between radiologists and AI, and the expert-alone strategy relying solely on radiologist decisions. Our findings underscore the significance of disease prevalence in relation to the trade-off between costs associated with false positives (e.g., follow-up expenses) and false negatives (e.g., litigation costs stemming from missed diagnoses) in shaping the AI strategy for healthcare organizations. We backtest our approach using data from an AI contest in which participants aimed to match or surpass radiologists' performance in assessing screening mammograms for women. The contest data supports the optimality of the delegation strategy, potentially leading to cost savings of 17.5% to 30.1% compared to relying solely on human experts. Our research provides guidance for healthcare organizations considering AI integration in mammography screening, with broader implications for work design and human-AI hybrid solutions in various fields.
Collapse
Affiliation(s)
- Mehmet Eren Ahsen
- Department of Business Administration, University of Illinois at Urbana-Champaign, Champaign, USA.
- Department of Biomedical and Translational Sciences, University of Illinois at Urbana-Champaign, Champaign, USA.
| | - Mehmet U S Ayvaci
- Jindal School of Management, University of Texas at Dallas, Richardson, USA
| | - Radha Mookerjee
- Jindal School of Management, University of Texas at Dallas, Richardson, USA
| | - Gustavo Stolovitzky
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
- Biomedical Data Sciences Hub, NYU Langone Health, New York, New York, USA
| |
Collapse
|
42
|
Chang YW, Ryu JK, An JK, Choi N, Park YM, Ko KH, Han K. Artificial intelligence for breast cancer screening in mammography (AI-STREAM): preliminary analysis of a prospective multicenter cohort study. Nat Commun 2025; 16:2248. [PMID: 40050619 PMCID: PMC11885569 DOI: 10.1038/s41467-025-57469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
Artificial intelligence (AI) improves the accuracy of mammography screening, but prospective evidence, particularly in a single-read setting, remains limited. This study compares the diagnostic accuracy of breast radiologists with and without AI-based computer-aided detection (AI-CAD) for screening mammograms in a real-world, single-read setting. A prospective multicenter cohort study is conducted within South Korea's national breast cancer screening program for women. The primary outcomes are screen-detected breast cancer within one year, with a focus on cancer detection rates (CDRs) and recall rates (RRs) of radiologists. A total of 24,543 women are included in the final cohort, with 140 (0.57%) screen-detected breast cancers. The CDR is significantly higher by 13.8% for breast radiologists using AI-CAD (n = 140 [5.70‰]) compared to those without AI (n = 123 [5.01‰]; p < 0.001), with no significant difference in RRs (p = 0.564). These preliminary results show a significant improvement in CDRs without affecting RRs in a radiologist's standard single-reading setting (ClinicalTrials.gov: NCT05024591).
Collapse
Affiliation(s)
- Yun-Woo Chang
- Department of Radiology, Soonchunhyang University Seoul Hospital, Seoul, Korea.
| | - Jung Kyu Ryu
- Department of Radiology, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Jin Kyung An
- Department of Radiology, Nowon Eulgi University Hospital, Seoul, Korea
| | - Nami Choi
- Department of Radiology, Konkuk University Medical center, Seoul, Korea
| | - Young Mi Park
- Department of Radiology, Inje University Busan Paik Hospital, Busan, Korea
| | - Kyung Hee Ko
- Department of Radiology, CHA Bundang Medical center, Seongnam, Korea
- Department of Radiology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Houssami N, Marinovich ML. AI for mammography: making double screen-reading history. Lancet Digit Health 2025; 7:e168-e169. [PMID: 39904653 DOI: 10.1016/j.landig.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025]
Affiliation(s)
- Nehmat Houssami
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Australia; School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia.
| | - M Luke Marinovich
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Australia; School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia
| |
Collapse
|
44
|
Behrens AS, Beckmann MW, Fasching PA, Huebner H, Emons J. [Personalized profiling in the field of senology]. RADIOLOGIE (HEIDELBERG, GERMANY) 2025; 65:194-200. [PMID: 39843711 DOI: 10.1007/s00117-024-01410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND The concept of personalized medicine is becoming increasingly important. The possibilities of diagnostics include not only genetic and molecular tumor profiles, but also the use of precise and individual imaging techniques. OBJECTIVES The development and implementation of suitable diagnostic procedures with high sensitivity and specificity, which are at the same time tailored to the individual risk factors and biological characteristics of the patient, remain a challenge. MATERIALS AND METHODS To enable personalized profiling, comprehensive diagnostics must be established that take into account all parameters such as imaging, molecular and genetic markers as well as real-world data and the use of artificial intelligence. This article sheds light on different approaches to personalized diagnostics in breast cancer and highlights the current clinical standard, innovative areas of research and the resulting challenges. CONCLUSION The highest hurdles for newer imaging techniques are the standardization of image analysis and the validation of these techniques in large clinical trials. The use of artificial intelligence requires not only appropriate technical and medical expertise, but also a sensitive approach to issues such as data protection and patient privacy. Real-world registries offer insights into real world treatment situations and are therefore of great importance.
Collapse
Affiliation(s)
- Annika S Behrens
- Universitätsklinikum Erlangen, Frauenklinik, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 21-23, 91054, Erlangen, Deutschland
| | - Matthias W Beckmann
- Universitätsklinikum Erlangen, Frauenklinik, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 21-23, 91054, Erlangen, Deutschland
| | - Peter A Fasching
- Universitätsklinikum Erlangen, Frauenklinik, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 21-23, 91054, Erlangen, Deutschland
| | - Hanna Huebner
- Universitätsklinikum Erlangen, Frauenklinik, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 21-23, 91054, Erlangen, Deutschland
| | - Julius Emons
- Universitätsklinikum Erlangen, Frauenklinik, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 21-23, 91054, Erlangen, Deutschland.
| |
Collapse
|
45
|
Dietzel M, Resch A, Baltzer PAT. [Artificial intelligence in breast imaging : Hopes and challenges]. RADIOLOGIE (HEIDELBERG, GERMANY) 2025; 65:187-193. [PMID: 39915299 PMCID: PMC11845416 DOI: 10.1007/s00117-024-01409-7] [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] [Accepted: 12/16/2024] [Indexed: 02/22/2025]
Abstract
CLINICAL/METHODICAL ISSUE Artificial intelligence (AI) is being increasingly integrated into clinical practice. However, the specific benefits are still unclear to many users. STANDARD RADIOLOGICAL METHODS In principle, AI applications are available for all imaging modalities, with a particular focus on mammography in breast diagnostics. METHODICAL INNOVATIONS AI promises to filter examinations into negative and clearly positive findings, and thereby reduces part of the radiological workload. Other applications are not yet as widely established. PERFORMANCE AI methods for mammography, and to a lesser extent tomosynthesis, have already reached the diagnostic quality of radiologists. ACHIEVEMENTS Except for second-opinion applications/triage in mammography, most methods are still under development. PRACTICAL RECOMMENDATIONS Currently, most AI applications must be critically evaluated by potential users regarding their maturity and practical benefits.
Collapse
Affiliation(s)
- Matthias Dietzel
- Department of Radiology, University Hospital Erlangen, Erlangen, Deutschland
| | - Alexandra Resch
- Department of Radiology, St. Francis Hospital Vienna, Sigmund Freud Private University Vienna, Vienna, Österreich
| | - Pascal A T Baltzer
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Waehringer-Guertel 18-20, 1090, Vienna, Österreich.
| |
Collapse
|
46
|
Shapiro J, Avitan-Hersh E, Greenfield B, Khamaysi Z, Dodiuk-Gad RP, Valdman-Grinshpoun Y, Freud T, Lyakhovitsky A. The use of a ChatGPT-4-based chatbot in teledermatology: A retrospective exploratory study. J Dtsch Dermatol Ges 2025; 23:311-319. [PMID: 39801186 DOI: 10.1111/ddg.15609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/10/2024] [Indexed: 03/08/2025]
Abstract
BACKGROUND AND OBJECTIVES Integration of artificial intelligence in healthcare, particularly ChatGPT, is transforming medical diagnostics and may benefit teledermatology. This exploratory study compared image description and differential diagnosis generation by a ChatGPT-4 based chatbot with human teledermatologists. PATIENTS AND METHODS This retrospective study compared 154 teledermatology consultations (December 2023-February 2024) with ChatGPT-4's performance in image descriptions and diagnoses. Diagnostic concordance was classified as "Top1" (exact match with the teledermatologist's diagnoses), "Top3" (correct diagnosis within one the top three diagnoses), and "Partial" (similar but not identical diagnoses). Image descriptions were rated and compared for quality parameters (location, color, size, morphology, and surrounding area), and accuracy (Yes, No, and Partial). RESULTS Out of 154 cases, ChatGPT-4 achieved a Top1 diagnostic concordance in 108 (70.8%), Top3 concordance in 137 (87.7%), partial concordance in four (2.6%), and was discordant in 15 (9.7%) cases. The quality of ChatGPT-4's image descriptions significantly surpassed teledermatologists in all five parameters. ChatGPT-4's descriptions were accurate in 130 (84.4%), partially accurate in 22 (14.3%), and inaccurate in two (1.3%) cases. CONCLUSIONS The preliminary findings of this study indicate that ChatGPT-4 demonstrates potential in generating accurate image descriptions and differential diagnoses. These results highlight the promise of integrating artificial intelligence into asynchronous teledermatology workflows.
Collapse
Affiliation(s)
| | - Emily Avitan-Hersh
- Rambam Health Care Campus, Haifa, Israel
- Dermatology Department, Bruce Rappaport Faculty of Medicine, Technion - Institute of Technology, Israel
| | | | - Ziad Khamaysi
- Rambam Health Care Campus, Haifa, Israel
- Dermatology Department, Bruce Rappaport Faculty of Medicine, Technion - Institute of Technology, Israel
| | - Roni P Dodiuk-Gad
- Dermatology Department, Bruce Rappaport Faculty of Medicine, Technion - Institute of Technology, Israel
- Emek Medical Center, Israel
- Department of Medicine, University of Toronto, Canada
| | | | - Tamar Freud
- Siaal Research Center for Family Medicine and Primary care, Department of Family Medicine, The Haim Doron Division of Community Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anna Lyakhovitsky
- Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Israel
| |
Collapse
|
47
|
Hernström V, Josefsson V, Sartor H, Schmidt D, Larsson AM, Hofvind S, Andersson I, Rosso A, Hagberg O, Lång K. Screening performance and characteristics of breast cancer detected in the Mammography Screening with Artificial Intelligence trial (MASAI): a randomised, controlled, parallel-group, non-inferiority, single-blinded, screening accuracy study. Lancet Digit Health 2025; 7:e175-e183. [PMID: 39904652 DOI: 10.1016/s2589-7500(24)00267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Emerging evidence suggests that artificial intelligence (AI) can increase cancer detection in mammography screening while reducing screen-reading workload, but further understanding of the clinical impact is needed. METHODS In this randomised, controlled, parallel-group, non-inferiority, single-blinded, screening-accuracy study, done within the Swedish national screening programme, women recruited at four screening sites in southwest Sweden (Malmö, Lund, Landskrona, and Trelleborg) who were eligible for mammography screening were randomly allocated (1:1) to AI-supported screening or standard double reading. The AI system (Transpara version 1.7.0 ScreenPoint Medical, Nijmegen, Netherlands) was used to triage screening examinations to single or double reading and as detection support highlighting suspicious findings. This is a protocol-defined analysis of the secondary outcome measures of recall, cancer detection, false-positive rates, positive predictive value of recall, type and stage of cancer detected, and screen-reading workload. This trial is registered at ClinicalTrials.gov, NCT04838756 and is closed to accrual. FINDINGS Between April 12, 2021, and Dec 7, 2022, 105 934 women were randomly assigned to the intervention or control group. 19 women were excluded from the analysis. The median age was 53·7 years (IQR 46·5-63·2). AI-supported screening among 53 043 participants resulted in 338 detected cancers and 1110 recalls. Standard screening among 52 872 participants resulted in 262 detected cancers and 1027 recalls. Cancer-detection rates were 6·4 per 1000 (95% CI 5·7-7·1) screened participants in the intervention group and 5·0 per 1000 (4·4-5·6) in the control group, a ratio of 1·29 (95% CI 1·09-1·51; p=0·0021). AI-supported screening resulted in an increased detection of invasive cancers (270 vs 217, a proportion ratio of 1·24 [95% CI 1·04-1·48]), wich were mainly small lymph-node negative cancers (58 more T1, 46 more lymph-node negative, and 21 more non-luminal A). AI-supported screening also resulted in an increased detection of in situ cancers (68 vs 45, a proportion ratio of 1·51 [1·03-2·19]), with about half of the increased detection being high-grade in situ cancer (12 more nuclear grade III, and no increase in nuclear grade I). The recall and false-positive rate were not significantly higher in the intervention group (a ratio of 1·08 [95% CI 0·99-1·17; p=0·084] and 1·01 [0·91-1·11; p=0·92], respectively). The positive predictive value of recall was significantly higher in the intervention group compared with the control group, with a ratio of 1·19 (95% CI 1·04-1·37; p=0·012). There were 61 248 screen readings in the intervention group and 109 692 in the control group, resulting in a 44·2% reduction in the screen-reading workload. INTERPRETATION The findings suggest that AI contributes to the early detection of clinically relevant breast cancer and reduces screen-reading workload without increasing false positives. FUNDING Swedish Cancer Society, Confederation of Regional Cancer Centres, and Swedish governmental funding for clinical research.
Collapse
Affiliation(s)
- Veronica Hernström
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Radiology Department, Skåne University Hospital, Malmö, Sweden
| | - Viktoria Josefsson
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Radiology Department, Skåne University Hospital, Malmö, Sweden
| | - Hanna Sartor
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Unilabs: Mammography Unit, Skåne University Hospital, Malmö, Sweden
| | - David Schmidt
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Unilabs: Mammography Unit, Skåne University Hospital, Malmö, Sweden
| | | | - Solveig Hofvind
- Section for Breast Cancer Screening, Cancer Registry of Norway, Oslo, Norway; Health and Care Sciences, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, Norway
| | - Ingvar Andersson
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Unilabs: Mammography Unit, Skåne University Hospital, Malmö, Sweden
| | - Aldana Rosso
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden
| | - Oskar Hagberg
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden
| | - Kristina Lång
- Diagnostic Radiology, Translational Medicine, Lund University, Lund, Sweden; Unilabs: Mammography Unit, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
48
|
Cui XW, Goudie A, Blaivas M, Chai YJ, Chammas MC, Dong Y, Stewart J, Jiang TA, Liang P, Sehgal CM, Wu XL, Hsieh PCC, Adrian S, Dietrich CF. WFUMB Commentary Paper on Artificial intelligence in Medical Ultrasound Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:428-438. [PMID: 39672681 DOI: 10.1016/j.ultrasmedbio.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 12/15/2024]
Abstract
Artificial intelligence (AI) is defined as the theory and development of computer systems able to perform tasks normally associated with human intelligence. At present, AI has been widely used in a variety of ultrasound tasks, including in point-of-care ultrasound, echocardiography, and various diseases of different organs. However, the characteristics of ultrasound, compared to other imaging modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI), poses significant additional challenges to AI. Application of AI can not only reduce variability during ultrasound image acquisition, but can standardize these interpretations and identify patterns that escape the human eye and brain. These advances have enabled greater innovations in ultrasound AI applications that can be applied to a variety of clinical settings and disease states. Therefore, The World Federation of Ultrasound in Medicine and Biology (WFUMB) is addressing the topic with a brief and practical overview of current and potential future AI applications in medical ultrasound, as well as discuss some current limitations and future challenges to AI implementation.
Collapse
Affiliation(s)
- Xin Wu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Adrian Goudie
- Department of Emergency, Fiona Stanley Hospital, Perth, Australia
| | - Michael Blaivas
- Department of Medicine, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Young Jun Chai
- Department of Surgery, Seoul National University College of Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Maria Cristina Chammas
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Yi Dong
- Department of Ultrasound, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jonathon Stewart
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Tian-An Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Chandra M Sehgal
- Ultrasound Research Lab, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Xing-Long Wu
- School of Computer Science & Engineering, Wuhan Institute of Technology, Wuhan, Hubei, China
| | | | - Saftoiu Adrian
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Christoph F Dietrich
- Department General Internal Medicine (DAIM), Hospitals Hirslanden Bern Beau Site, Salem and Permanence, Bern, Switzerland.
| |
Collapse
|
49
|
Eisemann N, Bunk S, Mukama T, Baltus H, Elsner SA, Gomille T, Hecht G, Heywang-Köbrunner S, Rathmann R, Siegmann-Luz K, Töllner T, Vomweg TW, Leibig C, Katalinic A. Nationwide real-world implementation of AI for cancer detection in population-based mammography screening. Nat Med 2025; 31:917-924. [PMID: 39775040 PMCID: PMC11922743 DOI: 10.1038/s41591-024-03408-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/11/2024] [Indexed: 01/11/2025]
Abstract
Artificial intelligence (AI) in mammography screening has shown promise in retrospective evaluations, but few prospective studies exist. PRAIM is an observational, multicenter, real-world, noninferiority, implementation study comparing the performance of AI-supported double reading to standard double reading (without AI) among women (50-69 years old) undergoing organized mammography screening at 12 sites in Germany. Radiologists in this study voluntarily chose whether to use the AI system. From July 2021 to February 2023, a total of 463,094 women were screened (260,739 with AI support) by 119 radiologists. Radiologists in the AI-supported screening group achieved a breast cancer detection rate of 6.7 per 1,000, which was 17.6% (95% confidence interval: +5.7%, +30.8%) higher than and statistically superior to the rate (5.7 per 1,000) achieved in the control group. The recall rate in the AI group was 37.4 per 1,000, which was lower than and noninferior to that (38.3 per 1,000) in the control group (percentage difference: -2.5% (-6.5%, +1.7%)). The positive predictive value (PPV) of recall was 17.9% in the AI group compared to 14.9% in the control group. The PPV of biopsy was 64.5% in the AI group versus 59.2% in the control group. Compared to standard double reading, AI-supported double reading was associated with a higher breast cancer detection rate without negatively affecting the recall rate, strongly indicating that AI can improve mammography screening metrics.
Collapse
Grants
- The study was funded by Vara. Vara was involved in study design, collection, interpretation of data, and in the writing of the report. All authors had access to all the data and were responsible for the decision to submit the manuscript. SB, TM, and CL are current employees of Vara with stock options as part of the standard compensation package. GH, RR, TG, TT, and TV actively participated in the study as radiologists and as customers of Vara. TT received speaker fees from Vara. AK received a payment from Vara for general consulting and speaker fees. KS-L received consulting fees from Hologic. SH-K has research cooperations with iCAD and ScreenPoint, no payments. NE, HB, and SE declare no conflict of interest.
- The study was funded by Vara.
Collapse
Affiliation(s)
- Nora Eisemann
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lubeck, Germany
| | | | | | - Hannah Baltus
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lubeck, Germany
| | - Susanne A Elsner
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lubeck, Germany
| | | | - Gerold Hecht
- Reference Center Mammography North, German Breast Cancer Screening Program, Oldenburg, Germany
| | - Sylvia Heywang-Köbrunner
- Reference Center Mammography Munich, German Breast Cancer Screening Program and FFB gGmbH, Munich, Germany
| | | | - Katja Siegmann-Luz
- Reference Center Mammography Berlin, German Breast Cancer Screening Program, Berlin, Germany
| | | | | | | | - Alexander Katalinic
- Institute for Social Medicine and Epidemiology, University of Lübeck, Lubeck, Germany.
| |
Collapse
|
50
|
Johnson LS, Zadrozniak P, Jasina G, Grotek-Cuprjak A, Andrade JG, Svennberg E, Diederichsen SZ, McIntyre WF, Stavrakis S, Benezet-Mazuecos J, Krisai P, Iakobishvili Z, Laish-Farkash A, Bhavnani S, Ljungström E, Bacevicius J, van Vreeswijk NL, Rienstra M, Spittler R, Marx JA, Oraii A, Miracle Blanco A, Lozano A, Mustafina I, Zafeiropoulos S, Bennett R, Bisson J, Linz D, Kogan Y, Glazer E, Marincheva G, Rahkovich M, Shaked E, Ruwald MH, Haugan K, Węcławski J, Radoslovich G, Jamal S, Brandes A, Matusik PT, Manninger M, Meyre PB, Blum S, Persson A, Måneheim A, Hammarlund P, Fedorowski A, Wodaje T, Lewinter C, Juknevicius V, Jakaite R, Shen C, Glotzer T, Platonov P, Engström G, Benz AP, Healey JS. Artificial intelligence for direct-to-physician reporting of ambulatory electrocardiography. Nat Med 2025; 31:925-931. [PMID: 39930139 PMCID: PMC11922735 DOI: 10.1038/s41591-025-03516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/16/2025] [Indexed: 03/21/2025]
Abstract
Developments in ambulatory electrocardiogram (ECG) technology have led to vast amounts of ECG data that currently need to be interpreted by human technicians. Here we tested an artificial intelligence (AI) algorithm for direct-to-physician reporting of ambulatory ECGs. Beat-by-beat annotation of 14,606 individual ambulatory ECG recordings (mean duration = 14 ± 10 days) was performed by certified ECG technicians (n = 167) and an ensemble AI model, called DeepRhythmAI. To compare the performance of the AI model and the technicians, a random sample of 5,235 rhythm events identified by the AI model or by technicians, of which 2,236 events were identified as critical arrhythmias, was selected for annotation by one of 17 cardiologist consensus panels. The mean sensitivity of the AI model for the identification of critical arrhythmias was 98.6% (95% confidence interval (CI) = 97.7-99.4), as compared to 80.3% (95% CI = 77.3-83.3%) for the technicians. False-negative findings were observed in 3.2/1,000 patients for the AI model versus 44.3/1,000 patients for the technicians. Accordingly, the relative risk of a missed diagnosis was 14.1 (95% CI = 10.4-19.0) times higher for the technicians. However, a higher false-positive event rate was observed for the AI model (12 (interquartile range (IQR) = 6-74)/1,000 patient days) as compared to the technicians (5 (IQR = 2-153)/1,000 patient days). We conclude that the DeepRhythmAI model has excellent negative predictive value for critical arrhythmias, substantially reducing false-negative findings, but at a modest cost of increased false-positive findings. AI-only analysis to facilitate direct-to-physician reporting could potentially reduce costs and improve access to care and outcomes in patients who need ambulatory ECG monitoring.
Collapse
Affiliation(s)
- L S Johnson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada.
| | | | - G Jasina
- Medicalgorithmics S.A., Warsaw, Poland
| | | | - J G Andrade
- Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - E Svennberg
- Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska University Hospital, Stockholm, Sweden
| | - S Z Diederichsen
- Department of Cardiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - W F McIntyre
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - S Stavrakis
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - P Krisai
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Z Iakobishvili
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
- Department of Cardiology, Clalit Health Services, Tel Aviv Jaffa District, Israel
| | - A Laish-Farkash
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - S Bhavnani
- Division of Cardiology, Scripps Clinic, San Diego, CA, USA
| | - E Ljungström
- Arrhythmia Clinic, Skåne University Hospital, Lund, Sweden
| | - J Bacevicius
- Clinic of Heart and Vessel Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - N L van Vreeswijk
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - M Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - R Spittler
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - J A Marx
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - A Oraii
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - A Miracle Blanco
- Cardiology Department Hospital Universitario La Luz, Madrid, Spain
| | - A Lozano
- Cardiology Department Hospital Universitario La Luz, Madrid, Spain
| | - I Mustafina
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - S Zafeiropoulos
- Feinstein Institutes for Medical Research at Northwell Health, Manhasset, NY, USA
- Department of Cardiology, University Hospital of Zurich, Zürich, Switzerland
| | - R Bennett
- Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - J Bisson
- Department of Cardiology, Centre hospitalier de l'Université de Montréal-Université de Montréal, Montréal, Quebec, Canada
| | - D Linz
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Y Kogan
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - E Glazer
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - G Marincheva
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - M Rahkovich
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - E Shaked
- Department of Cardiology, Assuta Ashdod University Hospital, Ben-Gurion University of the Negev, Ashdod, Israel
| | - M H Ruwald
- Department of Cardiology, Gentofte Hospital, Hellerup, Denmark
| | - K Haugan
- Department of Cardiology, Zealand University Hospital, Roskilde, Denmark
| | | | - G Radoslovich
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - S Jamal
- Hackensack University Medical Center, Hackensack, NJ, USA
- Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - A Brandes
- Department of Cardiology, Esbjerg Hospital-University Hospital of Southern Denmark, Esbjerg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark
| | - P T Matusik
- Department of Electrocardiology, Institute of Cardiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
- St. John Paul II Hospital, Kraków, Poland
| | - M Manninger
- Division of Cardiology, Department of Medicine, Medical University of Graz, Graz, Austria
| | - P B Meyre
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - S Blum
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - A Persson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Malmö, Sweden
| | - A Måneheim
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Clinical Physiology, Skåne University Hospital, Malmö, Sweden
| | - P Hammarlund
- Department of Cardiology, Helsingborg Hospital, Helsingborg, Sweden
| | - A Fedorowski
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - T Wodaje
- Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - C Lewinter
- Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- University of Glasgow, University of Glasgow, Institute of Wellbeing, Glasgow, UK
| | - V Juknevicius
- Clinic of Heart and Vessel Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - R Jakaite
- Clinic of Heart and Vessel Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - C Shen
- Division of Cardiology, Scripps Clinic, San Diego, CA, USA
| | - T Glotzer
- Hackensack University Medical Center, Hackensack, NJ, USA
- Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - P Platonov
- Arrhythmia Clinic, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - G Engström
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - A P Benz
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Cardiology, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - J S Healey
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|