1
|
Marret G, Herrera M, Siu LL. Turning the kaleidoscope: Innovations shaping the future of clinical trial design. Cancer Cell 2025; 43:597-605. [PMID: 40086439 DOI: 10.1016/j.ccell.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025]
Abstract
Current clinical trials are based on rigid designs and drug-centric approaches that can stifle flexibility and innovation. With advances in molecular biology and technology, there is an urgent call to revitalize trial designs to meet these evolving demands. We propose a reshaped, prismatic vision of clinical trials combining different knowledge layers, synergized with modern computational approaches. This paradigm based on iterative learning will enable a more adaptive and precise framework for oncology drug development.
Collapse
Affiliation(s)
- Grégoire Marret
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mercedes Herrera
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Li M, Xu P, Hu J, Tang Z, Yang G. From challenges and pitfalls to recommendations and opportunities: Implementing federated learning in healthcare. Med Image Anal 2025; 101:103497. [PMID: 39961211 DOI: 10.1016/j.media.2025.103497] [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: 09/15/2024] [Revised: 01/18/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025]
Abstract
Federated learning holds great potential for enabling large-scale healthcare research and collaboration across multiple centers while ensuring data privacy and security are not compromised. Although numerous recent studies suggest or utilize federated learning based methods in healthcare, it remains unclear which ones have potential clinical utility. This review paper considers and analyzes the most recent studies up to May 2024 that describe federated learning based methods in healthcare. After a thorough review, we find that the vast majority are not appropriate for clinical use due to their methodological flaws and/or underlying biases which include but are not limited to privacy concerns, generalization issues, and communication costs. As a result, the effectiveness of federated learning in healthcare is significantly compromised. To overcome these challenges, we provide recommendations and promising opportunities that might be implemented to resolve these problems and improve the quality of model development in federated learning with healthcare.
Collapse
Affiliation(s)
- Ming Li
- Bioengineering Department and Imperial-X, Imperial College London, London W12 7SL, UK; National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.
| | - Pengcheng Xu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA; State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China.
| | - Junjie Hu
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.
| | - Zeyu Tang
- Bioengineering Department and Imperial-X, Imperial College London, London W12 7SL, UK; Tri-Institutional Computational Biology & Medicine Program, Weill Cornell Medicine of Cornell University, NY, USA.
| | - Guang Yang
- Bioengineering Department and Imperial-X, Imperial College London, London W12 7SL, UK; National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK; Cardiovascular Research Centre, Royal Brompton Hospital, London SW3 6NP, UK; School of Biomedical Engineering & Imaging Sciences, King's College London, London WC2R 2LS, UK.
| |
Collapse
|
3
|
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
|
4
|
Gessain G, Lacroix-Triki M. Computational pathology for breast cancer: Where do we stand for prognostic applications? Breast 2025; 81:104464. [PMID: 40179582 DOI: 10.1016/j.breast.2025.104464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/24/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
The very early days of artificial intelligence (AI) in healthcare are behind us. AI is now spreading in the healthcare sector and is gradually being implemented in routine clinical practice. Driven by the increasing digitization of microscope slides, computational pathology (CPath) is further strengthening the role of AI in the field of oncology. CPath is transforming fundamental research as well as routine clinical practice, both for diagnostic and prognostic applications. In breast cancer, CPath holds the potential to address several unmet clinical needs, particularly in the areas of biomarkers and prognostic tools. Indeed, multiple applications are on their way, ranging from predicting clinically meaningful endpoints to offering alternatives to gene-expression testing and detecting molecular alterations directly from digitized whole slide images. However, to fully harness the potential of CPath, several challenges must be overcome. These include improving the availability of multimodal patient data, advancing the digitalization of pathology laboratories, increasing adoption within the medical community, and navigating regulatory hurdles. This review offers an overview of the current landscape of CPath in breast cancer, highlighting the progress made and the hurdles that remain for its widespread clinical adoption in prognostic applications.
Collapse
Affiliation(s)
- Grégoire Gessain
- Department of Pathology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, cedex, 94805, Villejuif, France; Université Paris-Cité, Faculté de Santé, Paris, France
| | - Magali Lacroix-Triki
- Department of Pathology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, cedex, 94805, Villejuif, France.
| |
Collapse
|
5
|
Tölle M, Garthe P, Scherer C, Seliger JM, Leha A, Krüger N, Simm S, Martin S, Eble S, Kelm H, Bednorz M, André F, Bannas P, Diller G, Frey N, Groß S, Hennemuth A, Kaderali L, Meyer A, Nagel E, Orwat S, Seiffert M, Friede T, Seidler T, Engelhardt S. Real world federated learning with a knowledge distilled transformer for cardiac CT imaging. NPJ Digit Med 2025; 8:88. [PMID: 39915633 PMCID: PMC11802793 DOI: 10.1038/s41746-025-01434-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Federated learning is a renowned technique for utilizing decentralized data while preserving privacy. However, real-world applications often face challenges like partially labeled datasets, where only a few locations have certain expert annotations, leaving large portions of unlabeled data unused. Leveraging these could enhance transformer architectures' ability in regimes with small and diversely annotated sets. We conduct the largest federated cardiac CT analysis to date (n = 8, 104) in a real-world setting across eight hospitals. Our two-step semi-supervised strategy distills knowledge from task-specific CNNs into a transformer. First, CNNs predict on unlabeled data per label type and then the transformer learns from these predictions with label-specific heads. This improves predictive accuracy and enables simultaneous learning of all partial labels across the federation, and outperforms UNet-based models in generalizability on downstream tasks. Code and model weights are made openly available for leveraging future cardiac CT analysis.
Collapse
Affiliation(s)
- Malte Tölle
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany.
- Heidelberg University, Heidelberg, Germany.
- Informatics for Life Institute, Heidelberg, Germany.
| | - Philipp Garthe
- Clinic for Cardiology III, University Hospital Münster, Münster, Germany
| | - Clemens Scherer
- DZHK (German Centre for Cardiovascular Research), partner site Munich, Munich, Germany
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jan Moritz Seliger
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Leha
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Göttingen, Germany
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Nina Krüger
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Stefan Simm
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Simon Martin
- DZHK (German Centre for Cardiovascular Research), partner site RhineMain, Frankfurt, Germany
- Institute for Experimental and Translational Cardiovascular Imaging, Goethe University, Frankfurt am Main, Germany
| | - Sebastian Eble
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
| | - Halvar Kelm
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
| | - Moritz Bednorz
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian André
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
- Informatics for Life Institute, Heidelberg, Germany
| | - Peter Bannas
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Diller
- Clinic for Cardiology III, University Hospital Münster, Münster, Germany
| | - Norbert Frey
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
- Informatics for Life Institute, Heidelberg, Germany
| | - Stefan Groß
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Anja Hennemuth
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Lars Kaderali
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Meyer
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
| | - Eike Nagel
- DZHK (German Centre for Cardiovascular Research), partner site RhineMain, Frankfurt, Germany
- Institute for Experimental and Translational Cardiovascular Imaging, Goethe University, Frankfurt am Main, Germany
| | - Stefan Orwat
- Clinic for Cardiology III, University Hospital Münster, Münster, Germany
| | - Moritz Seiffert
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Friede
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Göttingen, Germany
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Seidler
- DZHK (German Centre for Cardiovascular Research), partner site Lower Saxony, Göttingen, Germany
- Department of Cardiology, University Medicine Göttingen, Göttingen, Germany
- Department of Cardiology, Campus Kerckhoff of the Justus-Liebig-University at Gießen, Kerckhoff-Clinic, Gießen, Germany
| | - Sandy Engelhardt
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
- Informatics for Life Institute, Heidelberg, Germany
| |
Collapse
|
6
|
Gupta C, Gill NS, Gulia P, Alduaiji N, Shreyas J, Shukla PK. Applying YOLOv6 as an ensemble federated learning framework to classify breast cancer pathology images. Sci Rep 2025; 15:3769. [PMID: 39885198 PMCID: PMC11782635 DOI: 10.1038/s41598-024-80187-7] [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: 07/20/2024] [Accepted: 11/15/2024] [Indexed: 02/01/2025] Open
Abstract
The most common carcinoma-related cause of death among women is breast cancer. Early detection is crucial, and the manual screening method may lead to a delayed diagnosis, which would delay treatment and put lives at risk. Mammography imaging is advised for routine screening to diagnose breast cancer at an early stage. To improve generalizability, this study examines the implementation of Federated Learning (FedL) to detect breast cancer. Its performance is compared to a centralized training technique that diagnoses breast cancer. Although FedL has been famous as a safeguarding privacy algorithm, its similarities to ensemble learning methods, such as federated averaging (FEDAvrg), still need to be thoroughly investigated. This study examines explicitly how a YOLOv6 model trained with FedL performs across several clients. A new homomorphic encryption and decryption algorithm is also proposed to retain data privacy. A novel pruned YOLOv6 model with FedL is introduced in this study to differentiate benign and malignant tissues. The model is trained on the breast cancer pathological dataset BreakHis and BUSI. The proposed model achieved a validation accuracy of 98% on BreakHis dataset and 97% on BUSI dataset. The results are compared with the VGG-19, ResNet-50, and InceptionV3 algorithms, showing that the proposed model achieved better results. The tests reveal that federated learning is feasible, as FedAvrg trains models of outstanding quality with only a few communication rounds, as shown by the results on a range of model topologies such as ResNet50, VGG-19, InceptionV3, and the proposed Ensembled FedL YOLOv6.
Collapse
Affiliation(s)
- Chhaya Gupta
- Department of Computer Science and Applications, Maharshi Dayanand University, Rohtak, India
| | - Nasib Singh Gill
- Department of Computer Science and Applications, Maharshi Dayanand University, Rohtak, India
| | - Preeti Gulia
- Department of Computer Science and Applications, Maharshi Dayanand University, Rohtak, India
| | - Noha Alduaiji
- Department of Computer Science, College of Computer and Information Sciences, Majmaah University, 11952, Al Majmaah, Saudi Arabia
| | - J Shreyas
- Department of Information Technology, Manipal Institute of Technology Bengaluru, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Piyush Kumar Shukla
- Department of Computer Science and Engineering, University Institute of Technology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State Technological University of Madhya Pradesh), Madhya Pradesh, Bhopal, 462033, India
| |
Collapse
|
7
|
Li N, Lewin A, Ning S, Waito M, Zeller MP, Tinmouth A, Shih AW. Privacy-preserving federated data access and federated learning: Improved data sharing and AI model development in transfusion medicine. Transfusion 2025; 65:22-28. [PMID: 39610333 PMCID: PMC11747086 DOI: 10.1111/trf.18077] [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: 08/10/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Health data comprise data from different aspects of healthcare including administrative, digital health, and research-oriented data. Together, health data contribute to and inform healthcare operations, patient care, and research. Integrating artificial intelligence (AI) into healthcare requires understanding these data infrastructures and addressing challenges such as data availability, privacy, and governance. Federated learning (FL), a decentralized AI training approach, addresses these challenges by allowing models to learn from diverse datasets without data leaving its source, thus ensuring privacy and security are maintained. This report introduces FL and discusses its potential in transfusion medicine and blood supply chain management. METHODS AND DISCUSSION FL can offer significant benefits in transfusion medicine by enhancing predictive analytics, personalized medicine, and operational efficiency. Predictive models trained on diverse datasets by FL can improve accuracy in forecasting blood transfusion demands. Personalized treatment plans can be refined by aggregating patient data from multiple institutions using FL, reducing adverse reactions and improving outcomes. Operational efficiency can also be achieved through precise demand forecasting and optimized logistics. Despite its advantages, FL faces challenges such as data standardization, governance, and bias. Harmonizing diverse data sources and ensuring fair, unbiased models require advanced analytical solutions. Robust IT infrastructure and specialized expertise are needed for successful FL implementation. CONCLUSION FL represents a transformative approach to AI development in healthcare, particularly in transfusion medicine. By leveraging diverse datasets while maintaining data privacy, FL has the potential to enhance predictions, support personalized treatments, and optimize resource management, ultimately improving patient care and healthcare efficiency.
Collapse
Affiliation(s)
- Na Li
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Computing and SoftwareMcMaster UniversityHamiltonOntarioCanada
- Michael G. DeGroote Centre for Transfusion ResearchMcMaster UniversityHamiltonOntarioCanada
| | - Antoine Lewin
- Medical Affairs and Innovation, Héma‐Québec, MontrealQuebecCanada
| | - Shuoyan Ning
- Michael G. DeGroote Centre for Transfusion ResearchMcMaster UniversityHamiltonOntarioCanada
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
- Department of Medicine, Michael G. DeGroote School of MedicineMcMaster UniversityHamiltonOntarioCanada
- Canadian Blood Services, AncasterOntarioCanada
| | - Marianne Waito
- Integrated Supply Chain Planning, Canadian Blood ServicesOttawaOntarioCanada
| | - Michelle P. Zeller
- Michael G. DeGroote Centre for Transfusion ResearchMcMaster UniversityHamiltonOntarioCanada
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
- Department of Medicine, Michael G. DeGroote School of MedicineMcMaster UniversityHamiltonOntarioCanada
- Canadian Blood Services, AncasterOntarioCanada
| | - Alan Tinmouth
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Andrew W. Shih
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
- Centre for Innovation, Canadian Blood ServicesOttawaOntarioCanada
| | | |
Collapse
|
8
|
Bujotzek MR, Akünal Ü, Denner S, Neher P, Zenk M, Frodl E, Jaiswal A, Kim M, Krekiehn NR, Nickel M, Ruppel R, Both M, Döllinger F, Opitz M, Persigehl T, Kleesiek J, Penzkofer T, Maier-Hein K, Bucher A, Braren R. Real-world federated learning in radiology: hurdles to overcome and benefits to gain. J Am Med Inform Assoc 2025; 32:193-205. [PMID: 39455061 DOI: 10.1093/jamia/ocae259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE Federated Learning (FL) enables collaborative model training while keeping data locally. Currently, most FL studies in radiology are conducted in simulated environments due to numerous hurdles impeding its translation into practice. The few existing real-world FL initiatives rarely communicate specific measures taken to overcome these hurdles. To bridge this significant knowledge gap, we propose a comprehensive guide for real-world FL in radiology. Minding efforts to implement real-world FL, there is a lack of comprehensive assessments comparing FL to less complex alternatives in challenging real-world settings, which we address through extensive benchmarking. MATERIALS AND METHODS We developed our own FL infrastructure within the German Radiological Cooperative Network (RACOON) and demonstrated its functionality by training FL models on lung pathology segmentation tasks across six university hospitals. Insights gained while establishing our FL initiative and running the extensive benchmark experiments were compiled and categorized into the guide. RESULTS The proposed guide outlines essential steps, identified hurdles, and implemented solutions for establishing successful FL initiatives conducting real-world experiments. Our experimental results prove the practical relevance of our guide and show that FL outperforms less complex alternatives in all evaluation scenarios. DISCUSSION AND CONCLUSION Our findings justify the efforts required to translate FL into real-world applications by demonstrating advantageous performance over alternative approaches. Additionally, they emphasize the importance of strategic organization, robust management of distributed data and infrastructure in real-world settings. With the proposed guide, we are aiming to aid future FL researchers in circumventing pitfalls and accelerating translation of FL into radiological applications.
Collapse
Affiliation(s)
- Markus Ralf Bujotzek
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
- Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, 69120, Germany
| | - Ünal Akünal
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
| | - Stefan Denner
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
- Faculty of Mathematics and Computer Science, Heidelberg University, Heidelberg, 69120, Germany
| | - Peter Neher
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
- Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, 69120, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, Heidelberg, 69120, Germany
| | - Maximilian Zenk
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
- Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, 69120, Germany
| | - Eric Frodl
- Institute for Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt (Main), 60590, Germany
- Goethe University Frankfurt, Frankfurt, 60590, Germany
| | - Astha Jaiswal
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
| | - Moon Kim
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, 45131, Germany
| | - Nicolai R Krekiehn
- Intelligent Imaging Lab@Section Biomedical Imaging, Department of Radiology and Neuroradiology, University Medical Center Schleswig-Holstein (UKSH), Kel, 24118, Germany
| | - Manuel Nickel
- Institute for AI in Medicine, Technical University of Munich, Munich, 81675, Germany
| | - Richard Ruppel
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Marcus Both
- Department of Radiology and Neuroradiology, University Medical Centers Schleswig-Holstein, Kiel, 24105, Germany
| | - Felix Döllinger
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Marcel Opitz
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen (AÖR), Essen, 45131, Germany
| | - Thorsten Persigehl
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
| | - Jens Kleesiek
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, 45131, Germany
| | - Tobias Penzkofer
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin, 10117, Germany
- Berlin Institute of Health, Berlin, 10178, Germany
| | - Klaus Maier-Hein
- Division of Medical Image Computing, German Cancer Research Center Heidelberg, Heidelberg, 69120, Germany
- Pattern Analysis and Learning Group, Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, 69120, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, Heidelberg, 69120, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, A Partnership Between DKFZ and The University Medical Center Heidelberg, Heidelberg, 69120, Germany
| | - Andreas Bucher
- Institute for Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt (Main), 60590, Germany
- Goethe University Frankfurt, Frankfurt, 60590, Germany
| | - Rickmer Braren
- Institute for Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| |
Collapse
|
9
|
Zhang K, Yang X, Wang Y, Yu Y, Huang N, Li G, Li X, Wu JC, Yang S. Artificial intelligence in drug development. Nat Med 2025; 31:45-59. [PMID: 39833407 DOI: 10.1038/s41591-024-03434-4] [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: 05/01/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025]
Abstract
Drug development is a complex and time-consuming endeavor that traditionally relies on the experience of drug developers and trial-and-error experimentation. The advent of artificial intelligence (AI) technologies, particularly emerging large language models and generative AI, is poised to redefine this paradigm. The integration of AI-driven methodologies into the drug development pipeline has already heralded subtle yet meaningful enhancements in both the efficiency and effectiveness of this process. Here we present an overview of recent advancements in AI applications across the entire drug development workflow, encompassing the identification of disease targets, drug discovery, preclinical and clinical studies, and post-market surveillance. Lastly, we critically examine the prevailing challenges to highlight promising future research directions in AI-augmented drug development.
Collapse
Affiliation(s)
- Kang Zhang
- Eye Hospital and Institute for Advanced Study on Eye Health and Diseases, Institute for clinical Data Science, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, Wenzhou Medical University, Wenzhou, China.
| | - Xin Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yunfang Yu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute for AI in Medicine and faculty of Medicine, Macau University of Science and Technology, Macau, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Niu Huang
- National Institute of Biological Sciences, Beijing, China
| | - Gen Li
- Eye Hospital and Institute for Advanced Study on Eye Health and Diseases, Institute for clinical Data Science, Wenzhou Medical University, Wenzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Eye and Vision Innovation Center, Eye Valley, Wenzhou, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, Wenzhou Medical University, Wenzhou, China
| | - Joseph C Wu
- Cardiovascular Research Institute, Stanford University, Stanford, CA, USA
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Zhao Y, Liu Q, Liu P, Liu X, He K. Medical Federated Model With Mixture of Personalized and Shared Components. IEEE TRANSACTIONS ON PATTERN ANALYSIS AND MACHINE INTELLIGENCE 2025; 47:433-449. [PMID: 39331555 DOI: 10.1109/tpami.2024.3470072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Although data-driven methods usually have noticeable performance on disease diagnosis and treatment, they are suspected of leakage of privacy due to collecting data for model training. Recently, federated learning provides a secure and trustable alternative to collaboratively train model without any exchange of medical data among multiple institutes. Therefore, it has draw much attention due to its natural merit on privacy protection. However, when heterogenous medical data exists between different hospitals, federated learning usually has to face with degradation of performance. In the paper, we propose a new personalized framework of federated learning to handle the problem. It successfully yields personalized models based on awareness of similarity between local data, and achieves better tradeoff between generalization and personalization than existing methods. After that, we further design a differentially sparse regularizer to improve communication efficiency during procedure of model training. Additionally, we propose an effective method to reduce the computational cost, which improves computation efficiency significantly. Furthermore, we collect five real medical datasets, including two public medical image datasets and three private multi-center clinical diagnosis datasets, and evaluate its performance by conducting nodule classification, tumor segmentation, and clinical risk prediction tasks. Comparing with 14 existing related methods, the proposed method successfully achieves the best model performance, and meanwhile up to 60% improvement of communication efficiency.
Collapse
|
11
|
Schoenpflug LA, Nie Y, Sheikhzadeh F, Koelzer VH. A review on federated learning in computational pathology. Comput Struct Biotechnol J 2024; 23:3938-3945. [PMID: 39582895 PMCID: PMC11584763 DOI: 10.1016/j.csbj.2024.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024] Open
Abstract
Training generalizable computational pathology (CPATH) algorithms is heavily dependent on large-scale, multi-institutional data. Simultaneously, healthcare data underlies strict data privacy rules, hindering the creation of large datasets. Federated Learning (FL) is a paradigm addressing this dilemma, by allowing separate institutions to collaborate in a training process while keeping each institution's data private and exchanging model parameters instead. In this study, we identify and review key developments of FL for CPATH applications. We consider 15 studies, thereby evaluating the current status of exploring and adapting this emerging technology for CPATH applications. Proof-of-concept studies have been conducted across a wide range of CPATH use cases, showcasing the performance equivalency of models trained in a federated compared to a centralized manner. Six studies focus on model aggregation or model alignment methods reporting minor ( 0 ∼ 3 % ) performance improvement compared to conventional FL techniques, while four studies explore domain alignment methods, resulting in more significant performance improvements ( 4 ∼ 20 % ). To further reduce the privacy risk posed by sharing model parameters, four studies investigated the use of privacy preservation methods, where all methods demonstrated equivalent or slightly degraded performance ( 0.2 ∼ 6 % lower). To facilitate broader, real-world environment adoption, it is imperative to establish guidelines for the setup and deployment of FL infrastructure, alongside the promotion of standardized software frameworks. These steps are crucial to 1) further democratize CPATH research by allowing smaller institutions to pool data and computational resources 2) investigating rare diseases, 3) conducting multi-institutional studies, and 4) allowing rapid prototyping on private data.
Collapse
Affiliation(s)
- Lydia A. Schoenpflug
- Department of Pathology and Molecular Pathology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Yao Nie
- Roche Diagnostics, Digital Pathology, Santa Clara, CA, United States
| | | | - Viktor H. Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zürich, Zürich, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Department of Oncology, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Deng T, Huang Y, Han G, Shi Z, Lin J, Dou Q, Liu Z, Guo XJ, Philip Chen CL, Han C. FedDBL: Communication and Data Efficient Federated Deep-Broad Learning for Histopathological Tissue Classification. IEEE TRANSACTIONS ON CYBERNETICS 2024; 54:7851-7864. [PMID: 38923486 DOI: 10.1109/tcyb.2024.3403927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Histopathological tissue classification is a fundamental task in computational pathology. Deep learning (DL)-based models have achieved superior performance but centralized training suffers from the privacy leakage problem. Federated learning (FL) can safeguard privacy by keeping training samples locally, while existing FL-based frameworks require a large number of well-annotated training samples and numerous rounds of communication which hinder their viability in real-world clinical scenarios. In this article, we propose a lightweight and universal FL framework, named federated deep-broad learning (FedDBL), to achieve superior classification performance with limited training samples and only one-round communication. By simply integrating a pretrained DL feature extractor, a fast and lightweight broad learning inference system with a classical federated aggregation approach, FedDBL can dramatically reduce data dependency and improve communication efficiency. Five-fold cross-validation demonstrates that FedDBL greatly outperforms the competitors with only one-round communication and limited training samples, while it even achieves comparable performance with the ones under multiple-round communications. Furthermore, due to the lightweight design and one-round communication, FedDBL reduces the communication burden from 4.6 GB to only 138.4 KB per client using the ResNet-50 backbone at 50-round training. Extensive experiments also show the scalability of FedDBL on model generalization to the unseen dataset, various client numbers, model personalization and other image modalities. Since no data or deep model sharing across different clients, the privacy issue is well-solved and the model security is guaranteed with no model inversion attack risk. Code is available at https://github.com/tianpeng-deng/FedDBL.
Collapse
|
13
|
Gao Y, Ventura-Diaz S, Wang X, He M, Xu Z, Weir A, Zhou HY, Zhang T, van Duijnhoven FH, Han L, Li X, D'Angelo A, Longo V, Liu Z, Teuwen J, Kok M, Beets-Tan R, Horlings HM, Tan T, Mann R. An explainable longitudinal multi-modal fusion model for predicting neoadjuvant therapy response in women with breast cancer. Nat Commun 2024; 15:9613. [PMID: 39511143 PMCID: PMC11544255 DOI: 10.1038/s41467-024-53450-8] [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: 01/12/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Multi-modal image analysis using deep learning (DL) lays the foundation for neoadjuvant treatment (NAT) response monitoring. However, existing methods prioritize extracting multi-modal features to enhance predictive performance, with limited consideration on real-world clinical applicability, particularly in longitudinal NAT scenarios with multi-modal data. Here, we propose the Multi-modal Response Prediction (MRP) system, designed to mimic real-world physician assessments of NAT responses in breast cancer. To enhance feasibility, MRP integrates cross-modal knowledge mining and temporal information embedding strategy to handle missing modalities and remain less affected by different NAT settings. We validated MRP through multi-center studies and multinational reader studies. MRP exhibited comparable robustness to breast radiologists, outperforming humans in predicting pathological complete response in the Pre-NAT phase (ΔAUROC 14% and 10% on in-house and external datasets, respectively). Furthermore, we assessed MRP's clinical utility impact on treatment decision-making. MRP may have profound implications for enrolment into NAT trials and determining surgery extensiveness.
Collapse
Affiliation(s)
- Yuan Gao
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Sofia Ventura-Diaz
- Department of Radiology, St Joseph's Healthcare Hamilton, 50 Charlton Ave E, Hamilton, ON L8N 4A6, Ontario, Canada
| | - Xin Wang
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Muzhen He
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Zeyan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Arlene Weir
- Department of Radiology, Cork University Hospital, Wilton, Cork, T12 DC4A, Ireland
| | - Hong-Yu Zhou
- Department of Biomedical Informatics, Harvard Medical School, Boston, USA
| | - Tianyu Zhang
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Frederieke H van Duijnhoven
- Departments of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Luyi Han
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Xiaomei Li
- The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, 518020, China
| | - Anna D'Angelo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Rome, Italy
| | - Valentina Longo
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Rome, Italy
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jonas Teuwen
- Department of Radiation Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Marleen Kok
- Department of Tumor Biology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Regina Beets-Tan
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Hugo M Horlings
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Tao Tan
- Faculty of Applied Sciences, Macao Polytechnic University, 999078, Macao, China.
| | - Ritse Mann
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Medical Imaging, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Verlingue L, Boyer C, Olgiati L, Brutti Mairesse C, Morel D, Blay JY. Artificial intelligence in oncology: ensuring safe and effective integration of language models in clinical practice. THE LANCET REGIONAL HEALTH. EUROPE 2024; 46:101064. [PMID: 39290808 PMCID: PMC11406067 DOI: 10.1016/j.lanepe.2024.101064] [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: 02/21/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
In this Personal View, we address the latest advancements in automatic text analysis with artificial intelligence (AI) in medicine, with a focus on its implications in aiding treatment decisions in medical oncology. Acknowledging that a majority of hospital medical content is embedded in narrative format, natural language processing has become one of the most dynamic research fields for developing clinical decision support tools. In addition, large language models have recently reached unprecedented performance, notably when answering medical questions. Emerging applications include prognosis estimation, treatment recommendations, multidisciplinary tumor board recommendations and matching patients to recruiting clinical trials. Altogether, we advocate for a forward-looking approach in which the community efficiently initiates global prospective clinical evaluations of promising AI-based decision support systems. Such assessments will be essential to validate and evaluate potential biases, ensuring these innovations can be effectively and safely translated into practical tools for oncological practice. We are at a pivotal moment, where continued advancements in patient care must be pursued with scientific rigor.
Collapse
Affiliation(s)
- Loïc Verlingue
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
| | - Clara Boyer
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, France
| | - Louise Olgiati
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, France
| | | | - Daphné Morel
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - Jean-Yves Blay
- Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, France
| |
Collapse
|
15
|
Chen Z, Kim E, Davidsen T, Barnholtz-Sloan JS. Usage of the National Cancer Institute Cancer Research Data Commons by Researchers: A Scoping Review of the Literature. JCO Clin Cancer Inform 2024; 8:e2400116. [PMID: 39536277 PMCID: PMC11575903 DOI: 10.1200/cci.24.00116] [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: 05/14/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Over the past decade, significant surges in cancer data of all types have happened. To promote sharing and use of these rich data, the National Cancer Institute's Cancer Research Data Commons (CRDC) was developed as a cloud-based infrastructure that provides a large, comprehensive, and expanding collection of cancer data with tools for analysis. We conducted this scoping review of articles to provide an overview of how CRDC resources are being used by cancer researchers. METHODS A thorough literature search was conducted to identify all relevant publications. We included publications that directly cited CRDC resources to specifically examine the impact and contributions of CRDC by itself. We summarized the distributions and trends of how CRDC components were used by the research community and discussed current research gaps and future opportunities. RESULTS In terms of CRDC resources used by the research community, encouraging trends in utilization were observed, suggesting that CRDC has become an important building block for fostering a wide range of cancer research. We also noted a few areas where current applications are rather lacking and provided insights on how improvements can be made by CRDC and research community. CONCLUSION CRDC, as the foundation of a National Cancer Data Ecosystem, will continue empowering the research community to effectively leverage cancer-related data, uncover novel strategies, and address the needs of patients with cancer, ultimately combatting this disease more effectively.
Collapse
Affiliation(s)
- Zhaoyi Chen
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, MD
- Office of Data Science and Strategy, National Institutes of Health, Bethesda, MD
| | - Erika Kim
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, MD
| | - Tanja Davidsen
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, MD
| | - Jill S. Barnholtz-Sloan
- Informatics and Data Science Program, Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, MD
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
16
|
Abbas S, Asif M, Rehman A, Alharbi M, Khan MA, Elmitwally N. Emerging research trends in artificial intelligence for cancer diagnostic systems: A comprehensive review. Heliyon 2024; 10:e36743. [PMID: 39263113 PMCID: PMC11387343 DOI: 10.1016/j.heliyon.2024.e36743] [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: 04/27/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
This review article offers a comprehensive analysis of current developments in the application of machine learning for cancer diagnostic systems. The effectiveness of machine learning approaches has become evident in improving the accuracy and speed of cancer detection, addressing the complexities of large and intricate medical datasets. This review aims to evaluate modern machine learning techniques employed in cancer diagnostics, covering various algorithms, including supervised and unsupervised learning, as well as deep learning and federated learning methodologies. Data acquisition and preprocessing methods for different types of data, such as imaging, genomics, and clinical records, are discussed. The paper also examines feature extraction and selection techniques specific to cancer diagnosis. Model training, evaluation metrics, and performance comparison methods are explored. Additionally, the review provides insights into the applications of machine learning in various cancer types and discusses challenges related to dataset limitations, model interpretability, multi-omics integration, and ethical considerations. The emerging field of explainable artificial intelligence (XAI) in cancer diagnosis is highlighted, emphasizing specific XAI techniques proposed to improve cancer diagnostics. These techniques include interactive visualization of model decisions and feature importance analysis tailored for enhanced clinical interpretation, aiming to enhance both diagnostic accuracy and transparency in medical decision-making. The paper concludes by outlining future directions, including personalized medicine, federated learning, deep learning advancements, and ethical considerations. This review aims to guide researchers, clinicians, and policymakers in the development of efficient and interpretable machine learning-based cancer diagnostic systems.
Collapse
Affiliation(s)
- Sagheer Abbas
- Department of Computer Science, Prince Mohammad Bin Fahd University, Al-Khobar, KSA
| | - Muhammad Asif
- Department of Computer Science, Education University Lahore, Attock Campus, Pakistan
| | - Abdur Rehman
- School of Computer Science, National College of Business Administration and Economics, Lahore, 54000, Pakistan
| | - Meshal Alharbi
- Department of Computer Science, College of Computer Engineering and Sciences, Prince Sattam Bin Abdulaziz University, 11942, Alkharj, Saudi Arabia
| | - Muhammad Adnan Khan
- Riphah School of Computing & Innovation, Faculty of Computing, Riphah International University, Lahore Campus, Lahore, 54000, Pakistan
- School of Computing, Skyline University College, University City Sharjah, 1797, Sharjah, United Arab Emirates
- Department of Software, Faculty of Artificial Intelligence and Software, Gachon University, Seongnam-si, 13120, Republic of Korea
| | - Nouh Elmitwally
- Department of Computer Science, Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt
- School of Computing and Digital Technology, Birmingham City University, Birmingham, B4 7XG, UK
| |
Collapse
|
17
|
Yeom JC, Kim JH, Kim YJ, Kim J, Kim KG. A Comparative Study of Performance Between Federated Learning and Centralized Learning Using Pathological Image of Endometrial Cancer. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1683-1690. [PMID: 38381385 PMCID: PMC11300724 DOI: 10.1007/s10278-024-01020-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 02/22/2024]
Abstract
Federated learning, an innovative artificial intelligence training method, offers a secure solution for institutions to collaboratively develop models without sharing raw data. This approach offers immense promise and is particularly advantageous for domains dealing with sensitive information, such as patient data. However, when confronted with a distributed data environment, challenges arise due to data paucity or inherent heterogeneity, potentially impacting the performance of federated learning models. Hence, scrutinizing the efficacy of this method in such intricate settings is indispensable. To address this, we harnessed pathological image datasets of endometrial cancer from four hospitals for training and evaluating the performance of a federated learning model and compared it with a centralized learning model. With optimal processing techniques (data augmentation, color normalization, and adaptive optimizer), federated learning exhibited lower precision but higher recall and Dice similarity coefficient (DSC) than centralized learning. Hence, considering the critical importance of recall in the context of medical image processing, federated learning is demonstrated as a viable and applicable approach in this field, offering advantages in terms of both performance and data security.
Collapse
Affiliation(s)
- Jong Chan Yeom
- Department of Bio-health Medical Engineering, Gachon University, Seongnam, Republic of Korea
| | - Jae Hoon Kim
- Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Young Jae Kim
- Department of Biomedical Engineering, Gachon University, 191, Hambangmoe-ro, Yeonsu-gu, Incheon, 21936, Korea
- Department of Biomedical Engineering, Gachon University College of Medicine, Gil Medical Center, 38-13 Docjeom-ro 3 Beon-gil, Namdong-gu, Incheon, 21565, Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Seongnam-si, 13120, Korea
| | - Jisup Kim
- Department of Pathology, Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, Republic of Korea.
| | - Kwang Gi Kim
- Department of Biomedical Engineering, Gachon University, 191, Hambangmoe-ro, Yeonsu-gu, Incheon, 21936, Korea.
- Department of Biomedical Engineering, Gachon University College of Medicine, Gil Medical Center, 38-13 Docjeom-ro 3 Beon-gil, Namdong-gu, Incheon, 21565, Korea.
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Seongnam-si, 13120, Korea.
| |
Collapse
|
18
|
Wang Y, Sun W, Karlsson E, Kang Lövgren S, Ács B, Rantalainen M, Robertson S, Hartman J. Clinical evaluation of deep learning-based risk profiling in breast cancer histopathology and comparison to an established multigene assay. Breast Cancer Res Treat 2024; 206:163-175. [PMID: 38592541 PMCID: PMC11182789 DOI: 10.1007/s10549-024-07303-z] [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: 01/28/2024] [Accepted: 02/26/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE To evaluate the Stratipath Breast tool for image-based risk profiling and compare it with an established prognostic multigene assay for risk profiling in a real-world case series of estrogen receptor (ER)-positive and human epidermal growth factor receptor 2 (HER2)-negative early breast cancer patients categorized as intermediate risk based on classic clinicopathological variables and eligible for chemotherapy. METHODS In a case series comprising 234 invasive ER-positive/HER2-negative tumors, clinicopathological data including Prosigna results and corresponding HE-stained tissue slides were retrieved. The digitized HE slides were analysed by Stratipath Breast. RESULTS Our findings showed that the Stratipath Breast analysis identified 49.6% of the clinically intermediate tumors as low risk and 50.4% as high risk. The Prosigna assay classified 32.5%, 47.0% and 20.5% tumors as low, intermediate and high risk, respectively. Among Prosigna intermediate-risk tumors, 47.3% were stratified as Stratipath low risk and 52.7% as high risk. In addition, 89.7% of Stratipath low-risk cases were classified as Prosigna low/intermediate risk. The overall agreement between the two tests for low-risk and high-risk groups (N = 124) was 71.0%, with a Cohen's kappa of 0.42. For both risk profiling tests, grade and Ki67 differed significantly between risk groups. CONCLUSION The results from this clinical evaluation of image-based risk stratification shows a considerable agreement to an established gene expression assay in routine breast pathology.
Collapse
Affiliation(s)
- Yinxi Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Stratipath AB, Nanna Svartz väg 4, Stockholm, 171 65, Sweden
| | - Wenwen Sun
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Emelie Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Sandy Kang Lövgren
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Stratipath AB, Nanna Svartz väg 4, Stockholm, 171 65, Sweden
| | - Balázs Ács
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
| | - Stephanie Robertson
- Stratipath AB, Nanna Svartz väg 4, Stockholm, 171 65, Sweden.
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Lecuelle J, Truntzer C, Basile D, Laghi L, Greco L, Ilie A, Rageot D, Emile JF, Bibeau F, Taïeb J, Derangere V, Lepage C, Ghiringhelli F. Machine learning evaluation of immune infiltrate through digital tumour score allows prediction of survival outcome in a pooled analysis of three international stage III colon cancer cohorts. EBioMedicine 2024; 105:105207. [PMID: 38880067 PMCID: PMC11233898 DOI: 10.1016/j.ebiom.2024.105207] [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: 12/18/2023] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND T-cell immune infiltrates are robust prognostic variables in localised colon cancer. Evaluation of prognosis using artificial intelligence is an emerging field. We evaluated whether machine learning analysis improved prediction of patient outcome in comparison with analysis of T cell infiltrate only or in association with clinical variables. METHODS We used data from two phase III clinical trials (Prodige-13 and PETACC08) and one retrospective Italian cohort (HARMONY). Cohorts were split into training (N = 692), internal validation (N = 297) and external validation (N = 672) sets. Tumour slides were stained with CD3mAb. CD3 Machine Learning (CD3ML) score was computed using graphical parameters within the tumour tiles obtained from CD3 slides. CD3 infiltrates in tumour core and invasive margin were automatically detected. Associations of CD3 infiltrates and CD3ML with 5-year Disease-Free Survival (DFS) were examined using univariate and multivariable survival models by Cox regression. FINDINGS CD3 density both in the invasive margin and the tumour core were significantly associated with DFS in the different sets. Similarly, CD3ML score was significantly associated with DFS in all sets. CD3 assessment did not provide added value on top of CD3ML assessment (Likelihood Ratio Test (LRT), p = 0.13). In contrast, CD3ML improved prediction of DFS when combined with a clinical risk stage (LRT, p = 0.001). Stratified by clinical risk score (High or Low), patients with low CD3ML score had better DFS. INTERPRETATION In all tested sets, machine learning analysis of tumour cells improved prediction of prognosis compared to clinical parameters. Adding tumour-infiltrating lymphocytes assessment did not improve prognostic determination. FUNDING This research received no external funding.
Collapse
Affiliation(s)
- Julie Lecuelle
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Caroline Truntzer
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Debora Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, Crotone, Italy
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luana Greco
- Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alis Ilie
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - David Rageot
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Jean-François Emile
- Paris-Saclay University, Versailles SQY University (UVSQ), EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Ambroise Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| | - Fréderic Bibeau
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Normandie Université, Caen, France; Department of Pathology, Besançon University Hospital, Besançon, France
| | - Julien Taïeb
- Institut du Cancer Paris Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique, Sorbonne Université, Université Sorbonne Paris Cité, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Valentin Derangere
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France
| | - Come Lepage
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Fédération Francophone de Cancérologie Digestive, Centre de Randomisation Gestion Analyse, EPICAD LNC 1231, Dijon, France; Service d'Hépato-gastroentérologie et Oncologie digestive, CHU de Dijon, France
| | - François Ghiringhelli
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France.
| |
Collapse
|
20
|
Guan H, Yap PT, Bozoki A, Liu M. Federated learning for medical image analysis: A survey. PATTERN RECOGNITION 2024; 151:110424. [PMID: 38559674 PMCID: PMC10976951 DOI: 10.1016/j.patcog.2024.110424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Machine learning in medical imaging often faces a fundamental dilemma, namely, the small sample size problem. Many recent studies suggest using multi-domain data pooled from different acquisition sites/centers to improve statistical power. However, medical images from different sites cannot be easily shared to build large datasets for model training due to privacy protection reasons. As a promising solution, federated learning, which enables collaborative training of machine learning models based on data from different sites without cross-site data sharing, has attracted considerable attention recently. In this paper, we conduct a comprehensive survey of the recent development of federated learning methods in medical image analysis. We have systematically gathered research papers on federated learning and its applications in medical image analysis published between 2017 and 2023. Our search and compilation were conducted using databases from IEEE Xplore, ACM Digital Library, Science Direct, Springer Link, Web of Science, Google Scholar, and PubMed. In this survey, we first introduce the background of federated learning for dealing with privacy protection and collaborative learning issues. We then present a comprehensive review of recent advances in federated learning methods for medical image analysis. Specifically, existing methods are categorized based on three critical aspects of a federated learning system, including client end, server end, and communication techniques. In each category, we summarize the existing federated learning methods according to specific research problems in medical image analysis and also provide insights into the motivations of different approaches. In addition, we provide a review of existing benchmark medical imaging datasets and software platforms for current federated learning research. We also conduct an experimental study to empirically evaluate typical federated learning methods for medical image analysis. This survey can help to better understand the current research status, challenges, and potential research opportunities in this promising research field.
Collapse
Affiliation(s)
- Hao Guan
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pew-Thian Yap
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrea Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mingxia Liu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
21
|
Boissin C, Wang Y, Sharma A, Weitz P, Karlsson E, Robertson S, Hartman J, Rantalainen M. Deep learning-based risk stratification of preoperative breast biopsies using digital whole slide images. Breast Cancer Res 2024; 26:90. [PMID: 38831336 PMCID: PMC11145850 DOI: 10.1186/s13058-024-01840-7] [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: 12/21/2023] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Nottingham histological grade (NHG) is a well established prognostic factor in breast cancer histopathology but has a high inter-assessor variability with many tumours being classified as intermediate grade, NHG2. Here, we evaluate if DeepGrade, a previously developed model for risk stratification of resected tumour specimens, could be applied to risk-stratify tumour biopsy specimens. METHODS A total of 11,955,755 tiles from 1169 whole slide images of preoperative biopsies from 896 patients diagnosed with breast cancer in Stockholm, Sweden, were included. DeepGrade, a deep convolutional neural network model, was applied for the prediction of low- and high-risk tumours. It was evaluated against clinically assigned grades NHG1 and NHG3 on the biopsy specimen but also against the grades assigned to the corresponding resection specimen using area under the operating curve (AUC). The prognostic value of the DeepGrade model in the biopsy setting was evaluated using time-to-event analysis. RESULTS Based on preoperative biopsy images, the DeepGrade model predicted resected tumour cases of clinical grades NHG1 and NHG3 with an AUC of 0.908 (95% CI: 0.88; 0.93). Furthermore, out of the 432 resected clinically-assigned NHG2 tumours, 281 (65%) were classified as DeepGrade-low and 151 (35%) as DeepGrade-high. Using a multivariable Cox proportional hazards model the hazard ratio between DeepGrade low- and high-risk groups was estimated as 2.01 (95% CI: 1.06; 3.79). CONCLUSIONS DeepGrade provided prediction of tumour grades NHG1 and NHG3 on the resection specimen using only the biopsy specimen. The results demonstrate that the DeepGrade model can provide decision support to identify high-risk tumours based on preoperative biopsies, thus improving early treatment decisions.
Collapse
Affiliation(s)
- Constance Boissin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yinxi Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Abhinav Sharma
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Philippe Weitz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Emelie Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
22
|
Bryant AK, Zamora‐Resendiz R, Dai X, Morrow D, Lin Y, Jungles KM, Rae JM, Tate A, Pearson AN, Jiang R, Fritsche L, Lawrence TS, Zou W, Schipper M, Ramnath N, Yoo S, Crivelli S, Green MD. Artificial intelligence to unlock real-world evidence in clinical oncology: A primer on recent advances. Cancer Med 2024; 13:e7253. [PMID: 38899720 PMCID: PMC11187737 DOI: 10.1002/cam4.7253] [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: 08/17/2023] [Revised: 02/05/2024] [Accepted: 04/28/2024] [Indexed: 06/21/2024] Open
Abstract
PURPOSE Real world evidence is crucial to understanding the diffusion of new oncologic therapies, monitoring cancer outcomes, and detecting unexpected toxicities. In practice, real world evidence is challenging to collect rapidly and comprehensively, often requiring expensive and time-consuming manual case-finding and annotation of clinical text. In this Review, we summarise recent developments in the use of artificial intelligence to collect and analyze real world evidence in oncology. METHODS We performed a narrative review of the major current trends and recent literature in artificial intelligence applications in oncology. RESULTS Artificial intelligence (AI) approaches are increasingly used to efficiently phenotype patients and tumors at large scale. These tools also may provide novel biological insights and improve risk prediction through multimodal integration of radiographic, pathological, and genomic datasets. Custom language processing pipelines and large language models hold great promise for clinical prediction and phenotyping. CONCLUSIONS Despite rapid advances, continued progress in computation, generalizability, interpretability, and reliability as well as prospective validation are needed to integrate AI approaches into routine clinical care and real-time monitoring of novel therapies.
Collapse
Affiliation(s)
- Alex K. Bryant
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
| | - Rafael Zamora‐Resendiz
- Applied Mathematics and Computational Research Division, Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Xin Dai
- Computational Science Initiative, Brookhaven National LaboratoryUptonNew YorkUSA
| | - Destinee Morrow
- Applied Mathematics and Computational Research Division, Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Yuewei Lin
- Computational Science Initiative, Brookhaven National LaboratoryUptonNew YorkUSA
| | - Kassidy M. Jungles
- Department of PharmacologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - James M. Rae
- Department of PharmacologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of Internal MedicineUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Akshay Tate
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Ashley N. Pearson
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Ralph Jiang
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of StatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Lars Fritsche
- Department of StatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Theodore S. Lawrence
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Weiping Zou
- Department of StatisticsUniversity of MichiganAnn ArborMichiganUSA
- Center of Excellence for Cancer Immunology and ImmunotherapyUniversity of Michigan Rogel Cancer CenterAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Matthew Schipper
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of PharmacologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| | - Nithya Ramnath
- Division of Hematology Oncology, Department of MedicineUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Division of Hematology Oncology, Department of MedicineVeterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
| | - Shinjae Yoo
- Computational Science Initiative, Brookhaven National LaboratoryUptonNew YorkUSA
| | - Silvia Crivelli
- Applied Mathematics and Computational Research Division, Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Michael D. Green
- Department of Radiation OncologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare SystemAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in Cancer BiologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of Michigan School of MedicineAnn ArborMichiganUSA
| |
Collapse
|
23
|
Greeley B, Chung SS, Graves L, Song X. Combating Barriers to the Development of a Patient-Oriented Frailty Website. JMIR Aging 2024; 7:e53098. [PMID: 38807317 DOI: 10.2196/53098] [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: 09/25/2023] [Revised: 01/02/2024] [Accepted: 03/07/2024] [Indexed: 05/30/2024] Open
Abstract
Unlabelled This viewpoint article, which represents the opinions of the authors, discusses the barriers to developing a patient-oriented frailty website and potential solutions. A patient-oriented frailty website is a health resource where community-dwelling older adults can navigate to and answer a series of health-related questions to receive a frailty score and health summary. This information could then be shared with health care professionals to help with the understanding of health status prior to acute illness, as well as to screen and identify older adult individuals for frailty. Our viewpoints were drawn from 2 discussion sessions that included caregivers and care providers, as well as community-dwelling older adults. We found that barriers to a patient-oriented frailty website include, but are not limited to, its inherent restrictiveness to frail persons, concerns over data privacy, time commitment worries, and the need for health and lifestyle resources in addition to an assessment summary. For each barrier, we discuss potential solutions and caveats to those solutions, including assistance from caregivers, hosting the website on a trusted source, reducing the number of health questions that need to be answered, and providing resources tailored to each users' responses, respectively. In addition to screening and identifying frail older adults, a patient-oriented frailty website will help promote healthy aging in nonfrail adults, encourage aging in place, support real-time monitoring, and enable personalized and preventative care.
Collapse
|
24
|
Zhou J, Wang X, Li Y, Yang Y, Shi J. Federated-learning-based prognosis assessment model for acute pulmonary thromboembolism. BMC Med Inform Decis Mak 2024; 24:141. [PMID: 38802861 PMCID: PMC11131248 DOI: 10.1186/s12911-024-02543-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: 01/23/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Acute pulmonary thromboembolism (PTE) is a common cardiovascular disease and recognizing low prognosis risk patients with PTE accurately is significant for clinical treatment. This study evaluated the value of federated learning (FL) technology in PTE prognosis risk assessment while ensuring the security of clinical data. METHODS A retrospective dataset consisted of PTE patients from 12 hospitals were collected, and 19 physical indicators of patients were included to train the FL-based prognosis assessment model to predict the 30-day death event. Firstly, multiple machine learning methods based on FL were compared to choose the superior model. And then performance of models trained on the independent (IID) and non-independent identical distributed(Non-IID) datasets was calculated and they were tested further on Real-world data. Besides, the optimal model was compared with pulmonary embolism severity index (PESI), simplified PESI (sPESI), Peking Union Medical College Hospital (PUMCH). RESULTS The area under the receiver operating characteristic curve (AUC) of logistic regression(0.842) outperformed convolutional neural network (0.819) and multi layer perceptron (0.784). Under IID, AUC of model trained using FL(Fed) on the training, validation and test sets was 0.852 ± 0.002, 0.867 ± 0.012 and 0.829 ± 0.004. Under Real-world, AUC of Fed was 0.855 ± 0.005, 0.882 ± 0.003 and 0.835 ± 0.005. Under IID and Real-world, AUC of Fed surpassed centralization model(NonFed) (0.847 ± 0.001, 0.841 ± 0.001 and 0.811 ± 0.001). Under Non-IID, although AUC of Fed (0.846 ± 0.047) outperformed NonFed (0.841 ± 0.001) on validation set, it (0.821 ± 0.016 and 0.799 ± 0.031) slightly lagged behind NonFed (0.847 ± 0.001 and 0.811 ± 0.001) on the training and test sets. In practice, AUC of Fed (0.853, 0.884 and 0.842) outshone PESI (0.812, 0.789 and 0.791), sPESI (0.817, 0.770 and 0.786) and PUMCH(0.848, 0.814 and 0.832) on the training, validation and test sets. Additionally, Fed (0.842) exhibited higher AUC values across test sets compared to those trained directly on the clients (0.758, 0.801, 0.783, 0.741, 0.788). CONCLUSIONS In this study, the FL based machine learning model demonstrated commendable efficacy on PTE prognostic risk prediction, rendering it well-suited for deployment in hospitals.
Collapse
Affiliation(s)
- Jun Zhou
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, China
| | - Xin Wang
- Department of Ultrasound, Peking Union Medical College Hospital, Beijing, China
| | - Yiyao Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yuqing Yang
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, China.
| | - Juhong Shi
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
25
|
Lotter W, Hassett MJ, Schultz N, Kehl KL, Van Allen EM, Cerami E. Artificial Intelligence in Oncology: Current Landscape, Challenges, and Future Directions. Cancer Discov 2024; 14:711-726. [PMID: 38597966 PMCID: PMC11131133 DOI: 10.1158/2159-8290.cd-23-1199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/29/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Artificial intelligence (AI) in oncology is advancing beyond algorithm development to integration into clinical practice. This review describes the current state of the field, with a specific focus on clinical integration. AI applications are structured according to cancer type and clinical domain, focusing on the four most common cancers and tasks of detection, diagnosis, and treatment. These applications encompass various data modalities, including imaging, genomics, and medical records. We conclude with a summary of existing challenges, evolving solutions, and potential future directions for the field. SIGNIFICANCE AI is increasingly being applied to all aspects of oncology, where several applications are maturing beyond research and development to direct clinical integration. This review summarizes the current state of the field through the lens of clinical translation along the clinical care continuum. Emerging areas are also highlighted, along with common challenges, evolving solutions, and potential future directions for the field.
Collapse
Affiliation(s)
- William Lotter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael J. Hassett
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth L. Kehl
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M. Van Allen
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
26
|
Naik K, Goyal RK, Foschini L, Chak CW, Thielscher C, Zhu H, Lu J, Lehár J, Pacanoswki MA, Terranova N, Mehta N, Korsbo N, Fakhouri T, Liu Q, Gobburu J. Current Status and Future Directions: The Application of Artificial Intelligence/Machine Learning for Precision Medicine. Clin Pharmacol Ther 2024; 115:673-686. [PMID: 38103204 DOI: 10.1002/cpt.3152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023]
Abstract
Technological innovations, such as artificial intelligence (AI) and machine learning (ML), have the potential to expedite the goal of precision medicine, especially when combined with increased capacity for voluminous data from multiple sources and expanded therapeutic modalities; however, they also present several challenges. In this communication, we first discuss the goals of precision medicine, and contextualize the use of AI in precision medicine by showcasing innovative applications (e.g., prediction of tumor growth and overall survival, biomarker identification using biomedical images, and identification of patient population for clinical practice) which were presented during the February 2023 virtual public workshop entitled "Application of Artificial Intelligence and Machine Learning for Precision Medicine," hosted by the US Food and Drug Administration (FDA) and University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI). Next, we put forward challenges brought about by the multidisciplinary nature of AI, particularly highlighting the need for AI to be trustworthy. To address such challenges, we subsequently note practical approaches, viz., differential privacy, synthetic data generation, and federated learning. The proposed strategies - some of which are highlighted presentations from the workshop - are for the protection of personal information and intellectual property. In addition, methods such as the risk-based management approach and the need for an agile regulatory ecosystem are discussed. Finally, we lay out a call for action that includes sharing of data and algorithms, development of regulatory guidance documents, and pooling of expertise from a broad-spectrum of stakeholders to enhance the application of AI in precision medicine.
Collapse
Affiliation(s)
- Kunal Naik
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rahul K Goyal
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | | | | | | | - Hao Zhu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - James Lu
- Modeling & Simulation/Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | | | - Michael A Pacanoswki
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nadia Terranova
- Quantitative Pharmacology, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | - Neha Mehta
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Tala Fakhouri
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Qi Liu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jogarao Gobburu
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Khalighi S, Reddy K, Midya A, Pandav KB, Madabhushi A, Abedalthagafi M. Artificial intelligence in neuro-oncology: advances and challenges in brain tumor diagnosis, prognosis, and precision treatment. NPJ Precis Oncol 2024; 8:80. [PMID: 38553633 PMCID: PMC10980741 DOI: 10.1038/s41698-024-00575-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
This review delves into the most recent advancements in applying artificial intelligence (AI) within neuro-oncology, specifically emphasizing work on gliomas, a class of brain tumors that represent a significant global health issue. AI has brought transformative innovations to brain tumor management, utilizing imaging, histopathological, and genomic tools for efficient detection, categorization, outcome prediction, and treatment planning. Assessing its influence across all facets of malignant brain tumor management- diagnosis, prognosis, and therapy- AI models outperform human evaluations in terms of accuracy and specificity. Their ability to discern molecular aspects from imaging may reduce reliance on invasive diagnostics and may accelerate the time to molecular diagnoses. The review covers AI techniques, from classical machine learning to deep learning, highlighting current applications and challenges. Promising directions for future research include multimodal data integration, generative AI, large medical language models, precise tumor delineation and characterization, and addressing racial and gender disparities. Adaptive personalized treatment strategies are also emphasized for optimizing clinical outcomes. Ethical, legal, and social implications are discussed, advocating for transparency and fairness in AI integration for neuro-oncology and providing a holistic understanding of its transformative impact on patient care.
Collapse
Affiliation(s)
- Sirvan Khalighi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kartik Reddy
- Department of Radiology, Emory University, Atlanta, GA, USA
| | - Abhishek Midya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krunal Balvantbhai Pandav
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA.
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
- The Cell and Molecular Biology Program, Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
28
|
Sarfraz Z, Sarfraz A, Mehak O, Akhund R, Bano S, Aftab H. Racial and socioeconomic disparities in triple-negative breast cancer treatment. Expert Rev Anticancer Ther 2024; 24:107-116. [PMID: 38436305 DOI: 10.1080/14737140.2024.2326575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) continues to be a significant concern, especially among minority populations, where treatment disparities are notably pronounced. Addressing these disparities, especially among African American women and other minorities, is crucial for ensuring equitable healthcare. AREAS COVERED This review delves into the continuum of TNBC treatment, noting that the standard of care, previously restricted to chemotherapy, has now expanded due to emerging clinical trial results. With advances like PARP inhibitors, immunotherapy, and antibody-drug conjugates, a more personalized treatment approach is on the horizon. The review highlights innovative interventions tailored for minorities, such as utilizing technology like text messaging, smartphone apps, and targeted radio programming, coupled with church-based behavioral interventions. EXPERT OPINION Addressing TNBC treatment disparities demands a multifaceted approach, blending advanced medical treatments with culturally sensitive community outreach. The potential of technology, especially in the realm of promoting health awareness, is yet to be fully harnessed. As the field progresses, understanding and integrating the socio-economic, biological, and access-related challenges faced by minorities will be pivotal for achieving health equity in TNBC care.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Azza Sarfraz
- Department of Pediatrics, Aga Khan University, Karachi, Pakistan
| | - Onaiza Mehak
- Department of Medicine, Aziz Fatimah Medical and Dental College, Faisalabad, Pakistan
| | - Ramsha Akhund
- Department of Surgery, University of Alabama at Birmingham, Tuscaloosa, AL, USA
| | - Shehar Bano
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Hinna Aftab
- Department of Medicine, CMH Lahore Medical College, Lahore, Pakistan
| |
Collapse
|
29
|
Li P, Guo C, Xing Y, Shi Y, Feng L, Zhou F. Core network traffic prediction based on vertical federated learning and split learning. Sci Rep 2024; 14:4663. [PMID: 38409301 PMCID: PMC10897397 DOI: 10.1038/s41598-024-53193-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Wireless traffic prediction is vital for intelligent cellular network operations, such as load-aware resource management and predictive control. Traditional centralized training addresses this but poses issues like excessive data transmission, disregarding delays, and user privacy. Traditional federated learning methods can meet the requirement of jointly training models while protecting the privacy of all parties' data. However, challenges arise when the local data features among participating parties exhibit inconsistency, making the training process difficult to sustain. Our study introduces an innovative framework for wireless traffic prediction based on split learning (SL) and vertical federated learning. Multiple edge clients collaboratively train high-quality prediction models by utilizing diverse traffic data while maintaining the confidentiality of raw data locally. Each participant individually trains dimension-specific prediction models with their respective data, and the outcomes are aggregated through collaboration. A partially global model is formed and shared among clients to address statistical heterogeneity in distributed machine learning. Extensive experiments on real-world datasets demonstrate our method's superiority over current approaches, showcasing its potential for network traffic prediction and accurate forecasting.
Collapse
Affiliation(s)
- Pengyu Li
- 6G Research Center, China Telecom Research Institute, Beijing, 102209, China.
| | - Chengwei Guo
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Yanxia Xing
- 6G Research Center, China Telecom Research Institute, Beijing, 102209, China
| | - Yingji Shi
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Lei Feng
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Fanqin Zhou
- State Key Laboratory of Networking and Switching Technology, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| |
Collapse
|
30
|
Fisher TB, Saini G, Rekha TS, Krishnamurthy J, Bhattarai S, Callagy G, Webber M, Janssen EAM, Kong J, Aneja R. Digital image analysis and machine learning-assisted prediction of neoadjuvant chemotherapy response in triple-negative breast cancer. Breast Cancer Res 2024; 26:12. [PMID: 38238771 PMCID: PMC10797728 DOI: 10.1186/s13058-023-01752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Pathological complete response (pCR) is associated with favorable prognosis in patients with triple-negative breast cancer (TNBC). However, only 30-40% of TNBC patients treated with neoadjuvant chemotherapy (NAC) show pCR, while the remaining 60-70% show residual disease (RD). The role of the tumor microenvironment in NAC response in patients with TNBC remains unclear. In this study, we developed a machine learning-based two-step pipeline to distinguish between various histological components in hematoxylin and eosin (H&E)-stained whole slide images (WSIs) of TNBC tissue biopsies and to identify histological features that can predict NAC response. METHODS H&E-stained WSIs of treatment-naïve biopsies from 85 patients (51 with pCR and 34 with RD) of the model development cohort and 79 patients (41 with pCR and 38 with RD) of the validation cohort were separated through a stratified eightfold cross-validation strategy for the first step and leave-one-out cross-validation strategy for the second step. A tile-level histology label prediction pipeline and four machine-learning classifiers were used to analyze 468,043 tiles of WSIs. The best-trained classifier used 55 texture features from each tile to produce a probability profile during testing. The predicted histology classes were used to generate a histology classification map of the spatial distributions of different tissue regions. A patient-level NAC response prediction pipeline was trained with features derived from paired histology classification maps. The top graph-based features capturing the relevant spatial information across the different histological classes were provided to the radial basis function kernel support vector machine (rbfSVM) classifier for NAC treatment response prediction. RESULTS The tile-level prediction pipeline achieved 86.72% accuracy for histology class classification, while the patient-level pipeline achieved 83.53% NAC response (pCR vs. RD) prediction accuracy of the model development cohort. The model was validated with an independent cohort with tile histology validation accuracy of 83.59% and NAC prediction accuracy of 81.01%. The histological class pairs with the strongest NAC response predictive ability were tumor and tumor tumor-infiltrating lymphocytes for pCR and microvessel density and polyploid giant cancer cells for RD. CONCLUSION Our machine learning pipeline can robustly identify clinically relevant histological classes that predict NAC response in TNBC patients and may help guide patient selection for NAC treatment.
Collapse
Affiliation(s)
- Timothy B Fisher
- Department of Biology, Georgia State University, Atlanta, GA, 30302, USA
| | - Geetanjali Saini
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - T S Rekha
- JSSAHER (JSS Academy of Higher Education and Research) Medical College, Mysuru, Karnataka, India
| | - Jayashree Krishnamurthy
- JSSAHER (JSS Academy of Higher Education and Research) Medical College, Mysuru, Karnataka, India
| | - Shristi Bhattarai
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Grace Callagy
- Discipline of Pathology, University of Galway, Galway, Ireland
| | - Mark Webber
- Discipline of Pathology, University of Galway, Galway, Ireland
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, 30303, USA.
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30302, USA.
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
31
|
Boutros M, Baumann M, Bigas A, Chaabane L, Guérin J, Habermann JK, Jobard A, Pelicci PG, Stegle O, Tonon G, Valencia A, Winkler EC, Blanc P, De Maria R, Medema RH, Nagy P, Tabernero J, Solary E. UNCAN.eu: Toward a European Federated Cancer Research Data Hub. Cancer Discov 2024; 14:30-35. [PMID: 38213296 PMCID: PMC10784740 DOI: 10.1158/2159-8290.cd-23-1111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
To enable a collective effort that generates a new level of UNderstanding CANcer (UNCAN.eu) [Cancer Discov (2022) 12 (11): OF1], the European Union supports the creation of a sustainable platform that connects cancer research across Member States. A workshop hosted in Heidelberg gathered European cancer experts to identify ongoing initiatives that may contribute to building this platform and discuss the governance and long-term evolution of a European Federated Cancer Data Hub.
Collapse
Affiliation(s)
- Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics and Heidelberg University, Medical Faculty Heidelberg, Institute for Human Genetics, Heidelberg, Germany
| | | | - Anna Bigas
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Linda Chaabane
- Euro-BioImaging ERIC, Med-Hub, National Research Council of Italy (CNR), Turin, Italy
| | | | - Jens K. Habermann
- Interdisciplinary Center for Biobanking-Lübeck (ICB-L), University of Lübeck, Lübeck, Germany
| | - Aurélien Jobard
- Institut National du Cancer (INCa), Boulogne Billancourt, France
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milano, Italy
| | - Oliver Stegle
- DKFZ, Division of Computational Genomics and Systems Genetics, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology, Heidelberg, Germany
| | - Giovanni Tonon
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alfonso Valencia
- Barcelona Supercomputing Center, Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Eva C. Winkler
- National Center for Tumor Diseases (NCT), Heidelberg University, Section Translational Medical Ethics, Heidelberg, Germany
| | | | - Ruggero De Maria
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rene H. Medema
- Oncode Institute and The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Peter Nagy
- National Institute of Oncology and the National Tumor Biology Laboratory, Budapest, Department of Anatomy and Histology, HUN-REN–UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, and Chemistry Institute, University of Debrecen, Debrecen, Hungary
| | - Josep Tabernero
- DKFZ, Division of Computational Genomics and Systems Genetics, Heidelberg, Germany
- Vall d'Hebron Hospital Campus & Institute of Oncology (VHIO), Barcelona, Spain
| | - Eric Solary
- Université Paris-Saclay and INSERM, Gustave Roussy Cancer Center, Villejuif, France
| |
Collapse
|
32
|
Liu Z, Cai J, Jiang G, Wang M, Wu C, Su K, Hu W, Huang Y, Yu C, Huang X, Cao G, Wang H. Novel Platinum(IV) complexes intervene oxaliplatin resistance in colon cancer via inducing ferroptosis and apoptosis. Eur J Med Chem 2024; 263:115968. [PMID: 37995563 DOI: 10.1016/j.ejmech.2023.115968] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Platinum-based chemotherapeutics are widely used for cancer treatment but are frequently limited because of dosage-dependent side effects and drug resistance. To attenuate these drawbacks, a series of novel platinum(IV) prodrugs (15a-18c) were synthesized and evaluated for anti-cancer activity. Among them, 17a demonstrated superior anti-proliferative activity compared with oxaliplatin (OXA) in the cisplatin-resistant lung cancer cell line A549/CDDP and OXA-resistant colon cancer cell line HCT-116/OXA but showed a lower cytotoxic effect toward human normal cell lines HUVEC and L02. Mechanistic investigations suggested that 17a efficiently enhanced intracellular platinum accumulation, induced DNA damage, disturbed the homeostasis of intracellular reactive oxygen molecules and mitochondrial membrane potential, and thereby activated the mitochondrion-dependent apoptosis pathway. Moreover, 17a significantly induced ferroptosis in HCT-116/OXA via triggering the accumulation of lipid peroxides, disrupting iron homeostasis, and inhibiting solute carrier family 7 member 11 and glutathione peroxidase 4 axial pathway transduction by inhibiting the expression of the phosphorylated signal transducer and activator of transcription 3 and nuclear factor erythroid 2-related factor 2. Moreover, 17a exerted remarkable in vivo antitumor efficacy in the HCT-116/OXA xenograft models but showed attenuated toxicity. These results indicated that these novel platinum(IV) complexes provided an alternative strategy to develop novel platinum-based antineoplastic agents for cancer treatment.
Collapse
Affiliation(s)
- Zhikun Liu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Jinyuan Cai
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Guiyang Jiang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Meng Wang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China
| | - Chuang Wu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Kangning Su
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Weiwei Hu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Yaxian Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Chunhao Yu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Xiaochao Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| | - Guoxiu Cao
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China.
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
33
|
Chen X, Niu Y, Zhao Y, Qin X. An Efficient Group Federated Learning Framework for Large-Scale EEG-Based Driver Drowsiness Detection. Int J Neural Syst 2024; 34:2450003. [PMID: 37964570 DOI: 10.1142/s0129065724500035] [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: 11/16/2023]
Abstract
To avoid traffic accidents, monitoring the driver's electroencephalogram (EEG) signals to assess drowsiness is an effective solution. However, aggregating the personal data of these drivers may lead to insufficient data usage and pose a risk of privacy breaches. To address these issues, a framework called Group Federated Learning (Group-FL) for large-scale driver drowsiness detection is proposed, which can efficiently utilize diverse client data while protecting privacy. First, by arranging the clients into different levels of groups and gradually aggregating their model parameters from low-level groups to high-level groups, communication and time costs are reduced. In addition, to solve the problem of notable variations in EEG signals among different clients, a global-personalized deep neural network is designed. The global model extracts shared features from various clients, while the personalized model extracts fine-grained features from each client and outputs classification results. Finally, to address special issues such as scale/category imbalance and data pollution, three checking modules are designed for adjusting grouping, evaluating client data, and effectively applying personalized models. Through extensive experimentation, the effectiveness of each component within the framework was validated, and a mean accuracy, F1-score, and Area Under Curve (AUC) of 81.0%, 82.0%, and 87.9% was achieved, respectively, on a publicly available dataset comprising 11 subjects.
Collapse
Affiliation(s)
- Xinyuan Chen
- School of Information Science and Engineering, Shandong Normal University, Jinan 250014, P. R. China
| | - Yi Niu
- School of Information Science and Engineering, Shandong Normal University, Jinan 250014, P. R. China
| | - Yanna Zhao
- School of Information Science and Engineering, Shandong Normal University, Jinan 250014, P. R. China
| | - Xue Qin
- School of Information Science and Engineering, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
34
|
Shekar N, Vuong P, Kaur P. Analysing potent biomarkers along phytochemicals for breast cancer therapy: an in silico approach. Breast Cancer Res Treat 2024; 203:29-47. [PMID: 37726449 PMCID: PMC10771382 DOI: 10.1007/s10549-023-07107-7] [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: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE This research focused on the identification of herbal compounds as potential anti-cancer drugs, especially for breast cancer, that involved the recognition of Notch downstream targets NOTCH proteins (1-4) specifically expressed in breast tumours as biomarkers for prognosis, along with P53 tumour antigens, that were used as comparisons to check the sensitivity of the herbal bio-compounds. METHODS After investigating phytochemical candidates, we employed an approach for computer-aided drug design and analysis to find strong breast cancer inhibitors. The present study utilized in silico analyses and protein docking techniques to characterize and rank selected bio-compounds for their efficiency in oncogenic inhibition for use in precise carcinomic cell growth control. RESULTS Several of the identified phytocompounds found in herbs followed Lipinski's Rule of Five and could be further investigated as potential medicinal molecules. Based on the Vina score obtained after the docking process, the active compound Epigallocatechin gallate in green tea with NOTCH (1-4) and P53 proteins showed promising results for future drug repurposing. The stiffness and binding stability of green tea pharmacological complexes were further elucidated by the molecular dynamic simulations carried out for the highest scoring phytochemical ligand complex. CONCLUSION The target-ligand complex of green tea active compound Epigallocatechin gallate with NOTCH (1-4) had the potential to become potent anti-breast cancer therapeutic candidates following further research involving wet-lab experiments.
Collapse
Affiliation(s)
- Nivruthi Shekar
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Paton Vuong
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia
| | - Parwinder Kaur
- UWA School of Agriculture and Environment, University of Western Australia, 35-Stirling Highway, Perth, WA, 6009, Australia.
| |
Collapse
|
35
|
Almufareh MF, Tariq N, Humayun M, Almas B. A Federated Learning Approach to Breast Cancer Prediction in a Collaborative Learning Framework. Healthcare (Basel) 2023; 11:3185. [PMID: 38132075 PMCID: PMC10743267 DOI: 10.3390/healthcare11243185] [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: 10/05/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Breast cancer continues to pose a substantial worldwide public health concern, necessitating the use of sophisticated diagnostic methods to enable timely identification and management. The present research utilizes an iterative methodology for collaborative learning, using Deep Neural Networks (DNN) to construct a breast cancer detection model with a high level of accuracy. By leveraging Federated Learning (FL), this collaborative framework effectively utilizes the combined knowledge and data assets of several healthcare organizations while ensuring the protection of patient privacy and data security. The model described in this study showcases significant progress in the field of breast cancer diagnoses, with a maximum accuracy rate of 97.54%, precision of 96.5%, and recall of 98.0%, by using an optimum feature selection technique. Data augmentation approaches play a crucial role in decreasing loss and improving model performance. Significantly, the F1-Score, a comprehensive metric for evaluating performance, turns out to be 97%. This study signifies a notable advancement in the field of breast cancer screening, fostering hope for improved patient outcomes via increased accuracy and reliability. This study highlights the potential impact of collaborative learning, namely, in the field of FL, in transforming breast cancer detection. The incorporation of privacy considerations and the use of diverse data sources contribute to the advancement of early detection and the treatment of breast cancer, hence yielding significant benefits for patients on a global scale.
Collapse
Affiliation(s)
- Maram Fahaad Almufareh
- Department of Information Systems, College of Computer and Information Sciences, Jouf University, Al Jouf 72311, Saudi Arabia;
| | - Noshina Tariq
- Department of Avionics Engineering, Air University, Islamabad 44000, Pakistan;
| | - Mamoona Humayun
- Department of Information Systems, College of Computer and Information Sciences, Jouf University, Al Jouf 72311, Saudi Arabia;
| | - Bushra Almas
- Institute of Information Technology, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| |
Collapse
|
36
|
Tao S, Liu H, Sun C, Ji H, Ji G, Han Z, Gao R, Ma J, Ma R, Chen Y, Fu S, Wang Y, Sun Y, Rong Y, Zhang X, Zhou G, Sun H. Collaborative and privacy-preserving retired battery sorting for profitable direct recycling via federated machine learning. Nat Commun 2023; 14:8032. [PMID: 38052823 DOI: 10.1038/s41467-023-43883-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023] Open
Abstract
Unsorted retired batteries with varied cathode materials hinder the adoption of direct recycling due to their cathode-specific nature. The surge in retired batteries necessitates precise sorting for effective direct recycling, but challenges arise from varying operational histories, diverse manufacturers, and data privacy concerns of recycling collaborators (data owners). Here we show, from a unique dataset of 130 lithium-ion batteries spanning 5 cathode materials and 7 manufacturers, a federated machine learning approach can classify these retired batteries without relying on past operational data, safeguarding the data privacy of recycling collaborators. By utilizing the features extracted from the end-of-life charge-discharge cycle, our model exhibits 1% and 3% cathode sorting errors under homogeneous and heterogeneous battery recycling settings respectively, attributed to our innovative Wasserstein-distance voting strategy. Economically, the proposed method underscores the value of precise battery sorting for a prosperous and sustainable recycling industry. This study heralds a new paradigm of using privacy-sensitive data from diverse sources, facilitating collaborative and privacy-respecting decision-making for distributed systems.
Collapse
Affiliation(s)
- Shengyu Tao
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Haizhou Liu
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Chongbo Sun
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Haocheng Ji
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Guanjun Ji
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Zhiyuan Han
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Runhua Gao
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Jun Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Ruifei Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Yuou Chen
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Shiyi Fu
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Yu Wang
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Yaojie Sun
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Yu Rong
- Tencent AI Lab, Tencent, Shenzhen, China
| | - Xuan Zhang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.
| | - Guangmin Zhou
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.
| | - Hongbin Sun
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.
- Department of Electrical Engineering, Tsinghua University, Beijing, China.
- College of Electrical and Power Engineering, Taiyuan University of Technology, Taiyuan, China.
| |
Collapse
|
37
|
Heudel P, Crochet H, Durand T, Zrounba P, Blay JY. From data strategy to implementation to advance cancer research and cancer care: A French comprehensive cancer center experience. PLOS DIGITAL HEALTH 2023; 2:e0000415. [PMID: 38113207 PMCID: PMC10729983 DOI: 10.1371/journal.pdig.0000415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
In a comprehensive cancer center, effective data strategies are essential to evaluate practices, and outcome, understanding the disease and prognostic factors, identifying disparities in cancer care, and overall developing better treatments. To achieve these goals, the Center Léon Bérard (CLB) considers various data collection strategies, including electronic medical records (EMRs), clinical trial data, and research projects. Advanced data analysis techniques like natural language processing (NLP) can be used to extract and categorize information from these sources to provide a more complete description of patient data. Data sharing is also crucial for collaboration across comprehensive cancer centers, but it must be done securely and in compliance with regulations like GDPR. To ensure data is shared appropriately, CLB should develop clear data sharing policies and share data in a controlled, standardized format like OSIRIS RWD, OMOP and FHIR. The UNICANCER initiative has launched the CONSORE project to support the development of a structured and standardized repository of patient data to improve cancer research and patient outcomes. Real-world data (RWD) studies are vital in cancer research as they provide a comprehensive and accurate picture of patient outcomes and treatment patterns. By incorporating RWD into data collection, analysis, and sharing strategies, comprehensive cancer centers can take a more comprehensive and patient-centered approach to cancer research. In conclusion, comprehensive cancer centers must take an integrated approach to data collection, analysis, and sharing to enhance their understanding of cancer and improve patient outcomes. Leveraging advanced data analytics techniques and developing effective data sharing policies can help cancer centers effectively harness the power of data to drive progress in cancer research.
Collapse
Affiliation(s)
- Pierre Heudel
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Hugo Crochet
- Data and Artificial Intelligence Team, Centre Léon Bérard, Lyon, France
| | - Thierry Durand
- Data protection officer, Centre Léon Bérard, Lyon, France
| | - Philippe Zrounba
- Department of Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- General Director, Centre Léon Bérard, Lyon, France
| |
Collapse
|
38
|
Tavolara TE, Su Z, Gurcan MN, Niazi MKK. One label is all you need: Interpretable AI-enhanced histopathology for oncology. Semin Cancer Biol 2023; 97:70-85. [PMID: 37832751 DOI: 10.1016/j.semcancer.2023.09.006] [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: 10/24/2022] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
Artificial Intelligence (AI)-enhanced histopathology presents unprecedented opportunities to benefit oncology through interpretable methods that require only one overall label per hematoxylin and eosin (H&E) slide with no tissue-level annotations. We present a structured review of these methods organized by their degree of verifiability and by commonly recurring application areas in oncological characterization. First, we discuss morphological markers (tumor presence/absence, metastases, subtypes, grades) in which AI-identified regions of interest (ROIs) within whole slide images (WSIs) verifiably overlap with pathologist-identified ROIs. Second, we discuss molecular markers (gene expression, molecular subtyping) that are not verified via H&E but rather based on overlap with positive regions on adjacent tissue. Third, we discuss genetic markers (mutations, mutational burden, microsatellite instability, chromosomal instability) that current technologies cannot verify if AI methods spatially resolve specific genetic alterations. Fourth, we discuss the direct prediction of survival to which AI-identified histopathological features quantitatively correlate but are nonetheless not mechanistically verifiable. Finally, we discuss in detail several opportunities and challenges for these one-label-per-slide methods within oncology. Opportunities include reducing the cost of research and clinical care, reducing the workload of clinicians, personalized medicine, and unlocking the full potential of histopathology through new imaging-based biomarkers. Current challenges include explainability and interpretability, validation via adjacent tissue sections, reproducibility, data availability, computational needs, data requirements, domain adaptability, external validation, dataset imbalances, and finally commercialization and clinical potential. Ultimately, the relative ease and minimum upfront cost with which relevant data can be collected in addition to the plethora of available AI methods for outcome-driven analysis will surmount these current limitations and achieve the innumerable opportunities associated with AI-driven histopathology for the benefit of oncology.
Collapse
Affiliation(s)
- Thomas E Tavolara
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ziyu Su
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Metin N Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - M Khalid Khan Niazi
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
39
|
Fleming N. How Paris is becoming a happy home for health-technology start-up companies. Nature 2023:10.1038/d41586-023-03490-9. [PMID: 37938332 DOI: 10.1038/d41586-023-03490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
|
40
|
Stankevičiūtė K, Woillard JB, Peck RW, Marquet P, van der Schaar M. Bridging the Worlds of Pharmacometrics and Machine Learning. Clin Pharmacokinet 2023; 62:1551-1565. [PMID: 37803104 DOI: 10.1007/s40262-023-01310-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/08/2023]
Abstract
Precision medicine requires individualized modeling of disease and drug dynamics, with machine learning-based computational techniques gaining increasing popularity. The complexity of either field, however, makes current pharmacological problems opaque to machine learning practitioners, and state-of-the-art machine learning methods inaccessible to pharmacometricians. To help bridge the two worlds, we provide an introduction to current problems and techniques in pharmacometrics that ranges from pharmacokinetic and pharmacodynamic modeling to pharmacometric simulations, model-informed precision dosing, and systems pharmacology, and review some of the machine learning approaches to address them. We hope this would facilitate collaboration between experts, with complementary strengths of principled pharmacometric modeling and flexibility of machine learning leading to synergistic effects in pharmacological applications.
Collapse
Affiliation(s)
- Kamilė Stankevičiūtė
- Department of Computer Science and Technology, University of Cambridge, 15 JJ Thomson Avenue, Cambridge, CB3 0FD, UK
| | - Jean-Baptiste Woillard
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France.
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France.
| | - Richard W Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Pharma Research and Development, Roche Innovation Center, Basel, Switzerland
| | - Pierre Marquet
- INSERM U1248 P&T, University of Limoges, 2 rue du Pr Descottes, 87000, Limoges, France
- Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France
| | - Mihaela van der Schaar
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| |
Collapse
|
41
|
Jiang J, Chao WL, Cao T, Culp S, Napoléon B, El-Dika S, Machicado JD, Pannala R, Mok S, Luthra AK, Akshintala VS, Muniraj T, Krishna SG. Improving Pancreatic Cyst Management: Artificial Intelligence-Powered Prediction of Advanced Neoplasms through Endoscopic Ultrasound-Guided Confocal Endomicroscopy. Biomimetics (Basel) 2023; 8:496. [PMID: 37887627 PMCID: PMC10604893 DOI: 10.3390/biomimetics8060496] [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: 08/02/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Despite the increasing rate of detection of incidental pancreatic cystic lesions (PCLs), current standard-of-care methods for their diagnosis and risk stratification remain inadequate. Intraductal papillary mucinous neoplasms (IPMNs) are the most prevalent PCLs. The existing modalities, including endoscopic ultrasound and cyst fluid analysis, only achieve accuracy rates of 65-75% in identifying carcinoma or high-grade dysplasia in IPMNs. Furthermore, surgical resection of PCLs reveals that up to half exhibit only low-grade dysplastic changes or benign neoplasms. To reduce unnecessary and high-risk pancreatic surgeries, more precise diagnostic techniques are necessary. A promising approach involves integrating existing data, such as clinical features, cyst morphology, and data from cyst fluid analysis, with confocal endomicroscopy and radiomics to enhance the prediction of advanced neoplasms in PCLs. Artificial intelligence and machine learning modalities can play a crucial role in achieving this goal. In this review, we explore current and future techniques to leverage these advanced technologies to improve diagnostic accuracy in the context of PCLs.
Collapse
Affiliation(s)
- Joanna Jiang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei-Lun Chao
- Department of Computer Science and Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Troy Cao
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Stacey Culp
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Bertrand Napoléon
- Department of Gastroenterology, Jean Mermoz Private Hospital, 69008 Lyon, France
| | - Samer El-Dika
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA 94305, USA
| | - Jorge D. Machicado
- Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rahul Pannala
- Division of Gastroenterology and Hepatology, Mayo Clinic Arizona, Phoenix, AZ 85054, USA
| | - Shaffer Mok
- Division of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Anjuli K. Luthra
- Division of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Venkata S. Akshintala
- Division of Gastroenterology, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | - Thiruvengadam Muniraj
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
42
|
Blatter TU, Witte H, Fasquelle-Lopez J, Theodoros Naka C, Raisaro JL, Leichtle AB. The BioRef Infrastructure, a Framework for Real-Time, Federated, Privacy-Preserving, and Personalized Reference Intervals: Design, Development, and Application. J Med Internet Res 2023; 25:e47254. [PMID: 37851984 PMCID: PMC10620636 DOI: 10.2196/47254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Reference intervals (RIs) for patient test results are in standard use across many medical disciplines, allowing physicians to identify measurements indicating potentially pathological states with relative ease. The process of inferring cohort-specific RIs is, however, often ignored because of the high costs and cumbersome efforts associated with it. Sophisticated analysis tools are required to automatically infer relevant and locally specific RIs directly from routine laboratory data. These tools would effectively connect clinical laboratory databases to physicians and provide personalized target ranges for the respective cohort population. OBJECTIVE This study aims to describe the BioRef infrastructure, a multicentric governance and IT framework for the estimation and assessment of patient group-specific RIs from routine clinical laboratory data using an innovative decentralized data-sharing approach and a sophisticated, clinically oriented graphical user interface for data analysis. METHODS A common governance agreement and interoperability standards have been established, allowing the harmonization of multidimensional laboratory measurements from multiple clinical databases into a unified "big data" resource. International coding systems, such as the International Classification of Diseases, Tenth Revision (ICD-10); unique identifiers for medical devices from the Global Unique Device Identification Database; type identifiers from the Global Medical Device Nomenclature; and a universal transfer logic, such as the Resource Description Framework (RDF), are used to align the routine laboratory data of each data provider for use within the BioRef framework. With a decentralized data-sharing approach, the BioRef data can be evaluated by end users from each cohort site following a strict "no copy, no move" principle, that is, only data aggregates for the intercohort analysis of target ranges are exchanged. RESULTS The TI4Health distributed and secure analytics system was used to implement the proposed federated and privacy-preserving approach and comply with the limitations applied to sensitive patient data. Under the BioRef interoperability consensus, clinical partners enable the computation of RIs via the TI4Health graphical user interface for query without exposing the underlying raw data. The interface was developed for use by physicians and clinical laboratory specialists and allows intuitive and interactive data stratification by patient factors (age, sex, and personal medical history) as well as laboratory analysis determinants (device, analyzer, and test kit identifier). This consolidated effort enables the creation of extremely detailed and patient group-specific queries, allowing the generation of individualized, covariate-adjusted RIs on the fly. CONCLUSIONS With the BioRef-TI4Health infrastructure, a framework for clinical physicians and researchers to define precise RIs immediately in a convenient, privacy-preserving, and reproducible manner has been implemented, promoting a vital part of practicing precision medicine while streamlining compliance and avoiding transfers of raw patient data. This new approach can provide a crucial update on RIs and improve patient care for personalized medicine.
Collapse
Affiliation(s)
- Tobias Ueli Blatter
- University Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Harald Witte
- University Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
| | | | - Christos Theodoros Naka
- University Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
- Laboratory of Biometry, University of Thessaly, Volos, Greece
| | - Jean Louis Raisaro
- Biomedical Data Science Center, University Hospital Lausanne, Lausanne, Switzerland
| | - Alexander Benedikt Leichtle
- University Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
- Center for Artificial Intelligence in Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Ng FYC, Thirunavukarasu AJ, Cheng H, Tan TF, Gutierrez L, Lan Y, Ong JCL, Chong YS, Ngiam KY, Ho D, Wong TY, Kwek K, Doshi-Velez F, Lucey C, Coffman T, Ting DSW. Artificial intelligence education: An evidence-based medicine approach for consumers, translators, and developers. Cell Rep Med 2023; 4:101230. [PMID: 37852174 PMCID: PMC10591047 DOI: 10.1016/j.xcrm.2023.101230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
Current and future healthcare professionals are generally not trained to cope with the proliferation of artificial intelligence (AI) technology in healthcare. To design a curriculum that caters to variable baseline knowledge and skills, clinicians may be conceptualized as "consumers", "translators", or "developers". The changes required of medical education because of AI innovation are linked to those brought about by evidence-based medicine (EBM). We outline a core curriculum for AI education of future consumers, translators, and developers, emphasizing the links between AI and EBM, with suggestions for how teaching may be integrated into existing curricula. We consider the key barriers to implementation of AI in the medical curriculum: time, resources, variable interest, and knowledge retention. By improving AI literacy rates and fostering a translator- and developer-enriched workforce, innovation may be accelerated for the benefit of patients and practitioners.
Collapse
Affiliation(s)
- Faye Yu Ci Ng
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun James Thirunavukarasu
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore; University of Cambridge School of Clinical Medicine, Cambridge, UK; Oxford University Clinical Academic Graduate School, University of Oxford, Oxford, UK
| | - Haoran Cheng
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore; Rollins School of Public Health, Emory University, Atlanta, GA, USA; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ting Fang Tan
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore
| | - Laura Gutierrez
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore
| | - Yanyan Lan
- Institute for AI Industry Research (AIR), Tsinghua University, Beijing, China
| | | | - Yap Seng Chong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Dean's Office, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kee Yuan Ngiam
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Biomedical Engineering, School of Engineering, National University of Singapore, Singapore, Singapore
| | - Dean Ho
- Biomedical Engineering, School of Engineering, National University of Singapore, Singapore, Singapore; Insitute for Digital Medicine (WisDM), N.1 Institute for Health, National University of Singapore, Singapore, Singapore; Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Tien Yin Wong
- Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Kenneth Kwek
- Chief Executive Office, Singapore General Hospital, SingHealth, Singapore, Singapore
| | - Finale Doshi-Velez
- Harvard Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Catherine Lucey
- Executive Vice Chancellor and Provost Office, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Coffman
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Daniel Shu Wei Ting
- Artificial Intelligence and Digital Innovation, Singapore Eye Research Institute, Singapore National Eye Center, Singapore Health Service, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore; Byers Eye Institute, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
44
|
Fisher TB, Saini G, Ts R, Krishnamurthy J, Bhattarai S, Callagy G, Webber M, Janssen EAM, Kong J, Aneja R. Digital image analysis and machine learning-assisted prediction of neoadjuvant chemotherapy response in triple-negative breast cancer. RESEARCH SQUARE 2023:rs.3.rs-3243195. [PMID: 37645881 PMCID: PMC10462230 DOI: 10.21203/rs.3.rs-3243195/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Background Pathological complete response (pCR) is associated with favorable prognosis in patients with triple-negative breast cancer (TNBC). However, only 30-40% of TNBC patients treated with neoadjuvant chemotherapy (NAC) show pCR, while the remaining 60-70% show residual disease (RD). The role of the tumor microenvironment (TME) in NAC response in patients with TNBC remains unclear. In this study, we developed a machine learning-based two-step pipeline to distinguish between various histological components in hematoxylin and eosin (H&E)-stained whole slide images (WSIs) of TNBC tissue biopsies and to identify histological features that can predict NAC response. Methods H&E-stained WSIs of treatment-naïve biopsies from 85 patients (51 with pCR and 34 with RD) were separated through a stratified 8-fold cross validation strategy for the first step and leave one out cross validation strategy for the second step. A tile-level histology label prediction pipeline and four machine learning classifiers were used to analyze 468,043 tiles of WSIs. The best-trained classifier used 55 texture features from each tile to produce a probability profile during testing. The predicted histology classes were used to generate a histology classification map of the spatial distributions of different tissue regions. A patient-level NAC response prediction pipeline was trained with features derived from paired histology classification maps. The top graph-based features capturing the relevant spatial information across the different histological classes were provided to the radial basis function kernel support vector machine (rbfSVM) classifier for NAC treatment response prediction. Results The tile-level prediction pipeline achieved 86.72% accuracy for histology class classification, while the patient-level pipeline achieved 83.53% NAC response (pCR vs. RD) prediction accuracy. The histological class pairs with the strongest NAC response predictive ability were tumor and tumor tumor-infiltrating lymphocytes for pCR and microvessel density and polyploid giant cancer cells for RD. Conclusion Our machine learning pipeline can robustly identify clinically relevant histological classes that predict NAC response in TNBC patients and may help guide patient selection for NAC treatment.
Collapse
Affiliation(s)
| | | | - Rekha Ts
- JSSAHER (JSS Academy of Higher Education and Research) Medical College
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Fu X, Sahai E, Wilkins A. Application of digital pathology-based advanced analytics of tumour microenvironment organisation to predict prognosis and therapeutic response. J Pathol 2023; 260:578-591. [PMID: 37551703 PMCID: PMC10952145 DOI: 10.1002/path.6153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 08/09/2023]
Abstract
In recent years, the application of advanced analytics, especially artificial intelligence (AI), to digital H&E images, and other histological image types, has begun to radically change how histological images are used in the clinic. Alongside the recognition that the tumour microenvironment (TME) has a profound impact on tumour phenotype, the technical development of highly multiplexed immunofluorescence platforms has enhanced the biological complexity that can be captured in the TME with high precision. AI has an increasingly powerful role in the recognition and quantitation of image features and the association of such features with clinically important outcomes, as occurs in distinct stages in conventional machine learning. Deep-learning algorithms are able to elucidate TME patterns inherent in the input data with minimum levels of human intelligence and, hence, have the potential to achieve clinically relevant predictions and discovery of important TME features. Furthermore, the diverse repertoire of deep-learning algorithms able to interrogate TME patterns extends beyond convolutional neural networks to include attention-based models, graph neural networks, and multimodal models. To date, AI models have largely been evaluated retrospectively, outside the well-established rigour of prospective clinical trials, in part because traditional clinical trial methodology may not always be suitable for the assessment of AI technology. However, to enable digital pathology-based advanced analytics to meaningfully impact clinical care, specific measures of 'added benefit' to the current standard of care and validation in a prospective setting are important. This will need to be accompanied by adequate measures of explainability and interpretability. Despite such challenges, the combination of expanding datasets, increased computational power, and the possibility of integration of pre-clinical experimental insights into model development means there is exciting potential for the future progress of these AI applications. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xiao Fu
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| | - Erik Sahai
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Anna Wilkins
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Division of Radiotherapy and ImagingInstitute of Cancer ResearchLondonUK
- Royal Marsden Hospitals NHS TrustLondonUK
| |
Collapse
|
46
|
Lee M. Recent Advancements in Deep Learning Using Whole Slide Imaging for Cancer Prognosis. Bioengineering (Basel) 2023; 10:897. [PMID: 37627783 PMCID: PMC10451210 DOI: 10.3390/bioengineering10080897] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
This review furnishes an exhaustive analysis of the latest advancements in deep learning techniques applied to whole slide images (WSIs) in the context of cancer prognosis, focusing specifically on publications from 2019 through 2023. The swiftly maturing field of deep learning, in combination with the burgeoning availability of WSIs, manifests significant potential in revolutionizing the predictive modeling of cancer prognosis. In light of the swift evolution and profound complexity of the field, it is essential to systematically review contemporary methodologies and critically appraise their ramifications. This review elucidates the prevailing landscape of this intersection, cataloging major developments, evaluating their strengths and weaknesses, and providing discerning insights into prospective directions. In this paper, a comprehensive overview of the field aims to be presented, which can serve as a critical resource for researchers and clinicians, ultimately enhancing the quality of cancer care outcomes. This review's findings accentuate the need for ongoing scrutiny of recent studies in this rapidly progressing field to discern patterns, understand breakthroughs, and navigate future research trajectories.
Collapse
Affiliation(s)
- Minhyeok Lee
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
47
|
Karargyris A, Umeton R, Sheller MJ, Aristizabal A, George J, Wuest A, Pati S, Kassem H, Zenk M, Baid U, Narayana Moorthy P, Chowdhury A, Guo J, Nalawade S, Rosenthal J, Kanter D, Xenochristou M, Beutel DJ, Chung V, Bergquist T, Eddy J, Abid A, Tunstall L, Sanseviero O, Dimitriadis D, Qian Y, Xu X, Liu Y, Goh RSM, Bala S, Bittorf V, Reddy Puchala S, Ricciuti B, Samineni S, Sengupta E, Chaudhari A, Coleman C, Desinghu B, Diamos G, Dutta D, Feddema D, Fursin G, Huang X, Kashyap S, Lane N, Mallick I, Mascagni P, Mehta V, Ferro Moraes C, Natarajan V, Nikolov N, Padoy N, Pekhimenko G, Reddi VJ, Reina GA, Ribalta P, Singh A, Thiagarajan JJ, Albrecht J, Wolf T, Miller G, Fu H, Shah P, Xu D, Yadav P, Talby D, Awad MM, Howard JP, Rosenthal M, Marchionni L, Loda M, Johnson JM, Bakas S, Mattson P. Federated benchmarking of medical artificial intelligence with MedPerf. NAT MACH INTELL 2023; 5:799-810. [PMID: 38706981 PMCID: PMC11068064 DOI: 10.1038/s42256-023-00652-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 04/06/2023] [Indexed: 05/07/2024]
Abstract
Medical artificial intelligence (AI) has tremendous potential to advance healthcare by supporting and contributing to the evidence-based practice of medicine, personalizing patient treatment, reducing costs, and improving both healthcare provider and patient experience. Unlocking this potential requires systematic, quantitative evaluation of the performance of medical AI models on large-scale, heterogeneous data capturing diverse patient populations. Here, to meet this need, we introduce MedPerf, an open platform for benchmarking AI models in the medical domain. MedPerf focuses on enabling federated evaluation of AI models, by securely distributing them to different facilities, such as healthcare organizations. This process of bringing the model to the data empowers each facility to assess and verify the performance of AI models in an efficient and human-supervised process, while prioritizing privacy. We describe the current challenges healthcare and AI communities face, the need for an open platform, the design philosophy of MedPerf, its current implementation status and real-world deployment, our roadmap and, importantly, the use of MedPerf with multiple international institutions within cloud-based technology and on-premises scenarios. Finally, we welcome new contributions by researchers and organizations to further strengthen MedPerf as an open benchmarking platform.
Collapse
Affiliation(s)
- Alexandros Karargyris
- IHU Strasbourg, Strasbourg, France
- University of Strasbourg, Strasbourg, France
- These authors contributed equally: Alexandros Karargyris, Renato Umeton, Micah J. Sheller
| | - Renato Umeton
- Dana-Farber Cancer Institute, Boston, MA, USA
- Weill Cornell Medicine, New York, NY, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
- These authors contributed equally: Alexandros Karargyris, Renato Umeton, Micah J. Sheller
| | - Micah J. Sheller
- Intel, Santa Clara, CA, USA
- These authors contributed equally: Alexandros Karargyris, Renato Umeton, Micah J. Sheller
| | | | | | - Anna Wuest
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sarthak Pati
- Perelman School of Medicine, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Maximilian Zenk
- German Cancer Research Center, Heidelberg, Germany
- University of Heidelberg, Heidelberg, Germany
| | - Ujjwal Baid
- Perelman School of Medicine, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Junyi Guo
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Jacob Rosenthal
- Dana-Farber Cancer Institute, Boston, MA, USA
- Weill Cornell Medicine, New York, NY, USA
| | | | | | - Daniel J. Beutel
- University of Cambridge, Cambridge, UK
- Flower Labs, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Akshay Chaudhari
- Stanford University, Stanford, CA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | - Nicholas Lane
- University of Cambridge, Cambridge, UK
- Flower Labs, Hamburg, Germany
| | | | | | | | | | - Pietro Mascagni
- IHU Strasbourg, Strasbourg, France
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | | | | | - Nicolas Padoy
- IHU Strasbourg, Strasbourg, France
- University of Strasbourg, Strasbourg, France
| | - Gennady Pekhimenko
- University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | | | | | | | - Abhishek Singh
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | - Mark M. Awad
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeremy P. Howard
- fast.ai, San Francisco, CA, USA
- University of Queensland, Brisbane, Queensland, Australia
| | - Michael Rosenthal
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Massimo Loda
- Dana-Farber Cancer Institute, Boston, MA, USA
- Weill Cornell Medicine, New York, NY, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Spyridon Bakas
- Perelman School of Medicine, Philadelphia, PA, USA
- University of Pennsylvania, Philadelphia, PA, USA
- These authors jointly supervised this work: Spyridon Bakas, Peter Mattson
| | - Peter Mattson
- MLCommons, San Francisco, CA, USA
- Google, Mountain View, CA, USA
- These authors jointly supervised this work: Spyridon Bakas, Peter Mattson
| |
Collapse
|
48
|
Saillard C, Delecourt F, Schmauch B, Moindrot O, Svrcek M, Bardier-Dupas A, Emile JF, Ayadi M, Rebours V, de Mestier L, Hammel P, Neuzillet C, Bachet JB, Iovanna J, Dusetti N, Blum Y, Richard M, Kermezli Y, Paradis V, Zaslavskiy M, Courtiol P, Kamoun A, Nicolle R, Cros J. Pacpaint: a histology-based deep learning model uncovers the extensive intratumor molecular heterogeneity of pancreatic adenocarcinoma. Nat Commun 2023; 14:3459. [PMID: 37311751 PMCID: PMC10264377 DOI: 10.1038/s41467-023-39026-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Two tumor (Classical/Basal) and stroma (Inactive/active) subtypes of Pancreatic adenocarcinoma (PDAC) with prognostic and theragnostic implications have been described. These molecular subtypes were defined by RNAseq, a costly technique sensitive to sample quality and cellularity, not used in routine practice. To allow rapid PDAC molecular subtyping and study PDAC heterogeneity, we develop PACpAInt, a multi-step deep learning model. PACpAInt is trained on a multicentric cohort (n = 202) and validated on 4 independent cohorts including biopsies (surgical cohorts n = 148; 97; 126 / biopsy cohort n = 25), all with transcriptomic data (n = 598) to predict tumor tissue, tumor cells from stroma, and their transcriptomic molecular subtypes, either at the whole slide or tile level (112 µm squares). PACpAInt correctly predicts tumor subtypes at the whole slide level on surgical and biopsies specimens and independently predicts survival. PACpAInt highlights the presence of a minor aggressive Basal contingent that negatively impacts survival in 39% of RNA-defined classical cases. Tile-level analysis ( > 6 millions) redefines PDAC microheterogeneity showing codependencies in the distribution of tumor and stroma subtypes, and demonstrates that, in addition to the Classical and Basal tumors, there are Hybrid tumors that combine the latter subtypes, and Intermediate tumors that may represent a transition state during PDAC evolution.
Collapse
Affiliation(s)
| | - Flore Delecourt
- Université Paris Cité, Dpt of Pathology - FHU MOSAIC, Beaujon Hospital, INSERM U1149, Clichy, France
| | | | | | - Magali Svrcek
- Dpt of Pathology, Saint-Antoine Hospital - Sorbonne Universités, Paris, France
| | | | - Jean Francois Emile
- Dpt of Pathology, Ambroise Paré Hospital - Université Saint Quentin en Yvelines, Paris, France
| | - Mira Ayadi
- Integragen, Genomic Services & Precision Medicine, Paris, France
| | - Vinciane Rebours
- Université Paris Cité, Dpt of Pancreatology - FHU MOSAIC, Beaujon Hospital, INSERM U1149, Clichy, France
| | - Louis de Mestier
- Université Paris Cité, Dpt of Pancreatology - FHU MOSAIC, Beaujon Hospital, INSERM U1149, Clichy, France
| | - Pascal Hammel
- Dpt of Medical oncology, Paul Brousse Hospital, Villejuif, France
| | | | - Jean Baptiste Bachet
- Dpt of Gastroenterology, Pitié-Salpêtrière Hospital - Sorbonne Universités, Paris, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Institut Paoli-Calmettes, Aix Marseille Université, CNRS UMR 7258, Marseille, France
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, Institut Paoli-Calmettes, Aix Marseille Université, CNRS UMR 7258, Marseille, France
| | - Yuna Blum
- Institut Génétique et Développement de Rennes (IGDR), CNRS, Université de Rennes 1, UMR 6290, Rennes, France
| | - Magali Richard
- Techniques de l'Ingénierie Médicale et de la Complexité - Informatique, Mathématiques et Applications Grenoble (TIMC-IMAG), CNRS, Université Grenoble-Alpes, UMR5525, Grenoble, France
| | - Yasmina Kermezli
- Techniques de l'Ingénierie Médicale et de la Complexité - Informatique, Mathématiques et Applications Grenoble (TIMC-IMAG), CNRS, Université Grenoble-Alpes, UMR5525, Grenoble, France
| | - Valerie Paradis
- Université Paris Cité, Dpt of Pathology - FHU MOSAIC, Beaujon Hospital, INSERM U1149, Clichy, France
| | | | | | | | - Remy Nicolle
- Université Paris Cité, FHU MOSAIC, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018, Paris, France
| | - Jerome Cros
- Université Paris Cité, Dpt of Pathology - FHU MOSAIC, Beaujon Hospital, INSERM U1149, Clichy, France.
| |
Collapse
|
49
|
Vert JP. How will generative AI disrupt data science in drug discovery? Nat Biotechnol 2023:10.1038/s41587-023-01789-6. [PMID: 37156917 DOI: 10.1038/s41587-023-01789-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
|
50
|
Petinrin OO, Saeed F, Toseef M, Liu Z, Basurra S, Muyide IO, Li X, Lin Q, Wong KC. Machine learning in metastatic cancer research: Potentials, possibilities, and prospects. Comput Struct Biotechnol J 2023; 21:2454-2470. [PMID: 37077177 PMCID: PMC10106342 DOI: 10.1016/j.csbj.2023.03.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer has received extensive recognition for its high mortality rate, with metastatic cancer being the top cause of cancer-related deaths. Metastatic cancer involves the spread of the primary tumor to other body organs. As much as the early detection of cancer is essential, the timely detection of metastasis, the identification of biomarkers, and treatment choice are valuable for improving the quality of life for metastatic cancer patients. This study reviews the existing studies on classical machine learning (ML) and deep learning (DL) in metastatic cancer research. Since the majority of metastatic cancer research data are collected in the formats of PET/CT and MRI image data, deep learning techniques are heavily involved. However, its black-box nature and expensive computational cost are notable concerns. Furthermore, existing models could be overestimated for their generality due to the non-diverse population in clinical trial datasets. Therefore, research gaps are itemized; follow-up studies should be carried out on metastatic cancer using machine learning and deep learning tools with data in a symmetric manner.
Collapse
Affiliation(s)
| | - Faisal Saeed
- DAAI Research Group, Department of Computing and Data Science, School of Computing and Digital Technology, Birmingham City University, Birmingham B4 7XG, UK
| | - Muhammad Toseef
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Zhe Liu
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| | - Shadi Basurra
- DAAI Research Group, Department of Computing and Data Science, School of Computing and Digital Technology, Birmingham City University, Birmingham B4 7XG, UK
| | | | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Qiuzhen Lin
- School of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
- Hong Kong Institute for Data Science, City University of Hong Kong, Kowloon Tong, Kowloon, Hong Kong SAR
| |
Collapse
|