1
|
Timilsina M, Buosi S, Razzaq MA, Haque R, Judge C, Curry E. Harmonizing foundation models in healthcare: A comprehensive survey of their roles, relationships, and impact in artificial intelligence's advancing terrain. Comput Biol Med 2025; 189:109925. [PMID: 40081208 DOI: 10.1016/j.compbiomed.2025.109925] [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: 10/30/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
The lightning development of artificial intelligence (AI) has revolutionized healthcare, helping significant improvements in various applications. This paper provides a comprehensive review of foundation models in healthcare, highlighting their transformative potential in areas such as diagnostics, personalized treatment, and operational efficiency. We argue the key capabilities of these models, including their ability to process diverse data types such as medical images, clinical notes, and structured health records. Regardless their assurance, difficulties remain, including data privacy concerns, bias in AI algorithms, and the need for extensive computational resources. Our analysis identifies emerging trends and future directions, emphasizing the importance of ethical AI deployment, improved interoperability over healthcare systems, and the development of more robust, domain-specific models. Future research should focus on enhancing model interpretability, ensuring equitable access, and fostering collaboration between AI developers and healthcare professionals to maximize the advantages of these technologies.
Collapse
Affiliation(s)
- Mohan Timilsina
- Data Science Institute, Insight Centre for Data Analytics, University of Galway, Ireland.
| | - Samuele Buosi
- Data Science Institute, Insight Centre for Data Analytics, University of Galway, Ireland.
| | - Muhammad Asif Razzaq
- Data Science Institute, Insight Centre for Data Analytics, University of Galway, Ireland.
| | - Rafiqul Haque
- Data Science Institute, Insight Centre for Data Analytics, University of Galway, Ireland.
| | - Conor Judge
- College of Medicine, Nursing, Health Sciences, University of Galway, Ireland.
| | - Edward Curry
- Data Science Institute, Insight Centre for Data Analytics, University of Galway, Ireland.
| |
Collapse
|
2
|
Kong X, Shi J, Sun D, Cheng L, Wu C, Jiang Z, Zheng Y, Wang W, Wu H. A deep-learning model for predicting tyrosine kinase inhibitor response from histology in gastrointestinal stromal tumor. J Pathol 2025; 265:462-471. [PMID: 39950223 DOI: 10.1002/path.6399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/01/2024] [Accepted: 01/06/2025] [Indexed: 03/06/2025]
Abstract
Over 90% of gastrointestinal stromal tumors (GISTs) harbor mutations in KIT or PDGFRA that can predict response to tyrosine kinase inhibitor (TKI) therapies, as recommended by NCCN (National Comprehensive Cancer Network) guidelines. However, gene sequencing for mutation testing is expensive and time-consuming and is susceptible to a variety of preanalytical factors. To overcome the challenges associated with genetic screening by sequencing, in the current study we developed an artificial intelligence-based deep-learning (DL) model that uses convolutional neural networks (CNN) to analyze digitized hematoxylin and eosin staining in tumor histological sections to predict potential response to imatinib or avapritinib treatment in GIST patients. Assessment with an independent testing set showed that our DL model could predict imatinib sensitivity with an area under the curve (AUC) of 0.902 in case-wise analysis and 0.807 in slide-wise analysis. Case-level AUCs for predicting imatinib-dose-adjustment cases, avapritinib-sensitive cases, and wildtype GISTs were 0.920, 0.958, and 0.776, respectively, while slide-level AUCs for these respective groups were 0.714, 0.922, and 0.886, respectively. Our model showed comparable or better prediction of actual response to TKI than sequencing-based screening (accuracy 0.9286 versus 0.8929; DL model versus sequencing), while predictions of nonresponse to imatinib/avapritinib showed markedly higher accuracy than sequencing (0.7143 versus 0.4286). These results demonstrate the potential of a DL model to improve predictions of treatment response to TKI therapy from histology in GIST patients. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xue Kong
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
| | - Jun Shi
- School of Software, Hefei University of Technology, Hefei, PR China
| | - Dongdong Sun
- School of Computer Science and Information Engineering, Hefei University of Technology, Hefei, PR China
| | - Lanqing Cheng
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
| | - Can Wu
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
| | - Zhiguo Jiang
- Image Processing Center, School of Astronautics, Beihang University, Beijing, PR China
| | - Yushan Zheng
- School of Engineering Medicine, Beijing Advanced Innovation Center on Biomedical Engineering, Beihang University, Beijing, PR China
| | - Wei Wang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
| | - Haibo Wu
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of PR China, Hefei, PR China
| |
Collapse
|
3
|
Hölscher DL, Bülow RD. Decoding pathology: the role of computational pathology in research and diagnostics. Pflugers Arch 2025; 477:555-570. [PMID: 39095655 DOI: 10.1007/s00424-024-03002-2] [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: 04/18/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Traditional histopathology, characterized by manual quantifications and assessments, faces challenges such as low-throughput and inter-observer variability that hinder the introduction of precision medicine in pathology diagnostics and research. The advent of digital pathology allowed the introduction of computational pathology, a discipline that leverages computational methods, especially based on deep learning (DL) techniques, to analyze histopathology specimens. A growing body of research shows impressive performances of DL-based models in pathology for a multitude of tasks, such as mutation prediction, large-scale pathomics analyses, or prognosis prediction. New approaches integrate multimodal data sources and increasingly rely on multi-purpose foundation models. This review provides an introductory overview of advancements in computational pathology and discusses their implications for the future of histopathology in research and diagnostics.
Collapse
Affiliation(s)
- David L Hölscher
- Department for Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Roman D Bülow
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
4
|
Tahmasebzadeh A, Sadeghi M, Naseripour M, Mirshahi R, Ghaderi R. Artificial intelligence and different image modalities in uveal melanoma diagnosis and prognosis: A narrative review. Photodiagnosis Photodyn Ther 2025; 52:104528. [PMID: 39986588 DOI: 10.1016/j.pdpdt.2025.104528] [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: 01/18/2025] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND The most widespread primary intraocular tumor in adults is called uveal melanoma (UM), if detected early enough, it can be curable. Various methods are available to treat UM, but the most commonly used and effective approach is plaque radiotherapy using Iodine-125 and Ruthenium-106. METHOD The authors performed searches to distinguish relevant studies from 2017 to 2024 by three databases (PubMed, Scopus, and Google Scholar). RESULTS Imaging technologies such as ultrasound (US), fundus photography (FP), optical coherent tomography (OCT), fluorescein angiography (FA), and magnetic resonance images (MRI) play a vital role in the diagnosis and prognosis of UM. The present review assessed the power of different image modalities when integrated with artificial intelligence (AI) to diagnose and prognosis of patients affected by UM. CONCLUSION Finally, after reviewing the studies conducted, it was concluded that AI is a developing tool in image analysis and enhances workflows in diagnosis from data and image processing to clinical decisions, improving tailored treatment scenarios, response prediction, and prognostication.
Collapse
Affiliation(s)
- Atefeh Tahmasebzadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, , Iran
| | - Mahdi Sadeghi
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences, Tehran, , Iran.
| | - Masood Naseripour
- Eye Research Center, The Five Senses Institute, Moheb Kowsar Hospital, Iran University of Medical Sciences, Tehran, Iran; Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Reza Mirshahi
- Eye Research Center, The Five Senses Institute, Moheb Kowsar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ghaderi
- Department of Electrical Engineering, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
5
|
Zhou S, Xie Y, Feng X, Li Y, Shen L, Chen Y. Artificial intelligence in gastrointestinal cancer research: Image learning advances and applications. Cancer Lett 2025; 614:217555. [PMID: 39952597 DOI: 10.1016/j.canlet.2025.217555] [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: 12/04/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
With the rapid advancement of artificial intelligence (AI) technologies, including deep learning, large language models, and neural networks, these methodologies are increasingly being developed and integrated into cancer research. Gastrointestinal tumors are characterized by complexity and heterogeneity, posing significant challenges for early detection, diagnostic accuracy, and the development of personalized treatment strategies. The application of AI in digestive oncology has demonstrated its transformative potential. AI not only alleviates the diagnostic burden on clinicians, but it improves tumor screening sensitivity, specificity, and accuracy. Additionally, AI aids the detection of biomarkers such as microsatellite instability and mismatch repair, supports intraoperative assessments of tumor invasion depth, predicts treatment responses, and facilitates the design of personalized treatment plans to potentially significantly enhance patient outcomes. Moreover, the integration of AI with multiomics analyses and imaging technologies has led to substantial advancements in foundational research on the tumor microenvironment. This review highlights the progress of AI in gastrointestinal oncology over the past 5 years with focus on early tumor screening, diagnosis, molecular marker identification, treatment planning, and prognosis predictions. We also explored the potential of AI to enhance medical imaging analyses to aid tumor detection and characterization as well as its role in automating and refining histopathological assessments.
Collapse
Affiliation(s)
- Shengyuan Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing, 102200, China.
| |
Collapse
|
6
|
Le Vuong TT, Kwak JT. MoMA: Momentum contrastive learning with multi-head attention-based knowledge distillation for histopathology image analysis. Med Image Anal 2025; 101:103421. [PMID: 39671769 DOI: 10.1016/j.media.2024.103421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/07/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024]
Abstract
There is no doubt that advanced artificial intelligence models and high quality data are the keys to success in developing computational pathology tools. Although the overall volume of pathology data keeps increasing, a lack of quality data is a common issue when it comes to a specific task due to several reasons including privacy and ethical issues with patient data. In this work, we propose to exploit knowledge distillation, i.e., utilize the existing model to learn a new, target model, to overcome such issues in computational pathology. Specifically, we employ a student-teacher framework to learn a target model from a pre-trained, teacher model without direct access to source data and distill relevant knowledge via momentum contrastive learning with multi-head attention mechanism, which provides consistent and context-aware feature representations. This enables the target model to assimilate informative representations of the teacher model while seamlessly adapting to the unique nuances of the target data. The proposed method is rigorously evaluated across different scenarios where the teacher model was trained on the same, relevant, and irrelevant classification tasks with the target model. Experimental results demonstrate the accuracy and robustness of our approach in transferring knowledge to different domains and tasks, outperforming other related methods. Moreover, the results provide a guideline on the learning strategy for different types of tasks and scenarios in computational pathology.
Collapse
Affiliation(s)
- Trinh Thi Le Vuong
- School of Electrical Engineering, Korea University, Seoul, Republic of Korea
| | - Jin Tae Kwak
- School of Electrical Engineering, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Li R, Li J, Wang Y, Liu X, Xu W, Sun R, Xue B, Zhang X, Ai Y, Du Y, Jiang J. The artificial intelligence revolution in gastric cancer management: clinical applications. Cancer Cell Int 2025; 25:111. [PMID: 40119433 PMCID: PMC11929235 DOI: 10.1186/s12935-025-03756-4] [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: 08/27/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
Nowadays, gastric cancer has become a significant issue in the global cancer burden, and its impact cannot be ignored. The rapid development of artificial intelligence technology is attempting to address this situation, aiming to change the clinical management landscape of gastric cancer fundamentally. In this transformative change, machine learning and deep learning, as two core technologies, play a pivotal role, bringing unprecedented innovations and breakthroughs in the diagnosis, treatment, and prognosis evaluation of gastric cancer. This article comprehensively reviews the latest research status and application of artificial intelligence algorithms in gastric cancer, covering multiple dimensions such as image recognition, pathological analysis, personalized treatment, and prognosis risk assessment. These applications not only significantly improve the sensitivity of gastric cancer risk monitoring, the accuracy of diagnosis, and the precision of survival prognosis but also provide robust data support and a scientific basis for clinical decision-making. The integration of artificial intelligence, from optimizing the diagnosis process and enhancing diagnostic efficiency to promoting the practice of precision medicine, demonstrates its promising prospects for reshaping the treatment model of gastric cancer. Although most of the current AI-based models have not been widely used in clinical practice, with the continuous deepening and expansion of precision medicine, we have reason to believe that a new era of AI-driven gastric cancer care is approaching.
Collapse
Affiliation(s)
- Runze Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Jingfan Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yuman Wang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xiaoyu Liu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Weichao Xu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Runxue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Binqing Xue
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xinqian Zhang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yikun Ai
- North China University of Science and Technology, Tanshan 063000, China
| | - Yanru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, 050011, China.
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, 050011, China.
| | - Jianming Jiang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
| |
Collapse
|
8
|
Wang P, Zhang J, Li Y, Guo Y, Li P, Chen R. Histopathology image classification based on semantic correlation clustering domain adaptation. Artif Intell Med 2025; 163:103110. [PMID: 40107119 DOI: 10.1016/j.artmed.2025.103110] [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: 06/23/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Deep learning has been successfully applied to histopathology image classification tasks. However, the performance of deep models is data-driven, and the acquisition and annotation of pathological image samples are difficult, which limit the model's performance. Compared to whole slide images (WSI) of patients, histopathology image datasets of animal models are easier to acquire and annotate. Therefore, this paper proposes an unsupervised domain adaptation method based on semantic correlation clustering for histopathology image classification. The aim is to utilize Minmice model histopathology image dataset to achieve the classification and recognition of human WSIs. Firstly, the multi-scale fused features extracted from the source and target domains are normalized and mapped. In the new feature space, the cosine distance between class centers is used to measure the semantic correlation between categories. Then, the domain centers, class centers, and sample distributions are self-constrainedly aligned. Multi-granular information is applied to achieve cross-domain semantic correlation knowledge transfer between classes. Finally, the probabilistic heatmap is used to visualize the model's prediction results and annotate the cancerous regions in WSIs. Experimental results show that the proposed method has high classification accuracy for WSI, and the annotated result is close to manual annotation, indicating its potential for clinical applications.
Collapse
Affiliation(s)
- Pin Wang
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400030, PR China.
| | - Jinhua Zhang
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400030, PR China
| | - Yongming Li
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400030, PR China
| | - Yurou Guo
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400030, PR China
| | - Pufei Li
- School of Microelectronics and Communication Engineering, Chongqing University, Chongqing 400030, PR China
| | - Rui Chen
- Chongqing University Cancer Hospital, Chongqing 400030, PR China
| |
Collapse
|
9
|
Ma Y, Jamdade S, Konduri L, Sailem H. AI in Histopathology Explorer for comprehensive analysis of the evolving AI landscape in histopathology. NPJ Digit Med 2025; 8:156. [PMID: 40074858 PMCID: PMC11904230 DOI: 10.1038/s41746-025-01524-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Digital pathology and artificial intelligence (AI) hold immense transformative potential to revolutionize cancer diagnostics, treatment outcomes, and biomarker discovery. Gaining a deeper understanding of deep learning algorithm methods applied to histopathological data and evaluating their performance on different tasks is crucial for developing the next generation of AI technologies. To this end, we developed AI in Histopathology Explorer (HistoPathExplorer); an interactive dashboard with intelligent tools available at www.histopathexpo.ai . This real-time online resource enables users, including researchers, decision-makers, and various stakeholders, to assess the current landscape of AI applications for specific clinical tasks, analyze their performance, and explore the factors influencing their translation into practice. Moreover, a quality index was defined for evaluating the comprehensiveness of methodological details in published AI methods. HistoPathExplorer highlights opportunities and challenges for AI in histopathology, and offers a valuable resource for creating more effective methods and shaping strategies and guidelines for translating digital pathology applications into clinical practice.
Collapse
Affiliation(s)
- Yingrui Ma
- School of Cancer and Pharmaceutical Sciences, Stamford St., Franklin Wilkins Building, King's College London, London, UK
| | - Shivprasad Jamdade
- School of Cancer and Pharmaceutical Sciences, Stamford St., Franklin Wilkins Building, King's College London, London, UK
| | - Lakshmi Konduri
- School of Cancer and Pharmaceutical Sciences, Stamford St., Franklin Wilkins Building, King's College London, London, UK
| | - Heba Sailem
- School of Cancer and Pharmaceutical Sciences, Stamford St., Franklin Wilkins Building, King's College London, London, UK.
- King's Institute for Artificial Intelligence, King's College London, London, UK.
| |
Collapse
|
10
|
Lee K, Jeon J, Park JW, Yu S, Won JK, Kim K, Park CK, Park SH. SNUH methylation classifier for CNS tumors. Clin Epigenetics 2025; 17:47. [PMID: 40075518 PMCID: PMC11905536 DOI: 10.1186/s13148-025-01824-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Methylation profiling of central nervous system (CNS) tumors, pioneered by the German Cancer Research Center, has significantly improved diagnostic accuracy. This study aimed to further enhance the performance of methylation classifiers by leveraging publicly available data and innovative machine-learning techniques. RESULTS Seoul National University Hospital Methylation Classifier (SNUH-MC) addressed data imbalance using the Synthetic Minority Over-sampling Technique (SMOTE) algorithm and incorporated OpenMax within a Multi-Layer Perceptron to prevent labeling errors in low-confidence diagnoses. Compared to two published CNS tumor methylation classification models (DKFZ-MC: Deutsches Krebsforschungszentrum Methylation Classifier v11b4: RandomForest, 767-MC: Multi-Layer Perceptron), our SNUH-MC showed improved performance in F1-score. For 'Filtered Test Data Set 1,' the SNUH-MC achieved higher F1-micro (0.932) and F1-macro (0.919) scores compared to DKFZ-MC v11b4 (F1-micro: 0.907, F1-macro: 0.627). We evaluated the performance of three classifiers; SNUH-MC, DKFZ-MC v11b4, and DKFZ-MC v12.5, using specific criteria. We set established 'Decisions' categories based on histopathology, clinical information, and next-generation sequencing to assess the classification results. When applied to 193 unknown SNUH methylation data samples, SNUH-MC notably improved diagnosis compared to DKFZ-MC v11b4. Specifically, 17 cases were reclassified as 'Match' and 34 cases as 'Likely Match' when transitioning from DKFZ-MC v11b4 to SNUH-MC. Additionally, SNUH-MC demonstrated similar results to DKFZ-MC v12.5 for 23 cases that were unclassified by v11b4. CONCLUSIONS This study presents SNUH-MC, an innovative methylation-based classification tool that significantly advances the field of neuropathology and bioinformatics. Our classifier incorporates cutting-edge techniques such as the SMOTE and OpenMax resulting in improved diagnostic accuracy and robustness, particularly when dealing with unknown or noisy data.
Collapse
Affiliation(s)
- Kwanghoon Lee
- Department of Pathology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Jaemin Jeon
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Jin Woo Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suwan Yu
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Cannarozzi AL, Biscaglia G, Parente P, Latiano TP, Gentile A, Ciardiello D, Massimino L, Di Brina ALP, Guerra M, Tavano F, Ungaro F, Bossa F, Perri F, Latiano A, Palmieri O. Artificial intelligence and whole slide imaging, a new tool for the microsatellite instability prediction in colorectal cancer: Friend or foe? Crit Rev Oncol Hematol 2025; 210:104694. [PMID: 40064251 DOI: 10.1016/j.critrevonc.2025.104694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common and second most deadly cancer worldwide. Despite advances in screening and treatment, CRC is heterogeneous and the response to therapy varies significantly, limiting personalized treatment options. Certain molecular biomarkers, including microsatellite instability (MSI), are critical in planning personalized treatment, although only a subset of patients may benefit. Currently, the primary methods for assessing MSI status include immunohistochemistry (IHC) for DNA mismatch repair proteins (MMRs), polymerase chain reaction (PCR)-based molecular testing, or next-generation sequencing (NGS). However, these techniques have limitations, are expensive and time-consuming, and often result in inter-method inconsistencies. Deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H) are critical predictive biomarkers of response to immune checkpoint inhibitor (ICI) therapy and MSI testing is recommended to identify patients who may benefit. There is a pressing need for a more robust, reliable, and cost-effective approach that accurately assesses MSI status. Recent advances in computational pathology, in particular the development of technologies that digitally scan whole slide images (WSI) at high resolution, as well as new approaches to artificial intelligence (AI) in medicine, are increasingly gaining ground. This review aims to provide an overview of the latest findings on WSI and advances in AI methods for predicting MSI status, summarize their applications in CRC, and discuss their strengths and limitations in daily clinical practice.
Collapse
Affiliation(s)
- Anna Lucia Cannarozzi
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Giuseppe Biscaglia
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy.
| | - Tiziana Pia Latiano
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo 71013, Italy.
| | - Annamaria Gentile
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan.
| | - Luca Massimino
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Anna Laura Pia Di Brina
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Maria Guerra
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesca Tavano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Federica Ungaro
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Fabrizio Bossa
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Anna Latiano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Orazio Palmieri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
12
|
Matias-Guiu X, Temprana-Salvador J, Garcia Lopez P, Kammerer-Jacquet SF, Rioux-Leclercq N, Clark D, Schürch CM, Fend F, Mattern S, Snead D, Fusco N, Guerini-Rocco E, Rojo F, Brevet M, Salto Tellez M, Dei Tos A, di Maio T, Ramírez-Peinado S, Sheppard E, Bannister H, Gkiokas A, Arpaia M, Ben Dhia O, Martino N. Implementing digital pathology: qualitative and financial insights from eight leading European laboratories. Virchows Arch 2025:10.1007/s00428-025-04064-y. [PMID: 40056197 DOI: 10.1007/s00428-025-04064-y] [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: 01/17/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
Digital Pathology (DP) revolutionizes the diagnostic workflow. Digitized scanned slides enhance operational efficiency by facilitating remote access, slide storage, reporting and automated AI image analysis, and enabling collaboration and research. However, substantial upfront and maintenance costs remain significant barriers to adoption. This study evaluates DP's financial and qualitative value, exploring whether the long-term financial benefits justify investments and addressing implementation challenges in large public and private European laboratory settings. A targeted literature review, semi-structured interviews, surveys, and a net present value (NPV) model were employed to assess DP's impact on clinical practice and laboratory financials. Qualitative findings validate the key benefits of DP, including optimized workflow, enhanced logistics, and improved laboratory organization. Pathologists reported a smooth integration, improved training, teaching, and research capabilities, and increased flexibility through remote work. Collaboration within multidisciplinary teams was strengthened, while case examination efficiency and access to archival slides were notably improved. Quantitative results indicate that DP demonstrates strong financial potential, achieving cost recovery within 6 years. DP investment results in a 7-year NPV of + €0.21 million (m) driven by increased productivity and diagnosis volumes. Although the high upfront costs for scanners, training, and system integration pose a significant barrier to the adoption of DP, larger institutions are better positioned to leverage economies of scale. This study underscores the importance of sustained financial support to cope with the initial investment and regional collaboration in driving widespread adoption of DP. Expanding reimbursement policies for pathology procedures could significantly reduce financial barriers.
Collapse
Affiliation(s)
- Xavier Matias-Guiu
- Hospital Universitari de Bellvitge and Hospital Universitari Arnau de Vilanova IDIBELL, IRBLLEIDA, University of Lleida, CIBERONC, Lleida, Spain.
| | | | | | | | | | - David Clark
- Nottingham University Hospitals NHS Trust, HMDN, Dept of Histopathology, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
- Cluster of Excellence Ifit (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Falko Fend
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sven Mattern
- Institute of Pathology and Neuropathology, Tübingen University Hospital, Tübingen, Germany
| | - David Snead
- UHCW NHS Trust, Coventry, CV2 2DX, UK
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Nicola Fusco
- European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Marie Brevet
- Technipath, Dommartin, France & Biwako, Lyon, France
| | - Manuel Salto Tellez
- Precision Medicine Centre, Queen'S University Belfast, Belfast, UK
- Integrated Patholog Unit, Institute for Cancer Research, London, UK
| | - Angelo Dei Tos
- Department of Integrated Diagnostics, University of Padua, Padua, Italy
| | - Thomas di Maio
- Regional Diagnostics AstraZeneca, 6340, Basel, Zug/CH, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Ebigbo A, Messmann H, Lee SH. Artificial Intelligence Applications in Image-Based Diagnosis of Early Esophageal and Gastric Neoplasms. Gastroenterology 2025:S0016-5085(25)00471-8. [PMID: 40043857 DOI: 10.1053/j.gastro.2025.01.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 04/03/2025]
Abstract
Artificial intelligence (AI) holds the potential to transform the management of upper gastrointestinal (GI) conditions, such as Barrett's esophagus, esophageal squamous cell cancer, and early gastric cancer. Advancements in deep learning and convolutional neural networks offer improved diagnostic accuracy and reduced diagnostic variability across different clinical settings, particularly where human error or fatigue may impair diagnostic precision. Deep learning models have shown the potential to improve early cancer detection and lesion characterization, predict invasion depth, and delineate lesion margins with remarkable accuracy, all contributing to effective treatment planning. Several challenges, however, limit the broad application of AI in GI endoscopy, particularly in the upper GI tract. Subtle lesion morphology and restricted diversity in training datasets, which are often sourced from specialized centers, may constrain the generalizability of AI models in various clinical settings. Furthermore, the "black box" nature of some AI systems can impede explainability and clinician trust. To address these issues, efforts are underway to incorporate multimodal data, such as combining endoscopic and histopathologic imaging, to bolster model robustness and transparency. In the future, AI promises substantial advancements in automated real-time endoscopic guidance, personalized risk assessment, and optimized biopsy decision making. As it evolves, it would substantially impact not only early diagnosis and prognosis, but also the cost-effectiveness of managing upper GI diseases, ultimately leading to improved patient outcomes and more efficient health care delivery.
Collapse
Affiliation(s)
- Alanna Ebigbo
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Helmut Messmann
- Department of Gastroenterology, University Hospital Augsburg, Augsburg, Germany.
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea; Seoul St. Mary's Hospital, Seoul, South Korea.
| |
Collapse
|
14
|
de la Fouchardière C, Cammarota A, Svrcek M, Alsina M, Fleitas-Kanonnikoff T, Lordick Obermannová R, Wagner AD, Yap Wei Ting D, Enea D, Petrillo A, Smyth EC. How do I treat dMMR/MSI gastro-oesophageal adenocarcinoma in 2025? A position paper from the EORTC-GITCG gastro-esophageal task force. Cancer Treat Rev 2025; 134:102890. [PMID: 39933210 DOI: 10.1016/j.ctrv.2025.102890] [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: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
In less than a decade, immune checkpoint inhibitors (ICIs) have transformed the management of mismatch repair-deficient (dMMR) and microsatellite instability-high (MSI) cancers. However, beyond colorectal cancer (CRC), much of the evidence is mostly derived from non-randomized phase II studies or post-hoc analyses of broader clinical trials. dMMR/MSI tumours represent a specific subgroup of gastro-esophageal adenocarcinomas (GEA), accounting for approximately 9 % of cases, with a higher prevalence in early-stage compared to advanced-stage disease and older female patients. These tumours are predominantly sporadic, often linked to MLH1 promoter methylation, and rarely exhibit HER2 overexpression/ERBB2 amplification or other oncogenic drivers. The treatment landscape for early stage dMMR/MSI GEA is likely to change substantially soon, as ICIs have shown high pathological complete response (pCR) rates in small phase II trials, raising questions on optimisation of neoadjuvant therapy, and paving the way for organ preservation. The standard of treatment for untreated patients with advanced dMMR/MSI GEA is chemotherapy + ICI irrespectively of PDL-1 status. However, the role of chemotherapy-free regimen consisting of CTLA-4 plus PD-1 inhibitors remains undetermined. This review addresses these and other emerging questions, offering expert opinions and insights into the future therapeutic landscape for dMMR/MSI GEA.
Collapse
Affiliation(s)
- Christelle de la Fouchardière
- Institut PAOLI-CALMETTES, 232 Boulevard Sainte-Marguerite 13009, Marseille, France; Unicancer GI (UCGI) Group, Paris, France; EORTC-GITC Group, Brussels, Belgium.
| | - Antonella Cammarota
- EORTC-GITC Group, Brussels, Belgium; Hepatobiliary Immunopathology Lab, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Magali Svrcek
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France; LIMICS, UMRS 1142, Campus des Cordeliers 75006, Paris, France
| | - Maria Alsina
- EORTC-GITC Group, Brussels, Belgium; Hospital Universitario de Navarra, Navarrabiomed - IdiSNA, c. de Irunlarrea 3 31008, Pamplona, Spain
| | - Tania Fleitas-Kanonnikoff
- EORTC-GITC Group, Brussels, Belgium; Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Radka Lordick Obermannová
- EORTC-GITC Group, Brussels, Belgium; Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute and Faculty of Medicine, Masaryk University, Czech Republic
| | - Anna Dorothea Wagner
- EORTC-GITC Group, Brussels, Belgium; Anna Dorothea Wagner, Department of Oncology, Division of Medical Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), 1011, Lausanne, Switzerland
| | | | - Diana Enea
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Department of Pathology, France
| | - Angelica Petrillo
- EORTC-GITC Group, Brussels, Belgium; Medical Oncology Unit, Ospedale del Mare, Naples, Italy
| | - Elizabeth C Smyth
- EORTC-GITC Group, Brussels, Belgium; Oxford NIHRBiomedical Research Centre, Churchill Hospital, Oxford OX3 7LE, UK
| |
Collapse
|
15
|
Ligero M, El Nahhas OSM, Aldea M, Kather JN. Artificial intelligence-based biomarkers for treatment decisions in oncology. Trends Cancer 2025; 11:232-244. [PMID: 39814650 DOI: 10.1016/j.trecan.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The development of new therapeutic strategies such as immune checkpoint inhibitors (ICIs) and targeted therapies has increased the complexity of the treatment landscape for solid tumors. At the current rate of annual FDA approvals, the potential treatment options could increase by tenfold over the next 5 years. The cost of personalized medicine technologies limits its accessibility, thus increasing socioeconomic disparities in the treated population. In this review we describe artificial intelligence (AI)-based solutions - including deep learning (DL) methods for routine medical imaging and large language models (LLMs) for electronic health records (EHRs) - to support cancer treatment decisions with cost-effective biomarkers. We address the current limitations of these technologies and propose the next steps towards their adoption in routine clinical practice.
Collapse
Affiliation(s)
- Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Mihaela Aldea
- Department of Cancer Medicine, Institut Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Thoracic Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
16
|
Liao Y, Chen X, Hu S, Chen B, Zhuo X, Xu H, Wu X, Zeng X, Zeng H, Zhang D, Zhi Y, Zhao L. Artificial Intelligence for Predicting HER2 Status of Gastric Cancer Based on Whole-Slide Histopathology Images: A Retrospective Multicenter Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408451. [PMID: 39792693 PMCID: PMC11904990 DOI: 10.1002/advs.202408451] [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: 07/23/2024] [Revised: 12/15/2024] [Indexed: 01/12/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) positive gastric cancer (GC) shows a robust response to the combined therapy based HER2-targeted therapy. The application of these therapies is highly dependent on the evaluation of tumor HER2 status. However, there are many risks and challenges in HER2 assessment in GC. Therefore, an economically viable and readily available instrument is requisite for distinguishing HER2 status among patients diagnosed with GC. The study has innovatively developed a deep learning model, HER2Net, which can predict the HER2 status by quantitatively calculating the proportion of HER2 high-expression regions. The HER2Net is trained on an internal training set derived from 531 hematoxylin & eosin (H&E) whole slide images (WSI) of 520 patients. Subsequently, the performance of HER2Net is validated on an internal test set from 115 H&E WSI of 111 patients and an external multi-center test set from 102 H&E WSI of 101 patients. The HER2Net achieves an accuracy of 0.9043 on the internal test set, and an accuracy of 0.8922 on an external test set from multiple institutes. This discovery indicates that the HER2Net can potentially offer a novel methodology for the identification of HER2-positive GC.
Collapse
Affiliation(s)
- Yuhan Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xinhua Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shupeng Hu
- School of Computer Science, University of Manchester, Manchester, M13 9PL, UK
| | - Bing Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xinghua Zhuo
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaojin Wu
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Xiaofeng Zeng
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Huimin Zeng
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Donghui Zhang
- Department of Pathology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Yunfei Zhi
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| |
Collapse
|
17
|
Carrillo-Perez F, Pizurica M, Zheng Y, Nandi TN, Madduri R, Shen J, Gevaert O. Generation of synthetic whole-slide image tiles of tumours from RNA-sequencing data via cascaded diffusion models. Nat Biomed Eng 2025; 9:320-332. [PMID: 38514775 DOI: 10.1038/s41551-024-01193-8] [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: 12/27/2022] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Training machine-learning models with synthetically generated data can alleviate the problem of data scarcity when acquiring diverse and sufficiently large datasets is costly and challenging. Here we show that cascaded diffusion models can be used to synthesize realistic whole-slide image tiles from latent representations of RNA-sequencing data from human tumours. Alterations in gene expression affected the composition of cell types in the generated synthetic image tiles, which accurately preserved the distribution of cell types and maintained the cell fraction observed in bulk RNA-sequencing data, as we show for lung adenocarcinoma, kidney renal papillary cell carcinoma, cervical squamous cell carcinoma, colon adenocarcinoma and glioblastoma. Machine-learning models pretrained with the generated synthetic data performed better than models trained from scratch. Synthetic data may accelerate the development of machine-learning models in scarce-data settings and allow for the imputation of missing data modalities.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Ghent, Belgium
| | - Yuanning Zheng
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Tarak Nath Nandi
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Ravi Madduri
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Jeanne Shen
- Department of Pathology, Stanford University, School of Medicine, Palo Alto, CA, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
18
|
Zalila-Kolsi I, Dhieb D, Osman HA, Mekideche H. The Gut Microbiota and Colorectal Cancer: Understanding the Link and Exploring Therapeutic Interventions. BIOLOGY 2025; 14:251. [PMID: 40136508 PMCID: PMC11939563 DOI: 10.3390/biology14030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
CRC remains a significant public health challenge due to its high prevalence and mortality rates. Emerging evidence highlights the critical role of the gut microbiota in both the pathogenesis of CRC and the efficacy of treatment strategies, including chemotherapy and immunotherapy. Dysbiosis, characterized by imbalances in microbial communities, has been implicated in CRC progression and therapeutic outcomes. This review examines the intricate relationship between gut microbiota composition and CRC, emphasizing the potential for microbial profiles to serve as biomarkers for early detection and prognosis. Various interventions, such as prebiotics, probiotics, postbiotics, fecal microbiota transplantation, and dietary modifications, aim to restore microbiota balance and shift dysbiosis toward eubiosis, thereby improving health outcomes. Additionally, the integration of microbial profiling into clinical practice could enhance diagnostic capabilities and personalize treatment strategies, advancing the field of oncology. The study of intratumoral microbiota offers new diagnostic and prognostic tools that, combined with artificial intelligence algorithms, could predict treatment responses and assess the risk of adverse effects. Given the growing understanding of the gut microbiome-cancer axis, developing microbiota-oriented strategies for CRC prevention and treatment holds promise for improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Imen Zalila-Kolsi
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Dhoha Dhieb
- College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussam A. Osman
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Hadjer Mekideche
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| |
Collapse
|
19
|
Zhang P, Gao C, Zhang Z, Yuan Z, Zhang Q, Zhang P, Du S, Zhou W, Li Y, Li S. Systematic inference of super-resolution cell spatial profiles from histology images. Nat Commun 2025; 16:1838. [PMID: 39984438 PMCID: PMC11845739 DOI: 10.1038/s41467-025-57072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Inferring cell spatial profiles from histology images is critical for cancer diagnosis and treatment in clinical settings. In this study, we report a weakly-supervised deep-learning method, HistoCell, to directly infer super-resolution cell spatial profiles consisting of cell types, cell states and their spatial network from histology images at the single-nucleus-level. Benchmark analysis demonstrates that HistoCell robustly achieves state-of-the-art performance in terms of cell type/states prediction solely from histology images across multiple cancer tissues. HistoCell can significantly enhance the deconvolution accuracy for the spatial transcriptomics data and enable accurate annotation of subtle cancer tissue architectures. Moreover, HistoCell is applied to de novo discovery of clinically relevant spatial organization indicators, including prognosis and drug response biomarkers, across diverse cancer types. HistoCell also enable image-based screening of cell populations that drives phenotype of interest, and is applied to discover the cell population and corresponding spatial organization indicators associated with gastric malignant transformation risk. Overall, HistoCell emerges as a powerful and versatile tool for cancer studies in histology image-only cohorts.
Collapse
Affiliation(s)
- Peng Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Chaofei Gao
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhuoyu Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qian Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Shao Li
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
20
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 DOI: 10.1038/s41392-024-02082-x] [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: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
21
|
Gustav M, van Treeck M, Reitsam NG, Carrero ZI, Loeffler CML, Meneghetti AR, Märkl B, Boardman LA, French AJ, Goode EL, Gsur A, Brezina S, Gunter MJ, Murphy N, Hönscheid P, Sperling C, Foersch S, Steinfelder R, Harrison T, Peters U, Phipps A, Kather JN. Assessing Genotype-Phenotype Correlations with Deep Learning in Colorectal Cancer: A Multi-Centric Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.04.25321660. [PMID: 39973981 PMCID: PMC11838662 DOI: 10.1101/2025.02.04.25321660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Deep Learning (DL) has emerged as a powerful tool to predict genetic biomarkers directly from digitized Hematoxylin and Eosin (H&E) slides in colorectal cancer (CRC). However, few studies have systematically investigated the predictability of biomarkers beyond routinely available alterations such as microsatellite instability (MSI), and BRAF and KRAS mutations. Methods Our primary dataset comprised H&E slides of CRC tumors across five cohorts totaling 1,376 patients who underwent comprehensive panel sequencing, with an additional 536 patients from two public datasets for validation. We developed a DL model using a single transformer model to predict multiple genetic alterations directly from the slides. The model's performance was compared against conventional single-target models, and potential confounders were analyzed. Findings The multi-target model was able to predict numerous biomarkers from pathology slides, matching and partly exceeding single-target transformers. The Area Under the Receiver Operating Characteristic curve (AUROC, mean ± std) on the primary external validation cohorts was: BRAF (0·78 ± 0·01), hypermutation (0·88 ± 0·01), MSI (0·93 ± 0·01), RNF43 (0·86 ± 0·01); this biomarker predictability was mirrored across metrics and co-occurrence analyses. However, biomarkers with high AUROCs largely correlated with MSI, with model predictions depending considerably on MSI-associated morphology upon pathological examination. Interpretation Our study demonstrates that multi-target transformers can predict the biomarker status for numerous genetic alterations in CRC directly from H&E slides. However, their predictability is mainly associated with MSI phenotype, despite indications of slight biomarker-inherent contributions to a phenotype. Our findings underscore the need to analyze confounders in AI-based oncology biomarkers. To enable this, we developed a validated model applicable to other cancers and larger, diverse datasets. Funding The German Federal Ministry of Health, the Max-Eder-Programme of German Cancer Aid, the German Federal Ministry of Education and Research, the German Academic Exchange Service, and the EU.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Nic G. Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Zunamys I. Carrero
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Chiara M. L. Loeffler
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Asier Rabasco Meneghetti
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Lisa A. Boardman
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Amy J. French
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ellen L. Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Gsur
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Pia Hönscheid
- Institute of Pathology, University Hospital Carl Gustav Carus (UKD), Technical University Dresden (TUD), Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center Heidelberg, Dresden, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Sperling
- Institute of Pathology, University Hospital Carl Gustav Carus (UKD), Technical University Dresden (TUD), Dresden, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Robert Steinfelder
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tabitha Harrison
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Amanda Phipps
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
22
|
Guan B, Chu G, Wang Z, Li J, Yi B. Instance-level semantic segmentation of nuclei based on multimodal structure encoding. BMC Bioinformatics 2025; 26:42. [PMID: 39915737 PMCID: PMC11804060 DOI: 10.1186/s12859-025-06066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Accurate segmentation and classification of cell nuclei are crucial for histopathological image analysis. However, existing deep neural network-based methods often struggle to capture complex morphological features and global spatial distributions of cell nuclei due to their reliance on local receptive fields. METHODS This study proposes a graph neural structure encoding framework based on a vision-language model. The framework incorporates: (1) A multi-scale feature fusion and knowledge distillation module utilizing the Contrastive Language-Image Pre-training (CLIP) model's image encoder; (2) A method to transform morphological features of cells into textual descriptions for semantic representation; and (3) A graph neural network approach to learn spatial relationships and contextual information between cell nuclei. RESULTS Experimental results demonstrate that the proposed method significantly improves the accuracy of cell nucleus segmentation and classification compared to existing approaches. The framework effectively captures complex nuclear structures and global distribution features, leading to enhanced performance in histopathological image analysis. CONCLUSIONS By deeply mining the morphological features of cell nuclei and their spatial topological relationships, our graph neural structure encoding framework achieves high-precision nuclear segmentation and classification. This approach shows significant potential for enhancing histopathological image analysis, potentially leading to more accurate diagnoses and improved understanding of cellular structures in pathological tissues.
Collapse
Affiliation(s)
- Bo Guan
- Key Lab for Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China
| | - Guangdi Chu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ziying Wang
- Department of Medicine, Qingdao University, Qingdao, 266000, China
| | - Jianmin Li
- Key Lab for Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China.
| | - Bo Yi
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
23
|
Cui H, Guo Q, Xu J, Wu X, Cai C, Jiao Y, Ming W, Wen H, Wang X. Prediction of molecular subtypes for endometrial cancer based on hierarchical foundation model. BIOINFORMATICS (OXFORD, ENGLAND) 2025; 41:btaf059. [PMID: 39932017 DOI: 10.1093/bioinformatics/btaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/13/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
MOTIVATION Endometrial cancer is a prevalent gynecological malignancy that requires accurate identification of its molecular subtypes for effective diagnosis and treatment. Four molecular subtypes with different clinical outcomes have been identified: POLE mutation, mismatch repair deficient, p53 abnormal, and no specific molecular profile. However, determining these subtypes typically relies on expensive gene sequencing. To overcome this limitation, we propose a novel method that utilizes hematoxylin and eosin-stained whole slide images to predict endometrial cancer molecular subtypes. RESULTS Our approach leverages a hierarchical foundation model as a backbone, fine-tuned from the UNI computational pathology foundation model, to extract tissue embedding from different scales. We have achieved promising results through extensive experimentation on the Fudan University Shanghai Cancer Center cohort (N = 364). Our model demonstrates a macro-average AUROC of 0.879 (95% CI, 0.853-0.904) in a five-fold cross-validation. Compared to the current state-of-the-art molecular subtypes prediction for endometrial cancer, our method outperforms in terms of predictive accuracy and computational efficiency. Moreover, our method is highly reproducible, allowing for ease of implementation and widespread adoption. This study aims to address the cost and time constraints associated with traditional gene sequencing techniques. By providing a reliable and accessible alternative to gene sequencing, our method has the potential to revolutionize the field of endometrial cancer diagnosis and improve patient outcomes. AVAILABILITY AND IMPLEMENTATION The codes and data used for generating results in this study are available at https://github.com/HaoyuCui/hi-UNI for GitHub and https://doi.org/10.5281/zenodo.14627478 for Zenodo.
Collapse
Affiliation(s)
- Haoyu Cui
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Qinhao Guo
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jun Xu
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chengfei Cai
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Yiping Jiao
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Wenlong Ming
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Hao Wen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiangxue Wang
- Jiangsu Key Laboratory of Intelligent Medical Image Computing, Nanjing University of Information Science and Technology, Nanjing 210044, China
| |
Collapse
|
24
|
Gerussi A, Saldanha OL, Cazzaniga G, Verda D, Carrero ZI, Engel B, Taubert R, Bolis F, Cristoferi L, Malinverno F, Colapietro F, Akpinar R, Di Tommaso L, Terracciano L, Lleo A, Viganó M, Rigamonti C, Cabibi D, Calvaruso V, Gibilisco F, Caldonazzi N, Valentino A, Ceola S, Canini V, Nofit E, Muselli M, Calderaro J, Tiniakos D, L’Imperio V, Pagni F, Zucchini N, Invernizzi P, Carbone M, Kather JN. Deep learning helps discriminate between autoimmune hepatitis and primary biliary cholangitis. JHEP Rep 2025; 7:101198. [PMID: 39829723 PMCID: PMC11741034 DOI: 10.1016/j.jhepr.2024.101198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 01/03/2025] Open
Abstract
Background & Aims Biliary abnormalities in autoimmune hepatitis (AIH) and interface hepatitis in primary biliary cholangitis (PBC) occur frequently, and misinterpretation may lead to therapeutic mistakes with a negative impact on patients. This study investigates the use of a deep learning (DL)-based pipeline for the diagnosis of AIH and PBC to aid differential diagnosis. Methods We conducted a multicenter study across six European referral centers, and built a library of digitized liver biopsy slides dating from 1997 to 2023. A training set of 354 cases (266 AIH and 102 PBC) and an external validation set of 92 cases (62 AIH and 30 PBC) were available for analysis. A novel DL model, the autoimmune liver neural estimator (ALNE), was trained on whole-slide images (WSIs) with H&E staining, without human annotations. The ALNE model was evaluated against clinico-pathological diagnoses and tested for interobserver variability among general pathologists. Results The ALNE model demonstrated high accuracy in differentiating AIH from PBC, achieving an area under the receiver operating characteristic curve of 0.81 in external validation. Attention heatmaps showed that ALNE tends to focus more on areas with increased inflammation, associating such patterns predominantly with AIH. A multivariate explainable ML model revealed that PBC cases misclassified as AIH more often had ALP values between 1 × upper limit of normal (ULN) and 2 × ULN, coupled with AST values above 1 × ULN. Inconsistency among general pathologists was noticed when evaluating a random sample of the same cases (Fleiss's kappa value 0.09). Conclusions The ALNE model is the first system generating a quantitative and accurate differential diagnosis between cases with AIH or PBC. Impact and implications This study demonstrates the significant potential of the autoimmune liver neural estimator model, a transformer-based deep learning system, in accurately distinguishing between autoimmune hepatitis and primary biliary cholangitis using digitized liver biopsy slides without human annotation. The scientific justification for this work lies in addressing the challenge of differentiating these conditions, which often present with overlapping features and can lead to therapeutic mistakes. In addition, there is need for quantitative assessment of information embedded in liver biopsies, which are currently evaluated on qualitative or semi-quantitative methods. The results of this study are crucial for pathologists, researchers, and clinicians, providing a reliable diagnostic tool that reduces interobserver variability and improves diagnostic accuracy of these conditions. Potential methodological limitations, such as the diversity in scanning techniques and slide colorations, were considered, ensuring the robustness and generalizability of the findings.
Collapse
Affiliation(s)
- Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Oliver Lester Saldanha
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Giorgio Cazzaniga
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | | | - Zunamys I. Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Bastian Engel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Francesca Bolis
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Cristoferi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Federica Malinverno
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Francesca Colapietro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Reha Akpinar
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Pathology, IRCSS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Pathology, IRCSS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Luigi Terracciano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Pathology, IRCSS Humanitas Research Hospital, Rozzano-Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mauro Viganó
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Cristina Rigamonti
- Department of Translational Medicine, Università del Piemonte Orientale, Division of Internal Medicine, AOU Maggiore della Carità, Novara, Italy
| | - Daniela Cabibi
- Pathology Institute, PROMISE, University of Palermo, Palermo, Italy
| | - Vincenza Calvaruso
- Gastrointestinal and Liver Unit, Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Fabio Gibilisco
- Department of Pathology, Hospital “Gravina e Santo Pietro”, Caltagirone, Italy
- Department of Medical and Surgical Sciences and Advanced Technologies, “G. F. Ingrassia”, University of Catania, Catania, Italy
| | - Nicoló Caldonazzi
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Stefano Ceola
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | - Valentina Canini
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | - Eugenia Nofit
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - Dina Tiniakos
- Department of Pathology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Vincenzo L’Imperio
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | - Nicola Zucchini
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, Università di Milano-Bicocca, Monza, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marco Carbone
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Liver Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
- Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
25
|
Hezi H, Gelber M, Balabanov A, Maruvka YE, Freiman M. CIMIL-CRC: A clinically-informed multiple instance learning framework for patient-level colorectal cancer molecular subtypes classification from H&E stained images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 259:108513. [PMID: 39581068 DOI: 10.1016/j.cmpb.2024.108513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND AND OBJECTIVE Treatment approaches for colorectal cancer (CRC) are highly dependent on the molecular subtype, as immunotherapy has shown efficacy in cases with microsatellite instability (MSI) but is ineffective for the microsatellite stable (MSS) subtype. There is promising potential in utilizing deep neural networks (DNNs) to automate the differentiation of CRC subtypes by analyzing hematoxylin and eosin (H&E) stained whole-slide images (WSIs). Due to the extensive size of WSIs, multiple instance learning (MIL) techniques are typically explored. However, existing MIL methods focus on identifying the most representative image patches for classification, which may result in the loss of critical information. Additionally, these methods often overlook clinically relevant information, like the tendency for MSI class tumors to predominantly occur on the proximal (right side) colon. METHODS We introduce 'CIMIL-CRC', a DNN framework that: (1) solves the MSI/MSS MIL problem by efficiently combining a pre-trained feature extraction model with principal component analysis (PCA) to aggregate information from all patches, and (2) integrates clinical priors, particularly the tumor location within the colon, into the model to enhance patient-level classification accuracy. We assessed our CIMIL-CRC method using the average area under the receiver operating characteristic curve (AUROC) from a 5-fold cross-validation experimental setup for model development on the TCGA-CRC-DX cohort, contrasting it with a baseline patch-level classification, a MIL-only approach, and a clinically-informed patch-level classification approach. RESULTS Our CIMIL-CRC outperformed all methods (AUROC: 0.92±0.002 (95% CI 0.91-0.92), vs. 0.79±0.02 (95% CI 0.76-0.82), 0.86±0.01 (95% CI 0.85-0.88), and 0.87±0.01 (95% CI 0.86-0.88), respectively). The improvement was statistically significant. To the best of our knowledge, this is the best result achieved for MSI/MSS classification on this dataset. CONCLUSION Our CIMIL-CRC method holds promise for offering insights into the key representations of histopathological images and suggests a straightforward implementation.
Collapse
Affiliation(s)
- Hadar Hezi
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Matan Gelber
- Faculty of Electrical and Computer Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexander Balabanov
- Faculty of Electrical and Computer Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yosef E Maruvka
- Faculty of Food Engineering and Biotechnology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Moti Freiman
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
26
|
Cai Y, Chen X, Chen J, Liao J, Han M, Lin D, Hong X, Hu H, Hu J. Deep learning-assisted colonoscopy images for prediction of mismatch repair deficiency in colorectal cancer. Surg Endosc 2025; 39:859-867. [PMID: 39623175 DOI: 10.1007/s00464-024-11426-1] [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: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Deficient mismatch repair or microsatellite instability is a major predictive biomarker for the efficacy of immune checkpoint inhibitors of colorectal cancer. However, routine testing has not been uniformly implemented due to cost and resource constraints. METHODS We developed and validated a deep learning-based classifiers to detect mismatch repair-deficient status from routine colonoscopy images. We obtained the colonoscopy images from the imaging database at Endoscopic Center of the Sixth Affiliated Hospital, Sun Yat-sen University. Colonoscopy images from a prospective trial (Neoadjuvant PD-1 blockade by toripalimab with or without celecoxib in mismatch repair-deficient or microsatellite instability-high locally advanced colorectal cancer) were used to test the model. RESULTS A total of 5226 eligible images from 892 tumors from the consecutive patients were utilized to develop and validate the deep learning model. 2105 colorectal cancer images from 306 tumors were randomly selected to form model development dataset with a class-balanced approach. 3121 images of 488 proficient mismatch repair tumors and 98 deficient mismatch repair tumors were used to form the independent dataset. The model achieved an AUROC of 0.948 (95% CI 0.919-0.977) on the test dataset. On the independent validation dataset, the AUROC was 0.807 (0.760-0.854), and the NPV in was 94.2% (95% CI 0.918-0.967). On the prospective trial dataset, the model identified 29 tumors among the 33 deficient mismatch repair tumors (87.88%). CONCLUSIONS The model achieved a high NPV in detecting deficient mismatch repair colorectal cancers. This model might serve as an automatic screening tool.
Collapse
Affiliation(s)
- Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
| | - Xijie Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Gastric Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junguo Chen
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - James Liao
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Ming Han
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Dezheng Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoling Hong
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
| | - Jiancong Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Lin Y, Wang Y, Fang Z, Li Z, Guan X, Jiang D, Zhang Y. A Multi-Perspective Self-Supervised Generative Adversarial Network for FS to FFPE Stain Transfer. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:774-788. [PMID: 39283778 DOI: 10.1109/tmi.2024.3460795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
In clinical practice, frozen section (FS) images can be utilized to obtain the immediate pathological results of the patients in operation due to their fast production speed. However, compared with the formalin-fixed and paraffin-embedded (FFPE) images, the FS images greatly suffer from poor quality. Thus, it is of great significance to transfer the FS image to the FFPE one, which enables pathologists to observe high-quality images in operation. However, obtaining the paired FS and FFPE images is quite hard, so it is difficult to obtain accurate results using supervised methods. Apart from this, the FS to FFPE stain transfer faces many challenges. Firstly, the number and position of nuclei scattered throughout the image are hard to maintain during the transfer process. Secondly, transferring the blurry FS images to the clear FFPE ones is quite challenging. Thirdly, compared with the center regions of each patch, the edge regions are harder to transfer. To overcome these problems, a multi-perspective self-supervised GAN, incorporating three auxiliary tasks, is proposed to improve the performance of FS to FFPE stain transfer. Concretely, a nucleus consistency constraint is designed to enable the high-fidelity of nuclei, an FFPE guided image deblurring is proposed for improving the clarity, and a multi-field-of-view consistency constraint is designed to better generate the edge regions. Objective indicators and pathologists' evaluation for experiments on the five datasets across different countries have demonstrated the effectiveness of our method. In addition, the validation in the downstream task of microsatellite instability prediction has also proved the performance improvement by transferring the FS images to FFPE ones. Our code link is https://github.com/linyiyang98/Self-Supervised-FS2FFPE.git.
Collapse
|
28
|
Xiao H, Wang J, Weng Z, Lin X, Shu M, Shen J, Sun P, Cai M, Xiang X, Li B, Wei L, Shi Y, Lai J, Kuang M, Yun J, Chen S, Peng S. A histopathology-based artificial intelligence system assisting the screening of genetic alteration in intrahepatic cholangiocarcinoma. Br J Cancer 2025; 132:195-202. [PMID: 39623041 PMCID: PMC11747625 DOI: 10.1038/s41416-024-02910-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Targeted therapy for intrahepatic cholangiocarcinoma (ICC) shows superior survival outcomes but patients with certain targetable alterations are no more than 20%. Genetic alteration screening for all ICC patients is of high cost and not routinely performed. This study intends to develop a histopathology-based artificial intelligence (AI)-assisted system for predicting genetic alteration of ICC. METHODS We constructed a Genetic Alteration Prediction (GAP) system based on multi-instance learning and self-supervised learning to predict genetic alterations using whole-slide images (WSIs) of H&E-stained slides. A total of 2069 WSIs from 232 ICC patients underwent surgery of the FAH-SYSU dataset were used for model construction and adjustment by five-fold cross-validation. Another 150 patients from three medical centres were used as independent external validations. We also compared the cost-effectiveness of GAP-assisted precise treatment and all-sequencing strategy to non-sequencing strategy. RESULTS The GAP was able to predict actionable genetic alterations of ICC, including FGFR2 and IDH. The area under the receiver operating characteristic curves (AUC) for FGFR2 and IDH were 0.754 and 0.713 in the internal dataset, and 0.724 and 0.656 in the external dataset, respectively. Furthermore, compared to giving chemotherapy without sequencing for every patient, GAP-assisted precise treatment could increase 1 progression-free quality-adjusted life month with a cost of $13871.72, the co-responding figure for all-sequencing strategy is $44538.93. Decision curve analysis showed that AI-assisted strategy provides better clinical benefits. CONCLUSIONS We constructed an AI-assisted genetic alteration screening system which is predictable to ICC actionable targets and has potential to assist precise targeted treatment of advanced ICC.
Collapse
Affiliation(s)
- Han Xiao
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Wang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongpeng Weng
- Department of Biology and Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxuan Lin
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Man Shu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingxian Shen
- State Key Laboratory of Oncology in Southern China, Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Peng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Muyan Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao Xiang
- Center of Hepato‑Pancreato‑Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Bin Li
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lihong Wei
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yiyu Shi
- Department of CSE, University of Notre Dame, Notre Dame, IN, USA
| | - Jiaming Lai
- Center of Hepato‑Pancreato‑Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Kuang
- Center of Hepato‑Pancreato‑Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Shuling Chen
- Department of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Sui Peng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
29
|
Rakaee M, Tafavvoghi M, Ricciuti B, Alessi JV, Cortellini A, Citarella F, Nibid L, Perrone G, Adib E, Fulgenzi CAM, Hidalgo Filho CM, Di Federico A, Jabar F, Hashemi S, Houda I, Richardsen E, Rasmussen Busund LT, Donnem T, Bahce I, Pinato DJ, Helland Å, Sholl LM, Awad MM, Kwiatkowski DJ. Deep Learning Model for Predicting Immunotherapy Response in Advanced Non-Small Cell Lung Cancer. JAMA Oncol 2025; 11:109-118. [PMID: 39724105 PMCID: PMC11843371 DOI: 10.1001/jamaoncol.2024.5356] [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: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 12/28/2024]
Abstract
Importance Only a small fraction of patients with advanced non-small cell lung cancer (NSCLC) respond to immune checkpoint inhibitor (ICI) treatment. For optimal personalized NSCLC care, it is imperative to identify patients who are most likely to benefit from immunotherapy. Objective To develop a supervised deep learning-based ICI response prediction method; evaluate its performance alongside other known predictive biomarkers; and assess its association with clinical outcomes in patients with advanced NSCLC. Design, Setting, and Participants This multicenter cohort study developed and independently validated a deep learning-based response stratification model for predicting ICI treatment outcome in patients with advanced NSCLC from whole slide hematoxylin and eosin-stained images. Images for model development and validation were obtained from 1 participating center in the US and 3 in the European Union (EU) from August 2014 to December 2022. Data analyses were performed from September 2022 to May 2024. Exposure Monotherapy with ICIs. Main Outcomes and Measures Model performance measured by clinical end points and objective response rate (ORR) differentiation power vs other predictive biomarkers, ie, programmed death-ligand 1 (PD-L1), tumor mutational burden (TMB), and tumor-infiltrating lymphocytes (TILs). Results A total of 295 581 image tiles from 958 patients (mean [SD] age, 66.0 [10.6] years; 456 [48%] females and 502 [52%] males) treated with ICI for NSCLC were included in the analysis. The US-based development cohort consisted of 614 patients with median (IQR) follow-up time of 54.5 (38.2-68.1) months, and the EU-based validation cohort, 344 patients with 43.3 (27.4-53.9) months of follow-up. The ORR to ICI was 26% in the developmental cohort and 28% in the validation cohort. The deep learning model's area under the receiver operating characteristic curve (AUC) for ORR was 0.75 (95% CI, 0.64-0.85) in the internal test set and 0.66 (95% CI, 0.60-0.72) in the validation cohort. In a multivariable analysis, the deep learning model's score was an independent predictor of ICI response in the validation cohort for both progression-free (hazard ratio, 0.56; 95% CI, 0.42-0.76; P < .001) and overall survival (hazard ratio, 0.53; 95% CI, 0.39-0.73; P < .001). The tuned deep learning model achieved a higher AUC than TMB, TILs, and PD-L1 in the internal set; in the validation cohort, it was superior to TILs and comparable with PD-L1 (AUC, 0.67; 95% CI, 0.60-0.74), with a 10-percentage point improvement in specificity. In the validation cohort, combining the deep learning model with PD-L1 scores achieved an AUC of 0.70 (95% CI, 0.63-0.76), outperforming either marker alone, with a response rate of 51% compared to 41% for PD-L1 (≥50%) alone. Conclusions and Relevance The findings of this cohort study demonstrate a strong and independent deep learning-based feature associated with ICI response in patients with NSCLC across various cohorts. Clinical use of this deep learning model could refine treatment precision and better identify patients who are likely to benefit from ICI for treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Mehrdad Rakaee
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Masoud Tafavvoghi
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Joao V. Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alessio Cortellini
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology Operative Research Unit, Fondazione Policlinico Campus Bio-Medico, Rome, Italy
- Research Unit of Medical Oncology, Department of Medicine and Surgery, Universitá Campus Bio-Medico, Rome, Italy
| | - Fabrizio Citarella
- Medical Oncology Operative Research Unit, Fondazione Policlinico Campus Bio-Medico, Rome, Italy
- Research Unit of Medical Oncology, Department of Medicine and Surgery, Universitá Campus Bio-Medico, Rome, Italy
| | - Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Università Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Università Campus Bio-Medico, Rome, Italy
| | - Elio Adib
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Cassio Murilo Hidalgo Filho
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alessandro Di Federico
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Falah Jabar
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Sayed Hashemi
- Department of Pulmonary Medicine, Cancer Center Amsterdam, VU Medical Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ilias Houda
- Department of Pulmonary Medicine, Cancer Center Amsterdam, VU Medical Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Elin Richardsen
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway
| | - Idris Bahce
- Department of Pulmonary Medicine, Cancer Center Amsterdam, VU Medical Center, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - David J. Pinato
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Åslaug Helland
- Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Division of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lynette M. Sholl
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark M. Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - David J. Kwiatkowski
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
30
|
Fountzilas E, Pearce T, Baysal MA, Chakraborty A, Tsimberidou AM. Convergence of evolving artificial intelligence and machine learning techniques in precision oncology. NPJ Digit Med 2025; 8:75. [PMID: 39890986 PMCID: PMC11785769 DOI: 10.1038/s41746-025-01471-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: 06/18/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025] Open
Abstract
The confluence of new technologies with artificial intelligence (AI) and machine learning (ML) analytical techniques is rapidly advancing the field of precision oncology, promising to improve diagnostic approaches and therapeutic strategies for patients with cancer. By analyzing multi-dimensional, multiomic, spatial pathology, and radiomic data, these technologies enable a deeper understanding of the intricate molecular pathways, aiding in the identification of critical nodes within the tumor's biology to optimize treatment selection. The applications of AI/ML in precision oncology are extensive and include the generation of synthetic data, e.g., digital twins, in order to provide the necessary information to design or expedite the conduct of clinical trials. Currently, many operational and technical challenges exist related to data technology, engineering, and storage; algorithm development and structures; quality and quantity of the data and the analytical pipeline; data sharing and generalizability; and the incorporation of these technologies into the current clinical workflow and reimbursement models.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Panorama, Thessaloniki, Greece
| | | | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA.
| |
Collapse
|
31
|
Qi Y, Hu Y, Lin C, Song G, Shi L, Zhu H. A preoperative predictive model based on multi-modal features to predict pathological complete response after neoadjuvant chemoimmunotherapy in esophageal cancer patients. Front Immunol 2025; 16:1530279. [PMID: 39958355 PMCID: PMC11827421 DOI: 10.3389/fimmu.2025.1530279] [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: 11/18/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
Background This study aimed to develop a multi-modality model by incorporating pretreatment computed tomography (CT) radiomics and pathomics features along with clinical variables to predict pathologic complete response (pCR) to neoadjuvant chemoimmunotherapy in patients with locally advanced esophageal cancer (EC). Method A total of 223 EC patients who underwent neoadjuvant chemoimmunotherapy followed by surgical intervention between August 2021 and December 2023 were included in this study. Radiomics features were extracted from contrast-enhanced CT images using PyrRadiomics, while pathomics features were derived from whole-slide images (WSIs) of pathological specimens using a fine-tuned deep learning model (ResNet-50). After feature selection, three single-modality prediction models and a combined multi-modality model integrating two radiomics features, 11 pathomics features, and two clinicopathological features were constructed using the support vector machine (SVM) algorithm. The performance of the models were evaluated using receiver operating characteristic (ROC) analysis, calibration plots, and decision curve analysis (DCA). Shapley values were also utilized to explain the prediction model. Results The predictive capability of the multi-modality model in predicting pCR yielded an area under the curve (AUC) of 0.89 (95% confidence interval [CI], 0.75-1.00), outperforming the radiomics model (AUC 0.70 [95% CI 0.54-0.85]), pathomics model (AUC 0.77 [95% CI 0.53-1.00]), and clinical model (AUC 0.63 [95% CI 0.46-0.80]). Additionally, both the calibration plot and DCA curves support the clinical utility of the integrated multi-modality model. Conclusions The combined multi-modality model we propose can better predict the pCR status of esophageal cancer and help inform clinical treatment decisions.
Collapse
Affiliation(s)
- Yana Qi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanran Hu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chengting Lin
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ge Song
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Liting Shi
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
32
|
Ding L, Fan L, Shen M, Wang Y, Sheng K, Zou Z, An H, Jiang Z. Evaluating ChatGPT's diagnostic potential for pathology images. Front Med (Lausanne) 2025; 11:1507203. [PMID: 39917264 PMCID: PMC11798939 DOI: 10.3389/fmed.2024.1507203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025] Open
Abstract
Background Chat Generative Pretrained Transformer (ChatGPT) is a type of large language model (LLM) developed by OpenAI, known for its extensive knowledge base and interactive capabilities. These attributes make it a valuable tool in the medical field, particularly for tasks such as answering medical questions, drafting clinical notes, and optimizing the generation of radiology reports. However, keeping accuracy in medical contexts is the biggest challenge to employing GPT-4 in a clinical setting. This study aims to investigate the accuracy of GPT-4, which can process both text and image inputs, in generating diagnoses from pathological images. Methods This study analyzed 44 histopathological images from 16 organs and 100 colorectal biopsy photomicrographs. The initial evaluation was conducted using the standard GPT-4 model in January 2024, with a subsequent re-evaluation performed in July 2024. The diagnostic accuracy of GPT-4 was assessed by comparing its outputs to a reference standard using statistical measures. Additionally, four pathologists independently reviewed the same images to compare their diagnoses with the model's outputs. Both scanned and photographed images were tested to evaluate GPT-4's generalization ability across different image types. Results GPT-4 achieved an overall accuracy of 0.64 in identifying tumor imaging and tissue origins. For colon polyp classification, accuracy varied from 0.57 to 0.75 in different subtypes. The model achieved 0.88 accuracy in distinguishing low-grade from high-grade dysplasia and 0.75 in distinguishing high-grade dysplasia from adenocarcinoma, with a high sensitivity in detecting adenocarcinoma. Consistency between initial and follow-up evaluations showed slight to moderate agreement, with Kappa values ranging from 0.204 to 0.375. Conclusion GPT-4 demonstrates the ability to diagnose pathological images, showing improved performance over earlier versions. Its diagnostic accuracy in cancer is comparable to that of pathology residents. These findings suggest that GPT-4 holds promise as a supportive tool in pathology diagnostics, offering the potential to assist pathologists in routine diagnostic workflows.
Collapse
Affiliation(s)
- Liya Ding
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Fan
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Pathology, Ninghai County Traditional Chinese Medicine Hospital, Ningbo, China
| | - Miao Shen
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Pathology, Deqing People’s Hospital, Hangzhou, China
| | - Yawen Wang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Kaiqin Sheng
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijuan Zou
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huimin An
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhinong Jiang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
33
|
Romeo M, Dallio M, Napolitano C, Basile C, Di Nardo F, Vaia P, Iodice P, Federico A. Clinical Applications of Artificial Intelligence (AI) in Human Cancer: Is It Time to Update the Diagnostic and Predictive Models in Managing Hepatocellular Carcinoma (HCC)? Diagnostics (Basel) 2025; 15:252. [PMID: 39941182 PMCID: PMC11817573 DOI: 10.3390/diagnostics15030252] [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: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
In recent years, novel findings have progressively and promisingly supported the potential role of Artificial intelligence (AI) in transforming the management of various neoplasms, including hepatocellular carcinoma (HCC). HCC represents the most common primary liver cancer. Alarmingly, the HCC incidence is dramatically increasing worldwide due to the simultaneous "pandemic" spreading of metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD currently constitutes the leading cause of chronic hepatic damage (steatosis and steatohepatitis), fibrosis, and liver cirrhosis, configuring a scenario where an HCC onset has been reported even in the early disease stage. On the other hand, HCC represents a serious plague, significantly burdening the outcomes of chronic hepatitis B (HBV) and hepatitis C (HCV) virus-infected patients. Despite the recent progress in the management of this cancer, the overall prognosis for advanced-stage HCC patients continues to be poor, suggesting the absolute need to develop personalized healthcare strategies further. In this "cold war", machine learning techniques and neural networks are emerging as weapons, able to identify the patterns and biomarkers that would have normally escaped human observation. Using advanced algorithms, AI can analyze large volumes of clinical data and medical images (including routinely obtained ultrasound data) with an elevated accuracy, facilitating early diagnosis, improving the performance of predictive models, and supporting the multidisciplinary (oncologist, gastroenterologist, surgeon, radiologist) team in opting for the best "tailored" individual treatment. Additionally, AI can significantly contribute to enhancing the effectiveness of metabolomics-radiomics-based models, promoting the identification of specific HCC-pathogenetic molecules as new targets for realizing novel therapeutic regimens. In the era of precision medicine, integrating AI into routine clinical practice appears as a promising frontier, opening new avenues for liver cancer research and treatment.
Collapse
Affiliation(s)
- Mario Romeo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Marcello Dallio
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Carmine Napolitano
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Claudio Basile
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Fiammetta Di Nardo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Paolo Vaia
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | | | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| |
Collapse
|
34
|
Olawade DB, Clement David-Olawade A, Adereni T, Egbon E, Teke J, Boussios S. Integrating AI into Cancer Immunotherapy-A Narrative Review of Current Applications and Future Directions. Diseases 2025; 13:24. [PMID: 39851488 PMCID: PMC11764268 DOI: 10.3390/diseases13010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Cancer remains a leading cause of morbidity and mortality worldwide. Traditional treatments like chemotherapy and radiation often result in significant side effects and varied patient outcomes. Immunotherapy has emerged as a promising alternative, harnessing the immune system to target cancer cells. However, the complexity of immune responses and tumor heterogeneity challenges its effectiveness. OBJECTIVE This mini-narrative review explores the role of artificial intelligence [AI] in enhancing the efficacy of cancer immunotherapy, predicting patient responses, and discovering novel therapeutic targets. METHODS A comprehensive review of the literature was conducted, focusing on studies published between 2010 and 2024 that examined the application of AI in cancer immunotherapy. Databases such as PubMed, Google Scholar, and Web of Science were utilized, and articles were selected based on relevance to the topic. RESULTS AI has significantly contributed to identifying biomarkers that predict immunotherapy efficacy by analyzing genomic, transcriptomic, and proteomic data. It also optimizes combination therapies by predicting the most effective treatment protocols. AI-driven predictive models help assess patient response to immunotherapy, guiding clinical decision-making and minimizing side effects. Additionally, AI facilitates the discovery of novel therapeutic targets, such as neoantigens, enabling the development of personalized immunotherapies. CONCLUSIONS AI holds immense potential in transforming cancer immunotherapy. However, challenges related to data privacy, algorithm transparency, and clinical integration must be addressed. Overcoming these hurdles will likely make AI a central component of future cancer immunotherapy, offering more personalized and effective treatments.
Collapse
Affiliation(s)
- David B. Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London E16 2RD, UK
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Public Health, York St John University, London E14 2BA, UK
| | | | - Temitope Adereni
- Department of Public Health, University of Dundee, Dundee DD1 4HN, UK;
| | - Eghosasere Egbon
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Life Science Engineering, FH Technikum, 1200 Vienna, Austria;
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Surgery, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury, Kent CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
| |
Collapse
|
35
|
Schmauch B, Cabeli V, Domingues OD, Le Douget JE, Hardy A, Belbahri R, Maussion C, Romagnoni A, Eckstein M, Fuchs F, Swalduz A, Lantuejoul S, Crochet H, Ghiringhelli F, Derangere V, Truntzer C, Pass H, Moreira AL, Chiriboga L, Zheng Y, Ozawa M, Howitt BE, Gevaert O, Girard N, Rexhepaj E, Valtingojer I, Debussche L, de Rinaldis E, Nestle F, Spanakis E, Fantin VR, Durand EY, Classe M, Von Loga K, Pronier E, Cesaroni M. Deep learning uncovers histological patterns of YAP1/TEAD activity related to disease aggressiveness in cancer patients. iScience 2025; 28:111638. [PMID: 39868035 PMCID: PMC11758823 DOI: 10.1016/j.isci.2024.111638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/08/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025] Open
Abstract
Over the last decade, Hippo signaling has emerged as a major tumor-suppressing pathway. Its dysregulation is associated with abnormal expression of YAP1 and TEAD-family genes. Recent works have highlighted the role of YAP1/TEAD activity in several cancers and its potential therapeutic implications. Therefore, identifying patients with a dysregulated Hippo pathway is key to enhancing treatment impact. Although recent studies have derived RNA-seq-based signatures, there remains a need for a reproducible and cost-effective method to measure the pathway activation. In recent years, deep learning applied to histology slides have emerged as an effective way to predict molecular information from a data modality available in clinical routine. Here, we trained models to predict YAP1/TEAD activity from H&E-stained histology slides in multiple cancers. The robustness of our approach was assessed in seven independent validation cohorts. Finally, we showed that histological markers of disease aggressiveness were associated with dysfunctional Hippo signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Markus Eckstein
- Bavarian Cancer Research Center (Bayerisches Zentrum für Krebsforschung, BZKF), Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Florian Fuchs
- Bavarian Cancer Research Center (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aurélie Swalduz
- Claude Bernard University Lyon I & Léon Bérard Cancer Center, Lyon, France
| | - Sylvie Lantuejoul
- Grenoble Alpes University and Léon Bérard Cancer Center, Lyon, 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
| | - 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
- University of Burgundy Franche-Comté, Dijon, France
| | - Harvey Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - Andre L. Moreira
- Department of Pathology, NYU Langone New York University Langone Medical Center, New York, NY, USA
| | - Luis Chiriboga
- Department of Pathology, NYU Langone New York University Langone Medical Center, New York, NY, USA
| | - Yuanning Zheng
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Michael Ozawa
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Brooke E. Howitt
- Department of Medicine & Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Olivier Gevaert
- Department of Medicine & Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Jiang Y, Wang B, Xing W, Huo C. HCTTI: High-Performance Heterogeneous Computing Toolkit for Tissue Image Stain Normalization. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01398-6. [PMID: 39821779 DOI: 10.1007/s10278-025-01398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Whole slide imaging (WSI) has transformed diagnostic medicine, particularly in the field of cancer diagnosis and treatment. The use of deep learning algorithms for predicting WSIs has opened up new avenues for advanced medical diagnostics. Additionally, stain normalization can reduce the color and intensity variations present in WSI from different hospitals. As a result, deep learning classification accuracy improves. However, WSI reading and color normalization are still largely performed by using CPUs, leading to sub-optimal performance. We proposed a High-Performance Heterogeneous Computing Toolkit for Tissue Image (HCTTI) that integrates multiple computer system-level optimizations and encompasses WSI reading, tile normalization, and tile saving. We explored the potential advantages and limitations of different WSI readers and color normalization techniques in WSI analysis and the performance of different tile serialization formats for saving tiles. We found that HCTTI is 7 × faster than OpenSlide for reading WSIs, GPU implementation of the Macenko normalization algorithm is 9 × faster than TIAToolbox implementation, and HDF5 is faster than png and Zarr for storing normalized images in both writing (13 × acceleration compared to png) and reading (2 × acceleration compared to png), Specifically, HDF5 provides superior performance in handling large, complex datasets due to its efficient chunking and compression capabilities, as well as its broad support for hierarchical data management, making it very suitable for workloads like deep learning training I/O pattern that involves randomly reading large amount of small files in each training epoch. We also achieved linear acceleration in our multi-node distributed GPU implementation. To our knowledge, HCTTI is the first comprehensive toolkit that comprises distributed WSI reading, normalization, and serialization. It is 13 × speedup compared to TIAToolbox implementation for normalizing a single WSI. Our findings could help pave the way for more effective and efficient deep learning-based approaches to WSI analysis, with the potential to transform medical diagnosis and treatment for a wide range of conditions. The source code of HCTTI is available at https://github.com/wangbo00129/HCTTI .
Collapse
Affiliation(s)
- Yan Jiang
- School of Software, Quanzhou University of Information Engineering, Quanzhou, Fujian, 362000, China.
- Science Department, Geneis (Beijing) Co. Ltd., Beijing, 100102, P. R. China.
- School of Information Engineering, Changsha Medical University, Changsha, 410219, China.
| | - Bo Wang
- Science Department, Geneis (Beijing) Co. Ltd., Beijing, 100102, P. R. China
| | - Weipeng Xing
- School of Electrical & Information Engineering, Anhui University of Technology, Maanshan, Anhui, 243002, China
| | - Cheng Huo
- Department of Pathology, Sinopharm Tongmei General Hospital, Datong, 037003, Shanxi, China
| |
Collapse
|
37
|
Ren H, Song H, Cui S, Xiong H, Long B, Li Y. Deep learning of noncontrast CT for fast prediction of hemorrhagic transformation of acute ischemic stroke: a multicenter study. Eur Radiol Exp 2025; 9:8. [PMID: 39812734 PMCID: PMC11735721 DOI: 10.1186/s41747-024-00535-0] [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: 07/02/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Hemorrhagic transformation (HT) is a complication of reperfusion therapy following acute ischemic stroke (AIS). We aimed to develop and validate a model for predicting HT and its subtypes with poor prognosis-parenchymal hemorrhage (PH), including PH-1 (hematoma within infarcted tissue, occupying < 30%) and PH-2 (hematoma occupying ≥ 30% of the infarcted tissue)-in AIS patients following intravenous thrombolysis (IVT) based on noncontrast computed tomography (NCCT) and clinical data. METHODS In this six-center retrospective study, clinical and imaging data from 445 consecutive IVT-treated AIS patients were collected (01/2018-06/2023). The training cohort comprised 344 patients from five centers, and the test cohort included 101 patients from the sixth center. A clinical model was developed using eXtreme Gradient Boosting, an NCCT-based imaging model was created using deep learning, and an ensemble model integrated both models. Comparison with existing clinical scores (MSS, SEDAN, GRASPS) was performed using the DeLong test. RESULTS Of the 445 individuals, 202 (45.4%) had HT, 79 (17.8%) had hemorrhagic infarction, and 123 (27.6%) had PH. In the test cohort, the area under the receiver operating characteristic curve (AUROC) of the clinical, imaging, and ensemble model for HT prediction was 0.877, 0.920, and 0.937, respectively. The ensemble model for HT prediction outperformed MSS, SEDAN, and GRASPS scores (p ≤ 0.023). The ensemble model predicted PH and PH-2 with AUROC of 0.858 and 0.806, respectively. CONCLUSION Developing and validating an integrated model that can predict HT and its subtypes in AIS patients following IVT based on NCCT and clinical data is feasible. RELEVANCE STATEMENT The clinical, imaging, and ensemble models based on noncontrast CT and clinical data outperformed existing clinical scores in predicting hemorrhagic transformation of AIS and its subtypes with poor prognosis, facilitating personalized treatment decisions. KEY POINTS The models demonstrated the capability to predict hemorrhagic transformation of acute ischemic stroke quickly, accurately, and reliably. The proposed models outperformed existing clinical scores in predicting hemorrhagic transformation. The ensemble model provided risk assessment of parenchymal hemorrhage and parenchymal hemorrhage-2 outperforming existing clinical scores.
Collapse
Affiliation(s)
- Huanhuan Ren
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, China
| | - Haojie Song
- College of Computer and Information Science, Chongqing Normal University, Chongqing, China
| | - Shaoguo Cui
- College of Computer and Information Science, Chongqing Normal University, Chongqing, China
| | - Hua Xiong
- Department of Radiology, Shapingba Hospital affiliated to Chongqing University (Shapingba District People's Hospital of Chongqing), Chongqing, China
| | - Bangyuan Long
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
38
|
Hu K, Wu Y, Huang Y, Zhou M, Wang Y, Huang X. Annotation-free deep learning algorithm trained on hematoxylin & eosin images predicts epithelial-to-mesenchymal transition phenotype and endocrine response in estrogen receptor-positive breast cancer. Breast Cancer Res 2025; 27:6. [PMID: 39800743 PMCID: PMC11725188 DOI: 10.1186/s13058-025-01959-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
Recent evidence indicates that endocrine resistance in estrogen receptor-positive (ER+) breast cancer is closely correlated with phenotypic characteristics of epithelial-to-mesenchymal transition (EMT). Nonetheless, identifying tumor tissues with a mesenchymal phenotype remains challenging in clinical practice. In this study, we validated the correlation between EMT status and resistance to endocrine therapy in ER+ breast cancer from a transcriptomic perspective. To confirm the presence of morphological discrepancies in tumor tissues of ER+ breast cancer classified as epithelial- and mesenchymal-phenotypes according to EMT-related transcriptional features, we trained deep learning algorithms based on EfficientNetV2 architecture to assign the phenotypic status for each patient utilizing hematoxylin & eosin (H&E)-stained slides from The Cancer Genome Atlas database. Our classifier model accurately identified the precise phenotypic status, achieving an area under the curve (AUC) of 0.886 at the tile-level and an AUC of 0.910 at the slide-level. Furthermore, we evaluated the efficacy of the classifier in predicting endocrine response using data from an independent ER+ breast cancer patient cohort. Our classifier achieved a predicting accuracy of 81.25%, and 88.7% slides labeled as endocrine resistant were predicted as the mesenchymal-phenotype, while 75.6% slides labeled as sensitive were predicted as the epithelial-phenotype. Our work introduces an H&E-based framework capable of accurately predicting EMT phenotype and endocrine response for ER+ breast cancer, demonstrating its potential for clinical application and benefit.
Collapse
Affiliation(s)
- Kaimin Hu
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinan Wu
- Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China
| | - Yajing Huang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meiqi Zhou
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanyan Wang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingru Huang
- School of Electronic Engineering and Computer Science, Queen Mary University of London, London, UK.
| |
Collapse
|
39
|
Žigutytė L, Lenz T, Han T, Hewitt KJ, Reitsam NG, Foersch S, Carrero ZI, Unger M, Pearson AT, Truhn D, Kather JN. Counterfactual Diffusion Models for Mechanistic Explainability of Artificial Intelligence Models in Pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.29.620913. [PMID: 39554184 PMCID: PMC11565818 DOI: 10.1101/2024.10.29.620913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Background Deep learning can extract predictive and prognostic biomarkers from histopathology whole slide images, but its interpretability remains elusive. Methods We develop and validate MoPaDi (Morphing histoPathology Diffusion), which generates counterfactual mechanistic explanations. MoPaDi uses diffusion autoencoders to manipulate pathology image patches and flip their biomarker status by changing the morphology. Importantly, MoPaDi includes multiple instance learning for weakly supervised problems. We validate our method on four datasets classifying tissue types, cancer types within different organs, center of slide origin, and a biomarker - microsatellite instability. Counterfactual transitions were evaluated through pathologists' user studies and quantitative cell analysis. Results MoPaDi achieves excellent image reconstruction quality (multiscale structural similarity index measure 0.966-0.992) and good classification performance (AUCs 0.76-0.98). In a blinded user study for tissue-type counterfactuals, counterfactual images were realistic (63.3-73.3% of original images identified correctly). For other tasks, pathologists identified meaningful morphological features from counterfactual images. Conclusion MoPaDi generates realistic counterfactual explanations that reveal key morphological features driving deep learning model predictions in histopathology, improving interpretability.
Collapse
Affiliation(s)
- Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Tim Lenz
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Tianyu Han
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Germany
| | - Katherine J Hewitt
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Nic G Reitsam
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Germany
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Michaela Unger
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health (EKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
40
|
Wu Z, Wang T, Lan J, Wang J, Chen G, Tong T, Zhang H. Deep learning-based prediction of HER2 status and trastuzumab treatment efficacy of gastric adenocarcinoma based on morphological features. J Transl Med 2025; 23:13. [PMID: 39762854 PMCID: PMC11702172 DOI: 10.1186/s12967-024-06034-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND First-line treatment for advanced gastric adenocarcinoma (GAC) with human epidermal growth factor receptor 2 (HER2) is trastuzumab combined with chemotherapy. In clinical practice, HER2 positivity is identified through immunohistochemistry (IHC) or fluorescence in situ hybridization (FISH), whereas deep learning (DL) can predict HER2 status based on tumor histopathological features. However, it remains uncertain whether these deep learning-derived features can predict the efficacy of anti-HER2 therapy. METHODS We analyzed a cohort of 300 consecutive surgical specimens and 101 biopsy specimens, all undergoing HER2 testing, along with 41 biopsy specimens receiving trastuzumab-based therapy for HER2-positive GAC. RESULTS We developed a convolutional neural network (CNN) model using surgical specimens that achieved an area under the curve (AUC) value of 0.847 in predicting HER2 amplification, and achieved an AUC of 0.903 in predicting HER2 status specifically in patients with HER2 2 + expression. The model also predicted HER2 status in gastric biopsy specimens, achieving an AUC of 0.723. Furthermore, our classifier was trained using 41 HER2-positive gastric biopsy specimens that had undergone trastuzumab treatment, our model demonstrated an AUC of 0.833 for the (CR + PR) / (SD + PD) subgroup. CONCLUSION This work explores an algorithm that utilizes hematoxylin and eosin (H&E) staining to accurately predict HER2 status and assess the response to trastuzumab in GAC, potentially facilitating clinical decision-making.
Collapse
Affiliation(s)
- Zhida Wu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Tao Wang
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China
| | - Junlin Lan
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Tong Tong
- College of Physics and Information Engineering, Fuzhou University, Xueyuan Road No.2, Fuzhou, Fujian, 350108, China.
| | - Hejun Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
41
|
Chang L, Liu J, Zhu J, Guo S, Wang Y, Zhou Z, Wei X. Advancing precision medicine: the transformative role of artificial intelligence in immunogenomics, radiomics, and pathomics for biomarker discovery and immunotherapy optimization. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0376. [PMID: 39749734 PMCID: PMC11795263 DOI: 10.20892/j.issn.2095-3941.2024.0376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Artificial intelligence (AI) is significantly advancing precision medicine, particularly in the fields of immunogenomics, radiomics, and pathomics. In immunogenomics, AI can process vast amounts of genomic and multi-omic data to identify biomarkers associated with immunotherapy responses and disease prognosis, thus providing strong support for personalized treatments. In radiomics, AI can analyze high-dimensional features from computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT) images to discover imaging biomarkers associated with tumor heterogeneity, treatment response, and disease progression, thereby enabling non-invasive, real-time assessments for personalized therapy. Pathomics leverages AI for deep analysis of digital pathology images, and can uncover subtle changes in tissue microenvironments, cellular characteristics, and morphological features, and offer unique insights into immunotherapy response prediction and biomarker discovery. These AI-driven technologies not only enhance the speed, accuracy, and robustness of biomarker discovery but also significantly improve the precision, personalization, and effectiveness of clinical treatments, and are driving a shift from empirical to precision medicine. Despite challenges such as data quality, model interpretability, integration of multi-modal data, and privacy protection, the ongoing advancements in AI, coupled with interdisciplinary collaboration, are poised to further enhance AI's roles in biomarker discovery and immunotherapy response prediction. These improvements are expected to lead to more accurate, personalized treatment strategies and ultimately better patient outcomes, marking a significant step forward in the evolution of precision medicine.
Collapse
Affiliation(s)
- Luchen Chang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiamei Liu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuyue Guo
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yao Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhiwei Zhou
- Departments of Biochemistry and Radiation Oncology, UT Southwestern Medical Center, Dallas 75390, USA
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
42
|
Song Y, Liu S, Liu X, Jia H, Shi H, Liu X, Hao D, Wang H, Xing X. An Integrated Radiopathomics Machine Learning Model to Predict Pathological Response to Preoperative Chemotherapy in Gastric Cancer. Acad Radiol 2025; 32:134-145. [PMID: 39214816 DOI: 10.1016/j.acra.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
RATIONALE AND OBJECTIVES Accurately predicting the pathological response to chemotherapy before treatment is important for selecting the appropriate treatment groups, formulating individualized treatment plans, and improving the survival rates of patients with gastric cancer (GC). METHODS We retrospectively enrolled 151 patients diagnosed with GC who underwent preoperative chemotherapy and surgical resection at the Affiliated Hospital of Qingdao University between January 2015 and June 2023. Both pretreatment-enhanced computer technology images and whole slide images of pathological hematoxylin and eosin-stained sections were available for each patient. The image features were extracted and used to construct an ensemble radiopathomics machine learning model. In addition, a nomogram was developed by combining the imaging features and clinical characteristics. RESULTS In total, 962 radiomics and 999 pathomics signatures were extracted from 106 patients in the training cohort. A fusion radiopathomics model was constructed using 13 radiomics and 5 pathomics signatures. The fusion model showed favorable performance compared to single-omics models, with an area under the curve (AUC) of 0.789 in the validation cohort. Moreover, a combined radiopathomics nomogram (RPN) was developed based on radiopathomics features and the Borrmann type, which is a classification method for advanced GC according to tumor growth pattern and gross morphology. The RPN showed superior predictive performance in the training (AUC 0.880) and validation cohorts (AUC 0.797). The decision curve analysis showed that RPN could provide favorable clinical benefits to patients with GC. CONCLUSIONS RPN was able to predict the pathological response to preoperative chemotherapy with high accuracy, and therefore provides a novel tool for personalized treatment of GC.
Collapse
Affiliation(s)
- Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xinyu Liu
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Huiqing Jia
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Hailei Shi
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xianglan Liu
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Dapeng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Xiaoming Xing
- Department of Pathology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China.
| |
Collapse
|
43
|
Ying Y, Ju R, Wang J, Li W, Ji Y, Shi Z, Chen J, Chen M. Accuracy of machine learning in diagnosing microsatellite instability in gastric cancer: A systematic review and meta-analysis. Int J Med Inform 2025; 193:105685. [PMID: 39515046 DOI: 10.1016/j.ijmedinf.2024.105685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Significant challenges persist in the early identification of microsatellite instability (MSI) within current clinical practice. In recent years, with the growing utilization of machine learning (ML) in the diagnosis and management of gastric cancer (GC), numerous researchers have explored the effectiveness of ML methodologies in detecting MSI. Nevertheless, the predictive value of these approaches still lacks comprehensive evidence. Accordingly, this study was carried out to consolidate the accuracy of ML in the prompt detection of MSI in GC. METHODS PubMed, the Cochrane Library, the Web of Science, and Embase were retrieved up to March 20, 2024. The risk of bias in the encompassed studies was evaluated utilizing a risk assessment tool for predictive models. Models were then subjected to subgroup analysis based on the modeling variables. RESULTS A total of 12 studies, encompassing 11,912 patients with GC, satisfied the predefined inclusion criteria. ML models established in these studies were primarily based on pathological images, clinical features, and radiomics. The results suggested that in the validation sets, the pathological image-based models had a synthesized c-index of 0.86 [95 % CI (0.83-0.89)], with sensitivity and specificity being 0.86 [95 % CI (0.76-0.92)] and 0.83 [95 % CI (0.78-0.87)], respectively; radiomics feature-based models achieved respective values of 0.87 [95 % CI (0.81-0.92)], 0.77 [95 % CI (0.70-0.83)] and 0.81 [95 % CI (0.74-0.87)]; radiomics feature-based models + clinical feature-based models achieved respective values of 0.87 [95 % CI (0.81-0.93)], 0.78 [95 % CI (0.70-0.84)] and 0.79 [95 % CI (0.69-0.86)]. CONCLUSIONS ML has demonstrated optimal performance in detecting MSI in GC and could serve as a prospective early adjunctive detection tool for MSI in GC. Future research should contemplate minimally invasive or non-invasive, readily collectible, and efficient predictors to augment the predictive accuracy of ML.
Collapse
Affiliation(s)
- Yuou Ying
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Ruyi Ju
- Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jieyi Wang
- The Basic Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Wenkai Li
- Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Yuan Ji
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Zhenyu Shi
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jinhan Chen
- The Second Affiliated College of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Mingxian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Street Gucui No. 234, Region Xihu, Hangzhou 310012, Zhejiang Province, China.
| |
Collapse
|
44
|
Cai XJ, Peng CR, Cui YY, Li L, Huang MW, Zhang HY, Zhang JY, Li TJ. Identification of genomic alteration and prognosis using pathomics-based artificial intelligence in oral leukoplakia and head and neck squamous cell carcinoma: a multicenter experimental study. Int J Surg 2025; 111:426-438. [PMID: 39248300 PMCID: PMC11745750 DOI: 10.1097/js9.0000000000002077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Loss of chromosome 9p is an important biomarker in the malignant transformation of oral leukoplakia (OLK) to head and neck squamous cell carcinoma (HNSCC), and is associated with the prognosis of HNSCC patients. However, various challenges have prevented 9p loss from being assessed in clinical practice. The objective of this study was to develop a pathomics-based artificial intelligence (AI) model for the rapid and cost-effective prediction of 9p loss (9PLP). MATERIALS AND METHODS Three hundred thirty-three OLK cases were retrospectively collected with hematoxylin and eosin (H&E)-stained whole slide images and genomic alteration data from multicenter cohorts to develop the genomic alteration prediction AI model. They were divided into a training dataset ( n =217), a validation dataset ( n =93), and an external testing dataset ( n =23). The latest Transformer method and XGBoost algorithm were combined to develop the 9PLP model. The AI model was further applied and validated in two multicenter HNSCC datasets ( n =42 and n =365, respectively). Moreover, the combination of 9PLP with clinicopathological parameters was used to develop a nomogram model for assessing HNSCC patient prognosis. RESULTS 9PLP could predict chromosome 9p loss rapidly and effectively using both OLK and HNSCC images, with the area under the curve achieving 0.890 and 0.825, respectively. Furthermore, the predictive model showed high accuracy in HNSCC patient prognosis assessment (the area under the curve was 0.739 for 1-year prediction, 0.705 for 3-year prediction, and 0.691 for 5-year prediction). CONCLUSION To the best of our knowledge, this study developed the first genomic alteration prediction deep learning model in OLK and HNSCC. This novel AI model could predict 9p loss and assess patient prognosis by identifying pathomics features in H&E-stained images with good performance. In the future, the 9PLP model may potentially contribute to better clinical management of OLK and HNSCC.
Collapse
Affiliation(s)
- Xin-Jia Cai
- Central Laboratory, Peking University School and Hospital of Stomatology
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
| | - Chao-Ran Peng
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Ying-Ying Cui
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Long Li
- Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital, Central South University, Changsha, People’s Republic of China
| | - Ming-Wei Huang
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing
| | - He-Yu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
| | - Jian-Yun Zhang
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
| | - Tie-Jun Li
- National Center of Stomatology and National Clinical Research Center for Oral Diseases and National Engineering Research Center of Oral Biomaterials and Digital Medical Devices
- Department of Oral Pathology, Peking University School and Hospital of Stomatology
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034)
| |
Collapse
|
45
|
Syrykh C, van den Brand M, Kather JN, Laurent C. Role of artificial intelligence in haematolymphoid diagnostics. Histopathology 2025; 86:58-68. [PMID: 39435690 DOI: 10.1111/his.15327] [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: 10/23/2024]
Abstract
The advent of digital pathology and the deployment of high-throughput molecular techniques are generating an unprecedented mass of data. Thanks to advances in computational sciences, artificial intelligence (AI) approaches represent a promising avenue for extracting relevant information from complex data structures. From diagnostic assistance to powerful research tools, the potential fields of application of machine learning techniques in pathology are vast and constitute the subject of considerable research work. The aim of this article is to provide an overview of the potential applications of AI in the field of haematopathology and to define the role that these emerging technologies could play in our laboratories in the short to medium term.
Collapse
Affiliation(s)
- Charlotte Syrykh
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
| | - Michiel van den Brand
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Pathology-DNA, Arnhem, The Netherlands
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Camille Laurent
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
- INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| |
Collapse
|
46
|
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
|
47
|
Wang L, Chen L, Wei K, Zhou H, Zwiggelaar R, Fu W, Liu Y. Weakly supervised pathological differentiation of primary central nervous system lymphoma and glioblastoma on multi-site whole slide images. J Med Imaging (Bellingham) 2025; 12:017502. [PMID: 39802317 PMCID: PMC11724367 DOI: 10.1117/1.jmi.12.1.017502] [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: 06/10/2024] [Revised: 09/29/2024] [Accepted: 11/01/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Differentiating primary central nervous system lymphoma (PCNSL) and glioblastoma (GBM) is crucial because their prognosis and treatment differ substantially. Manual examination of their histological characteristics is considered the golden standard in clinical diagnosis. However, this process is tedious and time-consuming and might lead to misdiagnosis caused by morphological similarity between their histology and tumor heterogeneity. Existing research focuses on radiological differentiation, which mostly uses multi-parametric magnetic resonance imaging. By contrast, we investigate the pathological differentiation between the two types of tumors using whole slide images (WSIs) of postoperative formalin-fixed paraffin-embedded samples. Approach To learn the specific and intrinsic histological feature representations from the WSI patches, a self-supervised feature extractor is trained. Then, the patch representations are fused by feeding into a weakly supervised multiple-instance learning model for the WSI classification. We validate our approach on 134 PCNSL and 526 GBM cases collected from three hospitals. We also investigate the effect of feature extraction on the final prediction by comparing the performance of applying the feature extractors trained on the PCNSL/GBM slides from specific institutions, multi-site PCNSL/GBM slides, and large-scale histopathological images. Results Different feature extractors perform comparably with the overall area under the receiver operating characteristic curve value exceeding 85% for each dataset and close to 95% for the combined multi-site dataset. Using the institution-specific feature extractors generally obtains the best overall prediction with both of the PCNSL and GBM classification accuracies reaching 80% for each dataset. Conclusions The excellent classification performance suggests that our approach can be used as an assistant tool to reduce the pathologists' workload by providing an accurate and objective second diagnosis. Moreover, the discriminant regions indicated by the generated attention heatmap improve the model interpretability and provide additional diagnostic information.
Collapse
Affiliation(s)
- Liping Wang
- Shandong Normal University, School of Information Science and Engineering, Jinan, China
| | - Lin Chen
- The Affiliated Hospital of Southwest Medical University, Department of Neurosurgery, Luzhou, China
| | - Kaixi Wei
- The Affiliated Hospital of Southwest Medical University, Department of Neurosurgery, Luzhou, China
- Hejiang County Traditional Chinese Medicine Hospital, Department of Neurosurgery, Luzhou, China
| | - Huiyu Zhou
- University of Leicester, School of Computing and Mathematical Sciences, Leicester, United Kingdom
| | - Reyer Zwiggelaar
- Aberystwyth University, Department of Computer Science, Aberystwyth, United Kingdom
| | - Weiwei Fu
- The Affiliated Hospital of Qingdao University, Department of Pathology, Qingdao, China
| | - Yingchao Liu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Department of Neurosurgery, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Institute of Brain Science and Brain-inspired Research, Jinan, China
| |
Collapse
|
48
|
Tafenzi HA, Essaadi I, Belbaraka R. Digital Oncology in Morocco: Embracing Artificial Intelligence in a New Era. JCO Glob Oncol 2025; 11:e2400583. [PMID: 39787448 DOI: 10.1200/go-24-00583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Affiliation(s)
- Hassan Abdelilah Tafenzi
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | - Ismail Essaadi
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
- Medical Oncology Department, Avicenna Military Hospital of Marrakech, Marrakech, Morocco
| | - Rhizlane Belbaraka
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
49
|
El Nahhas OSM, van Treeck M, Wölflein G, Unger M, Ligero M, Lenz T, Wagner SJ, Hewitt KJ, Khader F, Foersch S, Truhn D, Kather JN. From whole-slide image to biomarker prediction: end-to-end weakly supervised deep learning in computational pathology. Nat Protoc 2025; 20:293-316. [PMID: 39285224 DOI: 10.1038/s41596-024-01047-2] [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: 12/17/2023] [Accepted: 07/04/2024] [Indexed: 01/11/2025]
Abstract
Hematoxylin- and eosin-stained whole-slide images (WSIs) are the foundation of diagnosis of cancer. In recent years, development of deep learning-based methods in computational pathology has enabled the prediction of biomarkers directly from WSIs. However, accurately linking tissue phenotype to biomarkers at scale remains a crucial challenge for democratizing complex biomarkers in precision oncology. This protocol describes a practical workflow for solid tumor associative modeling in pathology (STAMP), enabling prediction of biomarkers directly from WSIs by using deep learning. The STAMP workflow is biomarker agnostic and allows for genetic and clinicopathologic tabular data to be included as an additional input, together with histopathology images. The protocol consists of five main stages that have been successfully applied to various research problems: formal problem definition, data preprocessing, modeling, evaluation and clinical translation. The STAMP workflow differentiates itself through its focus on serving as a collaborative framework that can be used by clinicians and engineers alike for setting up research projects in the field of computational pathology. As an example task, we applied STAMP to the prediction of microsatellite instability (MSI) status in colorectal cancer, showing accurate performance for the identification of tumors high in MSI. Moreover, we provide an open-source code base, which has been deployed at several hospitals across the globe to set up computational pathology workflows. The STAMP workflow requires one workday of hands-on computational execution and basic command line knowledge.
Collapse
Affiliation(s)
- Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- StratifAI GmbH, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Georg Wölflein
- School of Computer Science, University of St Andrews, St Andrews, UK
| | - Michaela Unger
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tim Lenz
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Sophia J Wagner
- Helmholtz Munich-German Research Center for Environment and Health, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Katherine J Hewitt
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Firas Khader
- StratifAI GmbH, Dresden, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Sebastian Foersch
- Institute of Pathology-University Medical Center Mainz, Mainz, Germany
| | - Daniel Truhn
- StratifAI GmbH, Dresden, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- StratifAI GmbH, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
- Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
50
|
Komura D, Ochi M, Ishikawa S. Machine learning methods for histopathological image analysis: Updates in 2024. Comput Struct Biotechnol J 2024; 27:383-400. [PMID: 39897057 PMCID: PMC11786909 DOI: 10.1016/j.csbj.2024.12.033] [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: 09/30/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
The combination of artificial intelligence and digital pathology has emerged as a transformative force in healthcare and biomedical research. As an update to our 2018 review, this review presents comprehensive analysis of machine learning applications in histopathological image analysis, with focus on the developments since 2018. We highlight significant advances that have expanded the technical capabilities and practical applications of computational pathology. The review examines progress in addressing key challenges in the field as follows: processing of gigapixel whole slide images, insufficient labeled data, multidimensional analysis, domain shifts across institutions, and interpretability of machine learning models. We evaluate emerging trends, such as foundation models and multimodal integration, that are reshaping the field. Overall, our review highlights the potential of machine learning in enhancing both routine pathological analysis and scientific discovery in pathology. By providing this comprehensive overview, this review aims to guide researchers and clinicians in understanding the current state of the pathology image analysis field and its future trajectory.
Collapse
Affiliation(s)
- Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mieko Ochi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|