1
|
Angeloni M, Rizzi D, Schoen S, Caputo A, Merolla F, Hartmann A, Ferrazzi F, Fraggetta F. Closing the gap in the clinical adoption of computational pathology: a standardized, open-source framework to integrate deep-learning models into the laboratory information system. Genome Med 2025; 17:60. [PMID: 40420213 DOI: 10.1186/s13073-025-01484-y] [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: 10/25/2024] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Digital pathology (DP) has revolutionized cancer diagnostics and enabled the development of deep-learning (DL) models aimed at supporting pathologists in their daily work and improving patient care. However, the clinical adoption of such models remains challenging. Here, we describe a proof-of-concept framework that, leveraging Health Level 7 (HL7) standard and open-source DP resources, allows a seamless integration of both publicly available and custom developed DL models in the clinical workflow. METHODS Development and testing of the framework were carried out in a fully digitized Italian pathology department. A Python-based server-client architecture was implemented to interconnect through HL7 messaging the anatomic pathology laboratory information system (AP-LIS) with an external artificial intelligence-based decision support system (AI-DSS) containing 16 pre-trained DL models. Open-source toolboxes for DL model deployment were used to run DL model inference, and QuPath was used to provide an intuitive visualization of model predictions as colored heatmaps. RESULTS A default deployment mode runs continuously in the background as each new slide is digitized, choosing the correct DL model(s) on the basis of the tissue type and staining. In addition, pathologists can initiate the analysis on-demand by selecting a specific DL model from the virtual slide tray. In both cases, the AP-LIS transmits an HL7 message to the AI-DSS, which processes the message, runs DL model inference, and creates the appropriate visualization style for the employed classification model. The AI-DSS transmits model inference results to the AP-LIS, where pathologists can visualize the output in QuPath and/or directly as slide description in the virtual slide tray. CONCLUSIONS Taken together, the developed integration framework through the use of the HL7 standard and freely available DP resources offers a standardized, portable, and open-source solution that lays the groundwork for the future widespread adoption of DL models in pathology diagnostics.
Collapse
Affiliation(s)
- Miriam Angeloni
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | | | - Simon Schoen
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alessandro Caputo
- Department of Pathology, University Hospital of Salerno, Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstr. 8-10, Erlangen, 91054, Germany.
| | - Filippo Fraggetta
- Unit of Pathology, Gravina Hospital, Via Portosalvo 1, Caltagirone, 95041, Italy.
| |
Collapse
|
2
|
Park S, Pettigrew MF, Cha YJ, Kim IH, Kim M, Banerjee I, Barnfather I, Clemenceau JR, Jang I, Kim H, Kim Y, Pai RK, Park JH, Samadder NJ, Song KY, Sung JY, Cheong JH, Kang J, Lee SH, Wang SC, Hwang TH. Deep Gaussian process with uncertainty estimation for microsatellite instability and immunotherapy response prediction from histology. NPJ Digit Med 2025; 8:294. [PMID: 40389599 PMCID: PMC12089473 DOI: 10.1038/s41746-025-01580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/18/2025] [Indexed: 05/21/2025] Open
Abstract
Determining tumor microsatellite status has significant clinical value because tumors that are microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) respond well to immune checkpoint inhibitors (ICIs) and oftentimes not to chemotherapeutics. We propose MSI-SEER, a deep Gaussian process-based Bayesian model that analyzes H&E whole-slide images in weakly-supervised-learning to predict microsatellite status in gastric and colorectal cancers. We performed extensive validation using multiple large datasets comprised of patients from diverse racial backgrounds. MSI-SEER achieved state-of-the-art performance with MSI prediction by integrating uncertainty prediction. We achieved high accuracy for predicting ICI responsiveness by combining tumor MSI status with stroma-to-tumor ratio. Finally, MSI-SEER's tile-level predictions revealed novel insights into the role of spatial distribution of MSI-H regions in the tumor microenvironment and ICI response.
Collapse
Affiliation(s)
- Sunho Park
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Morgan F Pettigrew
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - In-Ho Kim
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Minji Kim
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Imon Banerjee
- Department of Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Isabel Barnfather
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | | | - Inyeop Jang
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Jeong Hwan Park
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - N Jewel Samadder
- Department of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, AZ, USA
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji-Youn Sung
- Department of Pathology, College of Medicine, Kyung Hee University hospital, Kyung Hee University, Seoul, Korea
| | - Jae-Ho Cheong
- Department of Surgery, Department of Biochemistry and Molecular Biology, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea.
| | - Jeonghyun Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Sam C Wang
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Tae Hyun Hwang
- Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
3
|
Dutta K, Pal D, Li S, Shyam C, Shoghi KI. Corr-A-Net: Interpretable Attention-Based Correlated Feature Learning framework for predicting of HER2 Score in Breast Cancer from H&E Images. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.22.25326227. [PMID: 40313277 PMCID: PMC12045401 DOI: 10.1101/2025.04.22.25326227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is a critical biomarker for assessing breast cancer (BC) severity and guiding targeted anti-HER2 therapies. The standard method for measuring HER2 expression is manual assessment of IHC slides by pathologists, which is both time intensive and prone to inter- and intra-observer variability. To address these challenges, we developed an interpretable deep-learning pipeline with Correlational Attention Neural Network (Corr-A-Net) to predict HER2 score from H&E images. Each prediction was accompanied with a confidence score generated by the surrogate confidence score estimation network trained using incentivized mechanism. The shared correlated representations generated using the attention mechanism of Corr-A-Net achieved the best predictive accuracy of 0.93 and AUC-ROC of 0.98. Additionally, correlated representations demonstrated the highest mean effective confidence (MEC) score of 0.85 indicating robust confidence level estimation for prediction. The Corr-A-Net can have profound implications in facilitating prediction of HER2 status from H&E images.
Collapse
Affiliation(s)
- Kaushik Dutta
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Debojyoti Pal
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Suya Li
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Chandresh Shyam
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
| | - Kooresh I. Shoghi
- Imaging Science Program, Washington University in St Louis, St Louis, MO USA
- Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO USA
- Department of Biomedical Engineering, Washington University in St Louis, St Louis, MO USA
- Lead contact
| |
Collapse
|
4
|
Nguyen T, Panwar V, Jamale V, Perny A, Dusek C, Cai Q, Kapur P, Danuser G, Rajaram S. Autonomous learning of pathologists' cancer grading rules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643999. [PMID: 40166226 PMCID: PMC11956981 DOI: 10.1101/2025.03.18.643999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Deep learning (DL) algorithms have demonstrated remarkable proficiency in histopathology classification tasks, presenting an opportunity to discover disease-related features escaping visual inspection. However, the "black box" nature of DL obfuscates the basis of the classification. Here, we develop an algorithm for interpretable Deep Learning (IDL) that sheds light on the links between tissue morphology and cancer biology. We make use of a generative model trained to represent images via a combination of a semantic latent space and a noise vector to capture low level image details. We traversed the latent space so as to induce prototypical image changes associated with the disease state, which we identified via a second DL model. Applied to a dataset of clear cell renal cell carcinoma (ccRCC) tissue images the AI system pinpoints nuclear size and nucleolus density in tumor cells (but not other cell types) as the decisive features of tumor progression from grade 1 to grade 4 - mirroring the rules that have been used for decades in the clinic and are taught in textbooks. Moreover, the AI system posits a decrease in vasculature with increasing grade. While the association has been illustrated by some previous reports, the correlation is not part of currently implemented grading systems. These results indicate the potential of IDL to autonomously formalize the connection between the histopathological presentation of a disease and the underlying tissue architectural drivers.
Collapse
Affiliation(s)
- Thuong Nguyen
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vandana Panwar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vipul Jamale
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Averi Perny
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cecilia Dusek
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qi Cai
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Satwik Rajaram
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Tiwari A, Ghose A, Hasanova M, Faria SS, Mohapatra S, Adeleke S, Boussios S. The current landscape of artificial intelligence in computational histopathology for cancer diagnosis. Discov Oncol 2025; 16:438. [PMID: 40167870 PMCID: PMC11961855 DOI: 10.1007/s12672-025-02212-z] [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: 12/15/2024] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Artificial intelligence (AI) marks a frontier in histopathologic analysis shift towards the clinic, becoming a mainstream choice to interpret histological images. Surveying studies assessing AI applications in histopathology from 2013 to 2024, we review key methods (including supervised, unsupervised, weakly supervised and transfer learning) in deep learning-based pattern recognition in computational histopathology for diagnostic and prognostic purposes. Deep learning methods also showed utility in identifying a wide range of genetic mutations and standard pathology biomarkers from routine histology. This survey of 41 primary studies also encompasses key regions of AI applicability in histopathology in a multi-cancer review while marking prospects to introduce AI into the clinical setting with key examples including Swarm Learning and Data Fusion.
Collapse
Affiliation(s)
- Aaditya Tiwari
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Barts Cancer Institute, Cancer Research UK City of London Centre, Queen Mary University of London, London, UK
- Department of Oncology, Princess Alexandra Hospital NHS Trust, Harlow, UK
| | - Aruni Ghose
- Barts Cancer Institute, Cancer Research UK City of London Centre, Queen Mary University of London, London, UK.
- Department of Oncology, Princess Alexandra Hospital NHS Trust, Harlow, UK.
- Barts Cancer Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK.
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, UK.
- Digital Health Network, European Cancer Organisation, Brussels, Belgium.
- OncoFlowTM, London, UK.
- United Kingdom and Ireland Global Cancer Network, Manchester, UK.
- Oncology Council, Royal Society of Medicine, London, UK.
| | - Maryam Hasanova
- OncoFlowTM, London, UK
- Division of Biosciences, University College London, London, UK
| | - Sara Socorro Faria
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
| | - Srishti Mohapatra
- General Internal Medicine Doctorate Programme, University of Hertfordshire, Hatfield, UK
- The Misdiagnosis Association and Research Institute, California, USA
| | - Sola Adeleke
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
- Guy's Cancer Centre, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, UK
- Kent and Medway Medical School, University of Kent, Canterbury, UK
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, UK
- Faculty of Life Sciences and Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- AELIA Organization, 9Th Km Thessaloniki-Thermi, 57001, Thessaloniki, Greece
| |
Collapse
|
6
|
Boge F, Mosig A. Causality and scientific explanation of artificial intelligence systems in biomedicine. Pflugers Arch 2025; 477:543-554. [PMID: 39470762 PMCID: PMC11958387 DOI: 10.1007/s00424-024-03033-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024]
Abstract
With rapid advances of deep neural networks over the past decade, artificial intelligence (AI) systems are now commonplace in many applications in biomedicine. These systems often achieve high predictive accuracy in clinical studies, and increasingly in clinical practice. Yet, despite their commonly high predictive accuracy, the trustworthiness of AI systems needs to be questioned when it comes to decision-making that affects the well-being of patients or the fairness towards patients or other stakeholders affected by AI-based decisions. To address this, the field of explainable artificial intelligence, or XAI for short, has emerged, seeking to provide means by which AI-based decisions can be explained to experts, users, or other stakeholders. While it is commonly claimed that explanations of artificial intelligence (AI) establish the trustworthiness of AI-based decisions, it remains unclear what traits of explanations cause them to foster trustworthiness. Building on historical cases of scientific explanation in medicine, we here propagate our perspective that, in order to foster trustworthiness, explanations in biomedical AI should meet the criteria of being scientific explanations. To further undermine our approach, we discuss its relation to the concepts of causality and randomized intervention. In our perspective, we combine aspects from the three disciplines of biomedicine, machine learning, and philosophy. From this interdisciplinary angle, we shed light on how the explanation and trustworthiness of artificial intelligence relate to the concepts of causality and robustness. To connect our perspective with AI research practice, we review recent cases of AI-based studies in pathology and, finally, provide guidelines on how to connect AI in biomedicine with scientific explanation.
Collapse
Affiliation(s)
- Florian Boge
- Institute for Philosophy and Political Science, Technical University Dortmund, Emil-Figge-Str. 50, 44227, Dortmund, Germany
| | - Axel Mosig
- Bioinformatics Group, Department for Biology and Biotechnology, Ruhr-University Bochum (RUB), Gesundheitscampus 4, 44801, Bochum, NRW, Germany.
- Center for Protein Diagnostics, Ruhr University Bochum, Gesundheitscampus 4, 44801, Bochum, Germany.
| |
Collapse
|
7
|
Hölscher DL, Bülow RD. Decoding pathology: the role of computational pathology in research and diagnostics. Pflugers Arch 2025; 477:555-570. [PMID: 39095655 PMCID: PMC11958429 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
|
8
|
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
|
9
|
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
|
10
|
Huang Q, Tang X, Gan C, Deng Q, Zhi S, Huang Q, Zheng X, Li X, Pan Z, Huang M. EFHD1 Activates SIK3 to Limit Colorectal Cancer Initiation and Progression via the Hippo Pathway. J Cancer 2025; 16:1348-1362. [PMID: 39895792 PMCID: PMC11786025 DOI: 10.7150/jca.103229] [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: 09/04/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers, with high rates of metastasis and lethality. EF-hand domain-containing protein D1 (EFHD1) and salt-inducible kinase 3 (SIK3) have been studied in several cancer types. Aberrant expression of EFHD1 and SIK3 has been observed in CRC, but little research has addressed their regulatory abilities and signaling pathways. In this study, we aimed to explore the efficacy of EFHD1 in inhibiting CRC proliferation and metastasis and to elucidate the underlying mechanisms involved in the upregulation of SIK3 expression. Cell viability, colony formation, wound healing, Transwell assay, orthotopic xenograft, and pulmonary metastasis mouse models were used to detect the antiproliferative and anti-metastatic effects of EFHD1 against CRC in vitro and in vivo. The Gene Expression Profiling Interactive Analysis (GEPIA) database was used to determine EFHD1 and SIK3 expression in CRC. The regulatory roles of EFHD1 and SIK3 in mediating anti-metastatic effects in CRC were measured using western blotting, immunohistochemical, and immunofluorescence analyses. The results showed that EFHD1 expression was significantly repressed in the clinical CRC samples. EFHD1 markedly suppressed cell proliferation, migration, and invasion in vitro and inhibited tumor growth and metastasis in vivo. Analysis of the GEPIA database revealed that EFHD1 expression positively correlated with SIK3 expression. SIK3 overexpression inhibited the migration of CRC cells, and SIK3 knockdown partially eliminated the inhibitory effects of EFHD1 on CRC metastasis. EFHD1 exerted anti-metastatic effects against CRC via upregulating SIK3 and inhibiting epithelial-mesenchymal transition (EMT) processing through modulating the Hippo signaling pathway. Collectively, these findings identify EFHD1 as a potent SIK3 agonist and highlight the EFHD1-SIK3 axis as a key modulator of the Hippo signaling pathway in CRC. EFHD1 serves as a novel regulator and is worthy of further development as a novel therapeutic target in CRC.
Collapse
Affiliation(s)
- Qionghui Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Xiaoyan Tang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Caiyan Gan
- Institute of basic medical sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Qiaoting Deng
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Shaobin Zhi
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Qingyan Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Xiaoqi Zheng
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Xueqiong Li
- Medical College of Jiaying University, Meizhou, China
| | - Zengfeng Pan
- Institute of basic medical sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Mingfeng Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| |
Collapse
|
11
|
Zheng Y, Wu K, Li J, Tang K, Shi J, Wu H, Jiang Z, Wang W. Partial-Label Contrastive Representation Learning for Fine-Grained Biomarkers Prediction From Histopathology Whole Slide Images. IEEE J Biomed Health Inform 2025; 29:396-408. [PMID: 39012745 DOI: 10.1109/jbhi.2024.3429188] [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] [Indexed: 07/18/2024]
Abstract
In the domain of histopathology analysis, existing representation learning methods for biomarkers prediction from whole slide images (WSIs) face challenges due to the complexity of tissue subtypes and label noise problems. This paper proposed a novel partial-label contrastive representation learning approach to enhance the discrimination of histopathology image representations for fine-grained biomarkers prediction. We designed a partial-label contrastive clustering (PLCC) module for partial-label disambiguation and a dynamic clustering algorithm to sample the most representative features of each category to the clustering queue during the contrastive learning process. We conducted comprehensive experiments on three gene mutation prediction datasets, including USTC-EGFR, BRCA-HER2, and TCGA-EGFR. The results show that our method outperforms 9 existing methods in terms of Accuracy, AUC, and F1 Score. Specifically, our method achieved an AUC of 0.950 in EGFR mutation subtyping of TCGA-EGFR and an AUC of 0.853 in HER2 0/1+/2+/3+ grading of BRCA-HER2, which demonstrates its superiority in fine-grained biomarkers prediction from histopathology whole slide images.
Collapse
|
12
|
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
|
13
|
Murchan P, Ó Broin P, Baird AM, Sheils O, P Finn S. Deep feature batch correction using ComBat for machine learning applications in computational pathology. J Pathol Inform 2024; 15:100396. [PMID: 39398947 PMCID: PMC11470259 DOI: 10.1016/j.jpi.2024.100396] [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: 07/08/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
Background Developing artificial intelligence (AI) models for digital pathology requires large datasets from multiple sources. However, without careful implementation, AI models risk learning confounding site-specific features in datasets instead of clinically relevant information, leading to overestimated performance, poor generalizability to real-world data, and potential misdiagnosis. Methods Whole-slide images (WSIs) from The Cancer Genome Atlas (TCGA) colon (COAD), and stomach adenocarcinoma datasets were selected for inclusion in this study. Patch embeddings were obtained using three feature extraction models, followed by ComBat harmonization. Attention-based multiple instance learning models were trained to predict tissue-source site (TSS), as well as clinical and genetic attributes, using raw, Macenko normalized, and Combat-harmonized patch embeddings. Results TSS prediction achieved high accuracy (AUROC > 0.95) with all three feature extraction models. ComBat harmonization significantly reduced the AUROC for TSS prediction, with mean AUROCs dropping to approximately 0.5 for most models, indicating successful mitigation of batch effects (e.g., CCL-ResNet50 in TCGA-COAD: Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, p < 0.001). Clinical attributes associated with TSS, such as race and treatment response, showed decreased predictability post-harmonization. Notably, the prediction of genetic features like MSI status remained robust after harmonization (e.g., MSI in TCGA-COAD: Pre-ComBat AUROC = 0.667, Post-ComBat AUROC = 0.669, p=0.952), indicating the preservation of true histological signals. Conclusion ComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates. This approach is promising for the integration of large-scale digital pathology datasets.
Collapse
Affiliation(s)
- Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
| | - Pilib Ó Broin
- The SFI Centre for Research Training in Genomics Data Science, Dublin, Ireland
- School of Mathematical & Statistical Sciences, University of Galway, Galway H91 TK33, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D02 A440, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin D08 W9RT, Ireland
- Department of Histopathology, St. James's Hospital, James's Street, Dublin D08 X4RX, Ireland
| |
Collapse
|
14
|
Xiaojian Y, Zhanbo Q, Jian C, Zefeng W, Jian L, Jin L, Yuefen P, Shuwen H. Deep learning application in prediction of cancer molecular alterations based on pathological images: a bibliographic analysis via CiteSpace. J Cancer Res Clin Oncol 2024; 150:467. [PMID: 39422817 PMCID: PMC11489169 DOI: 10.1007/s00432-024-05992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The advancements in artificial intelligence (AI) technology for image recognition were propelling molecular pathology research into a new era. OBJECTIVE To summarize the hot spots and research trends in the field of molecular pathology image recognition. METHODS Relevant articles from January 1st, 2010, to August 25th, 2023, were retrieved from the Web of Science Core Collection. Subsequently, CiteSpace was employed for bibliometric and visual analysis, generating diverse network diagrams illustrating keywords, highly cited references, hot topics, and research trends. RESULTS A total of 110 relevant articles were extracted from a pool of 10,205 articles. The overall publication count exhibited a rising trend each year. The leading contributors in terms of institutions, countries, and authors were Maastricht University (11 articles), the United States (38 articles), and Kather Jacob Nicholas (9 articles), respectively. Half of the top ten research institutions, based on publication volume, were affiliated with Germany. The most frequently cited article was authored by Nicolas Coudray et al. accumulating 703 citations. The keyword "Deep learning" had the highest frequency in 2019. Notably, the highlighted keywords from 2022 to 2023 included "microsatellite instability", and there were 21 articles focusing on utilizing algorithms to recognize microsatellite instability (MSI) in colorectal cancer (CRC) pathological images. CONCLUSION The use of DL is expected to provide a new strategy to effectively solve the current problem of time-consuming and expensive molecular pathology detection. Therefore, further research is needed to address issues, such as data quality and standardization, model interpretability, and resource and infrastructure requirements.
Collapse
Affiliation(s)
- Yu Xiaojian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Wang Zefeng
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jin
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Pan Yuefen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
15
|
Hyeon D, Kim Y, Hwang Y, Bae JM, Kang GH, Kim K. Deep learning-based histological predictions of chromosomal instability in colorectal cancer. Am J Cancer Res 2024; 14:4495-4505. [PMID: 39417190 PMCID: PMC11477831 DOI: 10.62347/jynd6488] [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: 06/25/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Colorectal cancer (CRC) is a lethal malignancy and a leading cause of cancer-related mortality worldwide. Chromosomal instability (CIN) is a key driver of genomic instability in CRC and is characterized by aneuploidy and somatic copy-number alterations. This study aimed to predict CIN in CRC using histological data from whole slide images (WSIs). CRC samples from TCGA were analyzed, with tumor regions segmented into tiles and nuclei for feature extraction using convolutional neural network (CNN) and morphologic analysis. Binary classification models were developed to distinguish high and low aneuploidy scores (AS) based on slide-level features. The analysis included 313 patients with 315 WSIs, resulting in over 350,000 tumor tiles and nearly 2.7 million tumor cell nuclei. The ResNet18-SSL model, pre-trained on histopathological images, demonstrated superior accuracy in tile-based AS prediction, while DenseNet121 excelled in nucleus-based prediction. Combining CNN-based and morphological features enhanced the classification accuracy of nucleus-based predictions. Additionally, significant correlations were observed between morphological features and copy-number signatures. Unsupervised clustering of nuclear features revealed that distinct groups are significantly correlated with CIN and TP53 mutations. This study underscores the potential of histological features from WSIs to predict CIN in CRC samples. Nuclear feature analysis, combined with deep-learning techniques, offers a robust method for CIN prediction, highlighting the importance of further research into the relationships between histological and molecular phenotypes.
Collapse
Affiliation(s)
- Dongwoo Hyeon
- Institute of Biomedical Research, Seoul National University HospitalSeoul, South Korea
| | - Younghoon Kim
- Department of Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of KoreaSeoul, South Korea
| | - Yaeeun Hwang
- Department of Veterinary Medicine, Seoul National UniversitySeoul, South Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University HospitalSeoul, South Korea
| | - Gyeong Hoon Kang
- Department of Pathology, College of Medicine, Seoul National UniversitySeoul, South Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University HospitalSeoul, South Korea
- Department of Medicine, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
16
|
Kumar A, Saha L. Colorectal cancer cell dormancy: An insight into pathways. World J Gastroenterol 2024; 30:3810-3817. [PMID: 39351431 PMCID: PMC11438629 DOI: 10.3748/wjg.v30.i33.3810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/02/2024] Open
Abstract
Cancer cell dormancy (CCD) in colorectal cancer (CRC) poses a significant challenge to effective treatment. In CRC, CCD contributes to tumour recurrence, drug resistance, and amplifying the disease's burden. The molecular mechanisms governing CCD and strategies for eliminating dormant cancer cells remain largely unexplored. Therefore, understanding the molecular mechanisms governing dormancy is crucial for improving patient outcomes and developing targeted therapies. This editorial highlights the complex interplay of signalling pathways and factors involved in colorectal CCD, emphasizing the roles of Hippo/YAP, pluripotent transcription factors such as NANOG, HIF-1α signalling, and Notch signalling pathways. Additionally, ERK/p38α/β/MAPK pathways, AKT signalling pathway, and Extracellular Matrix Metalloproteinase Inducer, along with some potential less explored pathways such as STAT/p53 switch and canonical and non-canonical Wnt and SMAD signalling, are also involved in promoting colorectal CCD. Highlighting their clinical significance, these findings may offer the potential for identifying key dormancy regulator pathways, improving treatment strategies, surmounting drug resistance, and advancing personalized medicine approaches. Moreover, insights into dormancy mechanisms could lead to the development of predictive biomarkers for identifying patients at risk of recurrence and the tailoring of targeted therapies based on individual dormancy profiles. It is essential to conduct further research into these pathways and their modulation to fully comprehend CRC dormancy mechanisms and enhance patient outcomes.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Lekha Saha
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
17
|
Carrillo-Perez F, Cramer EM, Pizurica M, Andor N, Gevaert O. Towards Digital Quantification of Ploidy from Pan-Cancer Digital Pathology Slides using Deep Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608555. [PMID: 39229200 PMCID: PMC11370345 DOI: 10.1101/2024.08.19.608555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Abnormal DNA ploidy, found in numerous cancers, is increasingly being recognized as a contributor in driving chromosomal instability, genome evolution, and the heterogeneity that fuels cancer cell progression. Furthermore, it has been linked with poor prognosis of cancer patients. While next-generation sequencing can be used to approximate tumor ploidy, it has a high error rate for near-euploid states, a high cost and is time consuming, motivating alternative rapid quantification methods. We introduce PloiViT, a transformer-based model for tumor ploidy quantification that outperforms traditional machine learning models, enabling rapid and cost-effective quantification directly from pathology slides. We trained PloiViT on a dataset of fifteen cancer types from The Cancer Genome Atlas and validated its performance in multiple independent cohorts. Additionally, we explored the impact of self-supervised feature extraction on performance. PloiViT, using self-supervised features, achieved the lowest prediction error in multiple independent cohorts, exhibiting better generalization capabilities. Our findings demonstrate that PloiViT predicts higher ploidy values in aggressive cancer groups and patients with specific mutations, validating PloiViT potential as complementary for ploidy assessment to next-generation sequencing data. To further promote its use, we release our models as a user-friendly inference application and a Python package for easy adoption and use.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, 94304, CA, USA
| | - Eric M. Cramer
- Department of Biomedical Engineering, Oregon Health & Science University (OHSU), Portland, 97239, OR, USA
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, 94304, CA, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Ghent, 9052, Ghent, Belgium
| | - Noemi Andor
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, 33612, FL, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, 94304, CA, USA
- Department of Biomedical Data Science (DBDS), Stanford University, Palo Alto, 94305, CA, USA
| |
Collapse
|
18
|
Faa G, Coghe F, Pretta A, Castagnola M, Van Eyken P, Saba L, Scartozzi M, Fraschini M. Artificial Intelligence Models for the Detection of Microsatellite Instability from Whole-Slide Imaging of Colorectal Cancer. Diagnostics (Basel) 2024; 14:1605. [PMID: 39125481 PMCID: PMC11311951 DOI: 10.3390/diagnostics14151605] [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: 07/08/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
With the advent of whole-slide imaging (WSI), a technology that can digitally scan whole slides in high resolution, pathology is undergoing a digital revolution. Detecting microsatellite instability (MSI) in colorectal cancer is crucial for proper treatment, as it identifies patients responsible for immunotherapy. Even though universal testing for MSI is recommended, particularly in patients affected by colorectal cancer (CRC), many patients remain untested, and they reside mainly in low-income countries. A critical need exists for accessible, low-cost tools to perform MSI pre-screening. Here, the potential predictive role of the most relevant artificial intelligence-driven models in predicting microsatellite instability directly from histology alone is discussed, focusing on CRC. The role of deep learning (DL) models in identifying the MSI status is here analyzed in the most relevant studies reporting the development of algorithms trained to this end. The most important performance and the most relevant deficiencies are discussed for every AI method. The models proposed for algorithm sharing among multiple research and clinical centers, including federal learning (FL) and swarm learning (SL), are reported. According to all the studies reported here, AI models are valuable tools for predicting MSI status on WSI alone in CRC. The use of digitized H&E-stained sections and a trained algorithm allow the extraction of relevant molecular information, such as MSI status, in a short time and at a low cost. The possible advantages related to introducing DL methods in routine surgical pathology are underlined here, and the acceleration of the digital transformation of pathology departments and services is recommended.
Collapse
Affiliation(s)
- Gavino Faa
- Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, 09123 Cagliari, Italy;
| | - Ferdinando Coghe
- UOC Laboratorio Analisi, AOU of Cagliari, 09123 Cagliari, Italy;
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (A.P.); (M.S.)
| | - Massimo Castagnola
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Peter Van Eyken
- Division of Pathology, Genk Regional Hospital, 3600 Genk, Belgium;
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, University of Cagliari, 40138 Cagliari, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (A.P.); (M.S.)
| | - Matteo Fraschini
- Dipartimento di Ingegneria Elettrica ed Elettronica, University of Cagliari, 09123 Cagliari, Italy
| |
Collapse
|
19
|
Maddox A, Fowler R, Solomon E, Rao A. Undergraduate Education in Computational Pathology Through Global Health Inspired Projects. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039320 DOI: 10.1109/embc53108.2024.10782173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Recent technological advancements are revolutionizing the field of pathology as the practice adopts digital workflows and computational tools to augment the analysis of tissue and enhance its role in patient care. These advancements are poised to make a particularly significant impact in low-income and middle-income countries, which are disproportionately affected by worsening pathology service shortages and rising cancer rates. Courses targeted towards undergraduate students interested in this emergent field of computational pathology (CPATH) are required to prepare the next generation of innovators and leaders in this space. However, such training courses have mostly been lacking and their design presents with several challenges. CPATH exists at the intersection of multiple complex specialties and so, following a traditional bottom-up curriculum, courses are often limited to the graduate level. In addition, standard didactic courses struggle to keep with the rapid pace of advancements driving the field, build the essential multidisciplinary teamwork skills, train the technical skillsets required for success, or emphasize innovation. Structured experiential learning (EL) has a long track record of success in addressing these issues and presents as a natural modality for early CPATH education. We have designed and piloted a project based EL course targeted towards undergraduates to address these limitations in CPATH education. At the core of the course experience, students work together to conceptualize, design, and implement innovative solutions to leverage CPATH towards addressing global health inequity. Here we present our design of the course, review insights from our first two years of piloting this course and share plans for course improvement drawn from these insights.Clinical relevance- CPATH is making a significant impact on the practice of pathology and is poised to play a major role in addressing global health inequities. This course is designed to prepare undergraduate and graduate students to innovate in this rapidly growing and developing field as members of multidisciplinary teams through structured project based EL. While open to students at the undergraduate and master's level from all backgrounds, it is directed at undergraduate biomedical engineering, computer science, and pre medicine students who are interested in future careers in this field.
Collapse
|
20
|
Reitsam NG, Enke JS, Vu Trung K, Märkl B, Kather JN. Artificial Intelligence in Colorectal Cancer: From Patient Screening over Tailoring Treatment Decisions to Identification of Novel Biomarkers. Digestion 2024; 105:331-344. [PMID: 38865982 PMCID: PMC11457979 DOI: 10.1159/000539678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Artificial intelligence (AI) is increasingly entering and transforming not only medical research but also clinical practice. In the last 10 years, new AI methods have enabled computers to perform visual tasks, reaching high performance and thereby potentially supporting and even outperforming human experts. This is in particular relevant for colorectal cancer (CRC), which is the 3rd most common cancer type in general, as along the CRC patient journey many complex visual tasks need to be performed: from endoscopy over imaging to histopathology; the screening, diagnosis, and treatment of CRC involve visual image analysis tasks. SUMMARY In all these clinical areas, AI models have shown promising results by supporting physicians, improving accuracy, and providing new biological insights and biomarkers. By predicting prognostic and predictive biomarkers from routine images/slides, AI models could lead to an improved patient stratification for precision oncology approaches in the near future. Moreover, it is conceivable that AI models, in particular together with innovative techniques such as single-cell or spatial profiling, could help identify novel clinically as well as biologically meaningful biomarkers that could pave the way to new therapeutic approaches. KEY MESSAGES Here, we give a comprehensive overview of AI in colorectal cancer, describing and discussing these developments as well as the next steps which need to be taken to incorporate AI methods more broadly into the clinical care of CRC.
Collapse
Affiliation(s)
- Nic Gabriel Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany,
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany,
| | - Johanna Sophie Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Kien Vu Trung
- Division of Gastroenterology, Medical Department II, University of Leipzig Medical Center, Leipzig, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
21
|
Niehues JM, Müller-Franzes G, Schirris Y, Wagner SJ, Jendrusch M, Kloor M, Pearson AT, Muti HS, Hewitt KJ, Veldhuizen GP, Zigutyte L, Truhn D, Kather JN. Using histopathology latent diffusion models as privacy-preserving dataset augmenters improves downstream classification performance. Comput Biol Med 2024; 175:108410. [PMID: 38678938 DOI: 10.1016/j.compbiomed.2024.108410] [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: 10/15/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
Latent diffusion models (LDMs) have emerged as a state-of-the-art image generation method, outperforming previous Generative Adversarial Networks (GANs) in terms of training stability and image quality. In computational pathology, generative models are valuable for data sharing and data augmentation. However, the impact of LDM-generated images on histopathology tasks compared to traditional GANs has not been systematically studied. We trained three LDMs and a styleGAN2 model on histology tiles from nine colorectal cancer (CRC) tissue classes. The LDMs include 1) a fine-tuned version of stable diffusion v1.4, 2) a Kullback-Leibler (KL)-autoencoder (KLF8-DM), and 3) a vector quantized (VQ)-autoencoder deploying LDM (VQF8-DM). We assessed image quality through expert ratings, dimensional reduction methods, distribution similarity measures, and their impact on training a multiclass tissue classifier. Additionally, we investigated image memorization in the KLF8-DM and styleGAN2 models. All models provided a high image quality, with the KLF8-DM achieving the best Frechet Inception Distance (FID) and expert rating scores for complex tissue classes. For simpler classes, the VQF8-DM and styleGAN2 models performed better. Image memorization was negligible for both styleGAN2 and KLF8-DM models. Classifiers trained on a mix of KLF8-DM generated and real images achieved a 4% improvement in overall classification accuracy, highlighting the usefulness of these images for dataset augmentation. Our systematic study of generative methods showed that KLF8-DM produces the highest quality images with negligible image memorization. The higher classifier performance in the generatively augmented dataset suggests that this augmentation technique can be employed to enhance histopathology classifiers for various tasks.
Collapse
Affiliation(s)
- Jan M Niehues
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Gustav Müller-Franzes
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Yoni Schirris
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Netherlands Cancer Institute, 1066 CX, Amsterdam, the Netherlands; University of Amsterdam, 1012 WP, Amsterdam, the Netherlands
| | - Sophia Janine Wagner
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany; School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Michael Jendrusch
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Kloor
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Hannah Sophie Muti
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Katherine J Hewitt
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Gregory P Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Laura Zigutyte
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
22
|
Perez-Lopez R, Ghaffari Laleh N, Mahmood F, Kather JN. A guide to artificial intelligence for cancer researchers. Nat Rev Cancer 2024; 24:427-441. [PMID: 38755439 DOI: 10.1038/s41568-024-00694-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Artificial intelligence (AI) has been commoditized. It has evolved from a specialty resource to a readily accessible tool for cancer researchers. AI-based tools can boost research productivity in daily workflows, but can also extract hidden information from existing data, thereby enabling new scientific discoveries. Building a basic literacy in these tools is useful for every cancer researcher. Researchers with a traditional biological science focus can use AI-based tools through off-the-shelf software, whereas those who are more computationally inclined can develop their own AI-based software pipelines. In this article, we provide a practical guide for non-computational cancer researchers to understand how AI-based tools can benefit them. We convey general principles of AI for applications in image analysis, natural language processing and drug discovery. In addition, we give examples of how non-computational researchers can get started on the journey to productively use AI in their own work.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Narmin Ghaffari Laleh
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
23
|
Xiao T, Kong S, Zhang Z, Hua D, Liu F. A review of big data technology and its application in cancer care. Comput Biol Med 2024; 176:108577. [PMID: 38739981 DOI: 10.1016/j.compbiomed.2024.108577] [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: 11/19/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
The development of modern medical devices and information technology has led to a rapid growth in the amount of data available for health protection information, with the concept of medical big data emerging globally, along with significant advances in cancer care relying on data-driven approaches. However, outstanding issues such as fragmented data governance, low-quality data specification, and data lock-in still make sharing challenging. Big data technology provides solutions for managing massive heterogeneous data while combining artificial intelligence (AI) techniques such as machine learning (ML) and deep learning (DL) to better mine the intrinsic connections between data. This paper surveys and organizes recent articles on big data technology and its applications in cancer, dividing them into three different types to outline their primary content and summarize their critical role in assisting cancer care. It then examines the latest research directions in big data technology in cancer and evaluates the current state of development of each type of application. Finally, current challenges and opportunities are discussed, and recommendations are made for the further integration of big data technology into the medical industry in the future.
Collapse
Affiliation(s)
- Tianyun Xiao
- Hebei Key Laboratory of Data Science and Application, North China University of Science and Technology, Tangshan, Hebei, 063210, China; The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, Hebei, 063210, China; College of Science, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Shanshan Kong
- College of Science, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| | - Zichen Zhang
- Hebei Key Laboratory of Data Science and Application, North China University of Science and Technology, Tangshan, Hebei, 063210, China; The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, Hebei, 063210, China; College of Science, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Dianbo Hua
- Beijing Sitairui Cancer Data Analysis Joint Laboratory, Beijing, 101149, China
| | - Fengchun Liu
- Hebei Key Laboratory of Data Science and Application, North China University of Science and Technology, Tangshan, Hebei, 063210, China; The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, Hebei, 063210, China; College of Science, North China University of Science and Technology, Tangshan, Hebei, 063210, China; Hebei Engineering Research Center for the Intelligentization of Iron Ore Optimization and Ironmaking Raw Materials Preparation Processes, North China University of Science and Technology, Tangshan, Hebei, China; Tangshan Intelligent Industry and Image Processing Technology Innovation Center, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
24
|
Williams DKA, Graifman G, Hussain N, Amiel M, Tran P, Reddy A, Haider A, Kavitesh BK, Li A, Alishahian L, Perera N, Efros C, Babu M, Tharakan M, Etienne M, Babu BA. Digital pathology, deep learning, and cancer: a narrative review. Transl Cancer Res 2024; 13:2544-2560. [PMID: 38881914 PMCID: PMC11170525 DOI: 10.21037/tcr-23-964] [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: 06/05/2023] [Accepted: 03/24/2024] [Indexed: 06/18/2024]
Abstract
Background and Objective Cancer is a leading cause of morbidity and mortality worldwide. The emergence of digital pathology and deep learning technologies signifies a transformative era in healthcare. These technologies can enhance cancer detection, streamline operations, and bolster patient care. A substantial gap exists between the development phase of deep learning models in controlled laboratory environments and their translations into clinical practice. This narrative review evaluates the current landscape of deep learning and digital pathology, analyzing the factors influencing model development and implementation into clinical practice. Methods We searched multiple databases, including Web of Science, Arxiv, MedRxiv, BioRxiv, Embase, PubMed, DBLP, Google Scholar, IEEE Xplore, Semantic Scholar, and Cochrane, targeting articles on whole slide imaging and deep learning published from 2014 and 2023. Out of 776 articles identified based on inclusion criteria, we selected 36 papers for the analysis. Key Content and Findings Most articles in this review focus on the in-laboratory phase of deep learning model development, a critical stage in the deep learning lifecycle. Challenges arise during model development and their integration into clinical practice. Notably, lab performance metrics may not always match real-world clinical outcomes. As technology advances and regulations evolve, we expect more clinical trials to bridge this performance gap and validate deep learning models' effectiveness in clinical care. High clinical accuracy is vital for informed decision-making throughout a patient's cancer care. Conclusions Deep learning technology can enhance cancer detection, clinical workflows, and patient care. Challenges may arise during model development. The deep learning lifecycle involves data preprocessing, model development, and clinical implementation. Achieving health equity requires including diverse patient groups and eliminating bias during implementation. While model development is integral, most articles focus on the pre-deployment phase. Future longitudinal studies are crucial for validating models in real-world settings post-deployment. A collaborative approach among computational pathologists, technologists, industry, and healthcare providers is essential for driving adoption in clinical settings.
Collapse
Affiliation(s)
| | | | - Nowair Hussain
- Department of Internal Medicine, Overlook Medical Center, Summit, NJ, USA
| | | | | | - Arjun Reddy
- Applied Mathematics & Statistics Stony Brook University, Stony Brook, NY, USA
| | - Ali Haider
- Department of Artificial Intelligence, Yeshiva University, New York, NY, USA
| | - Bali Kumar Kavitesh
- Centre for Frontier AI Research (CFAR), Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Austin Li
- New York Medical College, Valhalla, NY, USA
| | | | | | | | - Myoungmee Babu
- Artificial Intelligence and Mathematics, New York City Department of Education, New York, NY, USA
| | | | - Mill Etienne
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Benson A Babu
- New York Medical College, Valhalla, NY, USA
- Department of Hospital Medicine, Wyckoff, Medical Center, New York, NY, USA
| |
Collapse
|
25
|
Unger M, Kather JN. Deep learning in cancer genomics and histopathology. Genome Med 2024; 16:44. [PMID: 38539231 PMCID: PMC10976780 DOI: 10.1186/s13073-024-01315-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2024] [Indexed: 07/08/2024] Open
Abstract
Histopathology and genomic profiling are cornerstones of precision oncology and are routinely obtained for patients with cancer. Traditionally, histopathology slides are manually reviewed by highly trained pathologists. Genomic data, on the other hand, is evaluated by engineered computational pipelines. In both applications, the advent of modern artificial intelligence methods, specifically machine learning (ML) and deep learning (DL), have opened up a fundamentally new way of extracting actionable insights from raw data, which could augment and potentially replace some aspects of traditional evaluation workflows. In this review, we summarize current and emerging applications of DL in histopathology and genomics, including basic diagnostic as well as advanced prognostic tasks. Based on a growing body of evidence, we suggest that DL could be the groundwork for a new kind of workflow in oncology and cancer research. However, we also point out that DL models can have biases and other flaws that users in healthcare and research need to know about, and we propose ways to address them.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, 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
|
26
|
Chen RJ, Ding T, Lu MY, Williamson DFK, Jaume G, Song AH, Chen B, Zhang A, Shao D, Shaban M, Williams M, Oldenburg L, Weishaupt LL, Wang JJ, Vaidya A, Le LP, Gerber G, Sahai S, Williams W, Mahmood F. Towards a general-purpose foundation model for computational pathology. Nat Med 2024; 30:850-862. [PMID: 38504018 PMCID: PMC11403354 DOI: 10.1038/s41591-024-02857-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 159.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024]
Abstract
Quantitative evaluation of tissue images is crucial for computational pathology (CPath) tasks, requiring the objective characterization of histopathological entities from whole-slide images (WSIs). The high resolution of WSIs and the variability of morphological features present significant challenges, complicating the large-scale annotation of data for high-performance applications. To address this challenge, current efforts have proposed the use of pretrained image encoders through transfer learning from natural image datasets or self-supervised learning on publicly available histopathology datasets, but have not been extensively developed and evaluated across diverse tissue types at scale. We introduce UNI, a general-purpose self-supervised model for pathology, pretrained using more than 100 million images from over 100,000 diagnostic H&E-stained WSIs (>77 TB of data) across 20 major tissue types. The model was evaluated on 34 representative CPath tasks of varying diagnostic difficulty. In addition to outperforming previous state-of-the-art models, we demonstrate new modeling capabilities in CPath such as resolution-agnostic tissue classification, slide classification using few-shot class prototypes, and disease subtyping generalization in classifying up to 108 cancer types in the OncoTree classification system. UNI advances unsupervised representation learning at scale in CPath in terms of both pretraining data and downstream evaluation, enabling data-efficient artificial intelligence models that can generalize and transfer to a wide range of diagnostically challenging tasks and clinical workflows in anatomic pathology.
Collapse
Affiliation(s)
- Richard J Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Tong Ding
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Ming Y Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Guillaume Jaume
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew H Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bowen Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Zhang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Daniel Shao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Muhammad Shaban
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mane Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Lukas Oldenburg
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luca L Weishaupt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Judy J Wang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anurag Vaidya
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Georg Gerber
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sharifa Sahai
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Systems Biology, Harvard University, Cambridge, MA, USA
| | - Walt Williams
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
27
|
Murchan P, Baird AM, Ó Broin P, Sheils O, Finn SP. Surrogate Biomarker Prediction from Whole-Slide Images for Evaluating Overall Survival in Lung Adenocarcinoma. Diagnostics (Basel) 2024; 14:462. [PMID: 38472935 DOI: 10.3390/diagnostics14050462] [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: 01/23/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Recent advances in computational pathology have shown potential in predicting biomarkers from haematoxylin and eosin (H&E) whole-slide images (WSI). However, predicting the outcome directly from WSIs remains a substantial challenge. In this study, we aimed to investigate how gene expression, predicted from WSIs, could be used to evaluate overall survival (OS) in patients with lung adenocarcinoma (LUAD). METHODS Differentially expressed genes (DEGs) were identified from The Cancer Genome Atlas (TCGA)-LUAD cohort. Cox regression analysis was performed on DEGs to identify the gene prognostics of OS. Attention-based multiple instance learning (AMIL) models were trained to predict the expression of identified prognostic genes from WSIs using the TCGA-LUAD dataset. Models were externally validated in the Clinical Proteomic Tumour Analysis Consortium (CPTAC)-LUAD dataset. The prognostic value of predicted gene expression values was then compared to the true gene expression measurements. RESULTS The expression of 239 prognostic genes could be predicted in TCGA-LUAD with cross-validated Pearson's R > 0.4. Predicted gene expression demonstrated prognostic performance, attaining a cross-validated concordance index of up to 0.615 in TCGA-LUAD through Cox regression. In total, 36 genes had predicted expression in the external validation cohort that was prognostic of OS. CONCLUSIONS Gene expression predicted from WSIs is an effective method of evaluating OS in patients with LUAD. These results may open up new avenues of cost- and time-efficient prognosis assessment in LUAD treatment.
Collapse
Affiliation(s)
- Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- The SFI Centre for Research Training in Genomics Data Science, University of Galway, H91 CF50 Galway, Ireland
- Trinity St. James's Cancer Institute (TSJCI), St. James's Hospital, D08 RX0X Dublin, Ireland
| | - Anne-Marie Baird
- Trinity St. James's Cancer Institute (TSJCI), St. James's Hospital, D08 RX0X Dublin, Ireland
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D02 A440 Dublin, Ireland
| | - Pilib Ó Broin
- School of Mathematical & Statistical Sciences, University of Galway, H91 TK33 Galway, Ireland
| | - Orla Sheils
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Trinity St. James's Cancer Institute (TSJCI), St. James's Hospital, D08 RX0X Dublin, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Trinity St. James's Cancer Institute (TSJCI), St. James's Hospital, D08 RX0X Dublin, Ireland
- Department of Histopathology, St. James's Hospital, James's Street, D08 X4RX Dublin, Ireland
| |
Collapse
|
28
|
El Nahhas OSM, Loeffler CML, Carrero ZI, van Treeck M, Kolbinger FR, Hewitt KJ, Muti HS, Graziani M, Zeng Q, Calderaro J, Ortiz-Brüchle N, Yuan T, Hoffmeister M, Brenner H, Brobeil A, Reis-Filho JS, Kather JN. Regression-based Deep-Learning predicts molecular biomarkers from pathology slides. Nat Commun 2024; 15:1253. [PMID: 38341402 PMCID: PMC10858881 DOI: 10.1038/s41467-024-45589-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Deep Learning (DL) can predict biomarkers from cancer histopathology. Several clinically approved applications use this technology. Most approaches, however, predict categorical labels, whereas biomarkers are often continuous measurements. We hypothesize that regression-based DL outperforms classification-based DL. Therefore, we develop and evaluate a self-supervised attention-based weakly supervised regression method that predicts continuous biomarkers directly from 11,671 images of patients across nine cancer types. We test our method for multiple clinically and biologically relevant biomarkers: homologous recombination deficiency score, a clinically used pan-cancer biomarker, as well as markers of key biological processes in the tumor microenvironment. Using regression significantly enhances the accuracy of biomarker prediction, while also improving the predictions' correspondence to regions of known clinical relevance over classification. In a large cohort of colorectal cancer patients, regression-based prediction scores provide a higher prognostic value than classification-based scores. Our open-source regression approach offers a promising alternative for continuous biomarker analysis in computational pathology.
Collapse
Grants
- P30 CA008748 NCI NIH HHS
- JNK is supported by the German Federal Ministry of Health (DEEP LIVER, ZMVI1-2520DAT111) and the Max-Eder-Programme of the German Cancer Aid (grant #70113864), the German Federal Ministry of Education and Research (PEARL, 01KD2104C; CAMINO, 01EO2101; SWAG, 01KD2215A; TRANSFORM LIVER, 031L0312A), the German Academic Exchange Service (SECAI, 57616814), the German Federal Joint Committee (Transplant.KI, 01VSF21048) the European Union (ODELIA, 101057091; GENIAL, 101096312) and the National Institute for Health and Care Research (NIHR, NIHR213331) Leeds Biomedical Research Centre. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health and Social Care.
Collapse
Affiliation(s)
- Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Fiona R Kolbinger
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Katherine J Hewitt
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Hannah S Muti
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Mara Graziani
- University of Applied Sciences of Western Switzerland (HES-SO Valais), Rue du Technopole 3, 3960, Sierre, Valais, Switzerland
| | - Qinghe Zeng
- Centre d'Histologie, d'Imagerie et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Julien Calderaro
- Assistance Publique-Hôpitaux de Paris, Département de Pathologie, CHU Henri Mondor, F-94000, Créteil, France
| | - Nadina Ortiz-Brüchle
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Cologne, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
- Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
29
|
Calderaro J, Ghaffari Laleh N, Zeng Q, Maille P, Favre L, Pujals A, Klein C, Bazille C, Heij LR, Uguen A, Luedde T, Di Tommaso L, Beaufrère A, Chatain A, Gastineau D, Nguyen CT, Nguyen-Canh H, Thi KN, Gnemmi V, Graham RP, Charlotte F, Wendum D, Vij M, Allende DS, Aucejo F, Diaz A, Rivière B, Herrero A, Evert K, Calvisi DF, Augustin J, Leow WQ, Leung HHW, Boleslawski E, Rela M, François A, Cha AWH, Forner A, Reig M, Allaire M, Scatton O, Chatelain D, Boulagnon-Rombi C, Sturm N, Menahem B, Frouin E, Tougeron D, Tournigand C, Kempf E, Kim H, Ningarhari M, Michalak-Provost S, Gopal P, Brustia R, Vibert E, Schulze K, Rüther DF, Weidemann SA, Rhaiem R, Pawlotsky JM, Zhang X, Luciani A, Mulé S, Laurent A, Amaddeo G, Regnault H, De Martin E, Sempoux C, Navale P, Westerhoff M, Lo RCL, Bednarsch J, Gouw A, Guettier C, Lequoy M, Harada K, Sripongpun P, Wetwittayaklang P, Loménie N, Tantipisit J, Kaewdech A, Shen J, Paradis V, Caruso S, Kather JN. Deep learning-based phenotyping reclassifies combined hepatocellular-cholangiocarcinoma. Nat Commun 2023; 14:8290. [PMID: 38092727 PMCID: PMC10719304 DOI: 10.1038/s41467-023-43749-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Primary liver cancer arises either from hepatocytic or biliary lineage cells, giving rise to hepatocellular carcinoma (HCC) or intrahepatic cholangiocarcinoma (ICCA). Combined hepatocellular- cholangiocarcinomas (cHCC-CCA) exhibit equivocal or mixed features of both, causing diagnostic uncertainty and difficulty in determining proper management. Here, we perform a comprehensive deep learning-based phenotyping of multiple cohorts of patients. We show that deep learning can reproduce the diagnosis of HCC vs. CCA with a high performance. We analyze a series of 405 cHCC-CCA patients and demonstrate that the model can reclassify the tumors as HCC or ICCA, and that the predictions are consistent with clinical outcomes, genetic alterations and in situ spatial gene expression profiling. This type of approach could improve treatment decisions and ultimately clinical outcome for patients with rare and biphenotypic cancers such as cHCC-CCA.
Collapse
Affiliation(s)
- Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France.
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France.
- Inserm, U955, Team 18, Créteil, France.
- European Reference Network (ERN) RARE-LIVER, Créteil, France.
| | - Narmin Ghaffari Laleh
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany
| | - Qinghe Zeng
- Centre d'Histologie, d'Imagerie et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France
- Laboratoire d'Informatique Paris Descartes (LIPADE), Université Paris Cité, Paris, France
| | - Pascale Maille
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - Loetitia Favre
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - Anaïs Pujals
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - Christophe Klein
- Centre d'Histologie, d'Imagerie et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France
- INSERM, Sorbonne Université, Université Paris Cité, Paris, France
| | - Céline Bazille
- Caen University Hospital, Department of Pathology, Caen, France
| | - Lara R Heij
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Arnaud Uguen
- CHRU Brest, Department of Pathology, Brest, 29220, France
- Univ Brest, Inserm, CHU de Brest, LBAI, UMR1227, Brest, France
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Luca Di Tommaso
- Department of Pathology, Humanitas University, Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Aurélie Beaufrère
- Assistance Publique-Hôpitaux de Paris, Beaujon University Hospital, Department of Pathology, F-92110, Clichy, France
- Université de Paris, Inflammation Research Center, Inserm, U1149, CNRS, ERL8252, F-75018, Paris, France
| | | | | | - Cong Trung Nguyen
- Department of Pathology, E Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Hiep Nguyen-Canh
- Pathology Center, Bachmai Hospital, Hanoi, Vietnam
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Khuyen Nguyen Thi
- Pathology and Molecular biology Center, National Cancer Hospital, Hanoi, Vietnam
| | - Viviane Gnemmi
- University Lille, UMR9020-U1277, Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
- CHU Lille, Institute of Pathology, Lille, France
| | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Frédéric Charlotte
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Department of Pathology, Paris, France
| | - Dominique Wendum
- Assistance Publique-Hôpitaux de Paris, Saint-Antoine University Hospital, Department of Pathology, Paris, France
| | - Mukul Vij
- Department of Pathology, Dr Rela Institute and Medical Centre, Bharath Institute of Higher Education and Research, Chennai, India
| | - Daniela S Allende
- Department of Hepatobiliary Pathology, Cleveland Clinic Foundation, Cleveland, OH, USA
- Robert J. Tomsich Pathology & Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, L25, Cleveland, OH, 44195, USA
| | - Federico Aucejo
- Department of Gastrointestinal and Hepatobiliary Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Alba Diaz
- Barcelona Clinic Liver Cancer (BCLC) Group, Department of Pathology, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Benjamin Rivière
- Department of Pathology, Gui-de-Chauliac University Hospital, 80, avenue Augustin-Fliche, 34295, Montpellier, France
| | - Astrid Herrero
- Department of Digestive and Hepatobiliary Surgery, Gui-de-Chauliac University Hospital, 80, avenue Augustin-Fliche, 34295, Montpellier, France
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Diego Francesco Calvisi
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Jérémy Augustin
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - Wei Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Howard Ho Wai Leung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | - Mohamed Rela
- Dr Rela Institute and Medical Centre, Bharath Institute of Higher Education and Research, Chennai, India
| | - Arnaud François
- Rouen University Hospital, Department of Pathology, Rouen, France
| | - Anthony Wing-Hung Cha
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Alejandro Forner
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic of Barcelona, IDIBAPS, CIBEREHD, Universidad de Barcelona, Barcelona, Spain
| | - Maria Reig
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic of Barcelona, IDIBAPS, CIBEREHD, Universidad de Barcelona, Barcelona, Spain
| | - Manon Allaire
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Department of Hepatology, Paris, France
| | - Olivier Scatton
- Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Department of Digestive and Hepatobiliary Surgery, Paris, France
| | - Denis Chatelain
- Centre Hospitalier Universitaire d'Amiens, Département de Pathologie, Amiens, France
| | | | - Nathalie Sturm
- Department of Pathology, University Hospital, Grenoble, France
- Translational Innovation in Medicine and Complexity, Centre National de la Recherche Scientifique UMR5525, La Tronche, France
| | - Benjamin Menahem
- Caen University Hospital, Department of Digestive and Hepatobiliary Surgery, Caen, France
| | - Eric Frouin
- Poitiers University Hospital, Department of Pathology, Poitiers, France
- LITEC, Université de Poitiers, Poitiers, France
| | - David Tougeron
- Poitiers University Hospital, Department of Hepatogastroenterology and Oncology, Poitiers, France
| | - Christophe Tournigand
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Medical Oncology, Créteil, France
| | - Emmanuelle Kempf
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Medical Oncology, Créteil, France
| | - Haeryoung Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | | | | | - Purva Gopal
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Raffaele Brustia
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Digestive and Hepatobiliary Surgery, Créteil, France
| | - Eric Vibert
- Assistance Publique-Hôpitaux de Paris, Paul Brousse University Hospital, Department of Digestive and Hepatobiliary Surgery, Paris, France
| | - Kornelius Schulze
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Darius F Rüther
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sören A Weidemann
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rami Rhaiem
- Reims University Hospital, Department of Digestive and Hepatobiliary Surgery, Reims, France
| | - Jean-Michel Pawlotsky
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Inserm, U955, Team 18, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Paul Brousse University Hospital, Department of Digestive and Hepatobiliary Surgery, Paris, France
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Alain Luciani
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Medical Imaging, Créteil, France
| | - Sébastien Mulé
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Medical Imaging, Créteil, France
| | - Alexis Laurent
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Digestive and Hepatobiliary Surgery, Créteil, France
| | - Giuliana Amaddeo
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Hepatology, Créteil, France
| | - Hélène Regnault
- Assistance Publique-Hôpitaux de Paris, Henri Mondor University Hospital, Department of Hepatology, Créteil, France
| | - Eleonora De Martin
- Assistance Publique-Hôpitaux de Paris, Paul Brousse University Hospital, Department of Hepatology, Paris, France
| | - Christine Sempoux
- Institute of Pathology, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Pooja Navale
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Maria Westerhoff
- Department of Pathology University of Michigan, Ann Arbor, MI, USA
| | - Regina Cheuk-Lam Lo
- Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
- State Key Laboratory of Liver Research, (The University of Hong Kong), Pok Fu Lam, Hong Kong, China
| | - Jan Bednarsch
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
| | - Annette Gouw
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Catherine Guettier
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
- Assistance Publique-Hôpitaux de Paris, Paul Brousse University Hospital, Department of Pathology, Villejuif, France
| | - Marie Lequoy
- Assistance Publique-Hôpitaux de Paris, Saint Antoine University Hospital, Department of Hepatology, Paris, France
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Pimsiri Sripongpun
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | | | - Nicolas Loménie
- Laboratoire d'Informatique Paris Descartes (LIPADE), Université Paris Cité, Paris, France
| | - Jarukit Tantipisit
- Prince of Songkla University, Department of Pathology, Hat Yai, Thailand
| | - Apichat Kaewdech
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Jeanne Shen
- Center for Artificial Intelligence in Medicine and Imaging, Stanford University, 1701 Page Mill Road, Palo Alto, CA, 94304, USA
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305, USA
| | - Valérie Paradis
- Assistance Publique-Hôpitaux de Paris, Beaujon University Hospital, Department of Pathology, F-92110, Clichy, France
- Université de Paris, Inflammation Research Center, Inserm, U1149, CNRS, ERL8252, F-75018, Paris, France
| | - Stefano Caruso
- Université Paris Est Créteil, INSERM, IMRB, F-94010, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- Inserm, U955, Team 18, Créteil, France
| | - 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.
| |
Collapse
|
30
|
Tavolara TE, Su Z, Gurcan MN, Niazi MKK. One label is all you need: Interpretable AI-enhanced histopathology for oncology. Semin Cancer Biol 2023; 97:70-85. [PMID: 37832751 DOI: 10.1016/j.semcancer.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
Artificial Intelligence (AI)-enhanced histopathology presents unprecedented opportunities to benefit oncology through interpretable methods that require only one overall label per hematoxylin and eosin (H&E) slide with no tissue-level annotations. We present a structured review of these methods organized by their degree of verifiability and by commonly recurring application areas in oncological characterization. First, we discuss morphological markers (tumor presence/absence, metastases, subtypes, grades) in which AI-identified regions of interest (ROIs) within whole slide images (WSIs) verifiably overlap with pathologist-identified ROIs. Second, we discuss molecular markers (gene expression, molecular subtyping) that are not verified via H&E but rather based on overlap with positive regions on adjacent tissue. Third, we discuss genetic markers (mutations, mutational burden, microsatellite instability, chromosomal instability) that current technologies cannot verify if AI methods spatially resolve specific genetic alterations. Fourth, we discuss the direct prediction of survival to which AI-identified histopathological features quantitatively correlate but are nonetheless not mechanistically verifiable. Finally, we discuss in detail several opportunities and challenges for these one-label-per-slide methods within oncology. Opportunities include reducing the cost of research and clinical care, reducing the workload of clinicians, personalized medicine, and unlocking the full potential of histopathology through new imaging-based biomarkers. Current challenges include explainability and interpretability, validation via adjacent tissue sections, reproducibility, data availability, computational needs, data requirements, domain adaptability, external validation, dataset imbalances, and finally commercialization and clinical potential. Ultimately, the relative ease and minimum upfront cost with which relevant data can be collected in addition to the plethora of available AI methods for outcome-driven analysis will surmount these current limitations and achieve the innumerable opportunities associated with AI-driven histopathology for the benefit of oncology.
Collapse
Affiliation(s)
- Thomas E Tavolara
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ziyu Su
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Metin N Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - M Khalid Khan Niazi
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
31
|
Zhang C, Xu J, Tang R, Yang J, Wang W, Yu X, Shi S. Novel research and future prospects of artificial intelligence in cancer diagnosis and treatment. J Hematol Oncol 2023; 16:114. [PMID: 38012673 PMCID: PMC10680201 DOI: 10.1186/s13045-023-01514-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Research into the potential benefits of artificial intelligence for comprehending the intricate biology of cancer has grown as a result of the widespread use of deep learning and machine learning in the healthcare sector and the availability of highly specialized cancer datasets. Here, we review new artificial intelligence approaches and how they are being used in oncology. We describe how artificial intelligence might be used in the detection, prognosis, and administration of cancer treatments and introduce the use of the latest large language models such as ChatGPT in oncology clinics. We highlight artificial intelligence applications for omics data types, and we offer perspectives on how the various data types might be combined to create decision-support tools. We also evaluate the present constraints and challenges to applying artificial intelligence in precision oncology. Finally, we discuss how current challenges may be surmounted to make artificial intelligence useful in clinical settings in the future.
Collapse
Affiliation(s)
- Chaoyi Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianhui Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
32
|
Chatterji S, Niehues JM, van Treeck M, Loeffler CML, Saldanha OL, Veldhuizen GP, Cifci D, Carrero ZI, Abu-Eid R, Speirs V, Kather JN. Prediction models for hormone receptor status in female breast cancer do not extend to males: further evidence of sex-based disparity in breast cancer. NPJ Breast Cancer 2023; 9:91. [PMID: 37940649 PMCID: PMC10632426 DOI: 10.1038/s41523-023-00599-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
Breast cancer prognosis and management for both men and women are reliant upon estrogen receptor alpha (ERα) and progesterone receptor (PR) expression to inform therapy. Previous studies have shown that there are sex-specific binding characteristics of ERα and PR in breast cancer and, counterintuitively, ERα expression is more common in male than female breast cancer. We hypothesized that these differences could have morphological manifestations that are undetectable to human observers but could be elucidated computationally. To investigate this, we trained attention-based multiple instance learning prediction models for ERα and PR using H&E-stained images of female breast cancer from the Cancer Genome Atlas (TCGA) (n = 1085) and deployed them on external female (n = 192) and male breast cancer images (n = 245). Both targets were predicted in the internal (AUROC for ERα prediction: 0.86 ± 0.02, p < 0.001; AUROC for PR prediction = 0.76 ± 0.03, p < 0.001) and external female cohorts (AUROC for ERα prediction: 0.78 ± 0.03, p < 0.001; AUROC for PR prediction = 0.80 ± 0.04, p < 0.001) but not the male cohort (AUROC for ERα prediction: 0.66 ± 0.14, p = 0.43; AUROC for PR prediction = 0.63 ± 0.04, p = 0.05). This suggests that subtle morphological differences invisible upon visual inspection may exist between the sexes, supporting previous immunohistochemical, genomic, and transcriptomic analyses.
Collapse
Affiliation(s)
- Subarnarekha Chatterji
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Aberdeen Cancer Centre, University of Aberdeen, Aberdeen, UK
| | - Jan Moritz Niehues
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
| | - Marko van Treeck
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
| | - Chiara Maria Lavinia Loeffler
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine, Technical University of Dresden, Dresden, Germany
| | - Oliver Lester Saldanha
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
| | - Gregory Patrick Veldhuizen
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
| | - Didem Cifci
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
| | - Zunamys Itzell Carrero
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
| | - Rasha Abu-Eid
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Aberdeen Cancer Centre, University of Aberdeen, Aberdeen, UK
- Institute of Dentistry, University of Aberdeen, Aberdeen, UK
| | - Valerie Speirs
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
- Aberdeen Cancer Centre, University of Aberdeen, Aberdeen, UK.
| | - Jakob Nikolas Kather
- Else Kröner Fresenius Centre for Digital Health, Carl Gustav Carus Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Department of Medicine III, University Hospital RWTH (Rheinisch-Westfälische Technische Hochschule) Aachen, Aachen, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK
| |
Collapse
|
33
|
Waqas A, Bui MM, Glassy EF, El Naqa I, Borkowski P, Borkowski AA, Rasool G. Revolutionizing Digital Pathology With the Power of Generative Artificial Intelligence and Foundation Models. J Transl Med 2023; 103:100255. [PMID: 37757969 DOI: 10.1016/j.labinv.2023.100255] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/06/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Digital pathology has transformed the traditional pathology practice of analyzing tissue under a microscope into a computer vision workflow. Whole-slide imaging allows pathologists to view and analyze microscopic images on a computer monitor, enabling computational pathology. By leveraging artificial intelligence (AI) and machine learning (ML), computational pathology has emerged as a promising field in recent years. Recently, task-specific AI/ML (eg, convolutional neural networks) has risen to the forefront, achieving above-human performance in many image-processing and computer vision tasks. The performance of task-specific AI/ML models depends on the availability of many annotated training datasets, which presents a rate-limiting factor for AI/ML development in pathology. Task-specific AI/ML models cannot benefit from multimodal data and lack generalization, eg, the AI models often struggle to generalize to new datasets or unseen variations in image acquisition, staining techniques, or tissue types. The 2020s are witnessing the rise of foundation models and generative AI. A foundation model is a large AI model trained using sizable data, which is later adapted (or fine-tuned) to perform different tasks using a modest amount of task-specific annotated data. These AI models provide in-context learning, can self-correct mistakes, and promptly adjust to user feedback. In this review, we provide a brief overview of recent advances in computational pathology enabled by task-specific AI, their challenges and limitations, and then introduce various foundation models. We propose to create a pathology-specific generative AI based on multimodal foundation models and present its potentially transformative role in digital pathology. We describe different use cases, delineating how it could serve as an expert companion of pathologists and help them efficiently and objectively perform routine laboratory tasks, including quantifying image analysis, generating pathology reports, diagnosis, and prognosis. We also outline the potential role that foundation models and generative AI can play in standardizing the pathology laboratory workflow, education, and training.
Collapse
Affiliation(s)
- Asim Waqas
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; Department of Electrical Engineering, University of South Florida, Tampa, Florida.
| | - Marilyn M Bui
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; University of South Florida, Morsani College of Medicine, Tampa, Florida
| | - Eric F Glassy
- Affiliated Pathologists Medical Group, Inc., Rancho Dominguez, California
| | - Issam El Naqa
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Piotr Borkowski
- Quest Diagnostics/Ameripath, Tampa, Florida; Center of Excellence for Digital and AI-Empowered Pathology, Quest Diagnostics, Tampa, Florida
| | - Andrew A Borkowski
- University of South Florida, Morsani College of Medicine, Tampa, Florida; James A. Haley Veterans' Hospital, Tampa, Florida; National Artificial Intelligence Institute, Washington, District of Columbia
| | - Ghulam Rasool
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; Department of Electrical Engineering, University of South Florida, Tampa, Florida; University of South Florida, Morsani College of Medicine, Tampa, Florida; Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
34
|
Ke X, Liu W, Shen L, Zhang Y, Liu W, Wang C, Wang X. Early Screening of Colorectal Precancerous Lesions Based on Combined Measurement of Multiple Serum Tumor Markers Using Artificial Neural Network Analysis. BIOSENSORS 2023; 13:685. [PMID: 37504084 PMCID: PMC10377288 DOI: 10.3390/bios13070685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/29/2023]
Abstract
Many patients with colorectal cancer (CRC) are diagnosed in the advanced stage, resulting in delayed treatment and reduced survival time. It is urgent to develop accurate early screening methods for CRC. The purpose of this study is to develop an artificial intelligence (AI)-based artificial neural network (ANN) model using multiple protein tumor markers to assist in the early diagnosis of CRC and precancerous lesions. In this retrospective analysis, 148 cases with CRC and precancerous diseases were included. The concentrations of multiple protein tumor markers (CEA, CA19-9, CA 125, CYFRA 21-1, CA 72-4, CA 242) were measured by electrochemical luminescence immunoassays. By combining these markers with an ANN algorithm, a diagnosis model (CA6) was developed to distinguish between normal healthy and abnormal subjects, with an AUC of 0.97. The prediction score derived from the CA6 model also performed well in assisting in the diagnosis of precancerous lesions and early CRC (with AUCs of 0.97 and 0.93 and cut-off values of 0.39 and 0.34, respectively), which was better than that of individual protein tumor indicators. The CA6 model established by ANN provides a new and effective method for laboratory auxiliary diagnosis, which might be utilized for early colorectal lesion screening by incorporating more tumor markers with larger sample size.
Collapse
Affiliation(s)
- Xing Ke
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai 200092, China
| | - Wenxue Liu
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai 200092, China
| | - Yue Zhang
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Liu
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co., Ltd., Beijing 100080, China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xu Wang
- Department of Pathology, Ruijin Hospital and College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Nanning Jiuzhouyuan Biotechnology Co., Ltd., Nanning 530007, China
| |
Collapse
|
35
|
Hewitt KJ, Löffler CML, Muti HS, Berghoff AS, Eisenlöffel C, van Treeck M, Carrero ZI, El Nahhas OSM, Veldhuizen GP, Weil S, Saldanha OL, Bejan L, Millner TO, Brandner S, Brückmann S, Kather JN. Direct image to subtype prediction for brain tumors using deep learning. Neurooncol Adv 2023; 5:vdad139. [PMID: 38106649 PMCID: PMC10724115 DOI: 10.1093/noajnl/vdad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Background Deep Learning (DL) can predict molecular alterations of solid tumors directly from routine histopathology slides. Since the 2021 update of the World Health Organization (WHO) diagnostic criteria, the classification of brain tumors integrates both histopathological and molecular information. We hypothesize that DL can predict molecular alterations as well as WHO subtyping of brain tumors from hematoxylin and eosin-stained histopathology slides. Methods We used weakly supervised DL and applied it to three large cohorts of brain tumor samples, comprising N = 2845 patients. Results We found that the key molecular alterations for subtyping, IDH and ATRX, as well as 1p19q codeletion, were predictable from histology with an area under the receiver operating characteristic curve (AUROC) of 0.95, 0.90, and 0.80 in the training cohort, respectively. These findings were upheld in external validation cohorts with AUROCs of 0.90, 0.79, and 0.87 for prediction of IDH, ATRX, and 1p19q codeletion, respectively. Conclusions In the future, such DL-based implementations could ease diagnostic workflows, particularly for situations in which advanced molecular testing is not readily available.
Collapse
Affiliation(s)
- Katherine J Hewitt
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, North Rhine-Westphalia, Germany
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
| | - Chiara M L Löffler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, North Rhine-Westphalia, Germany
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Saxony, Germany
| | - Hannah Sophie Muti
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Dresden, Dresden, Saxony, Germany
| | - Anna Sophie Berghoff
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Vienna, Austria
| | - Christian Eisenlöffel
- Department of Pathology, St. Georg Teaching Hospital, University of Leipzig, Leipzig, Saxony, Germany
| | - Marko van Treeck
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
| | - Zunamys I Carrero
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
| | - Omar S M El Nahhas
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
| | - Gregory P Veldhuizen
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
| | - Sophie Weil
- Neurology Clinic, Department of Neurology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Baden- Württemberg, Germany
- Clinical Cooperation Unit Neuro-oncology, Department of Neurology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Baden- Württemberg, Germany
| | - Oliver Lester Saldanha
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, North Rhine-Westphalia, Germany
| | - Laura Bejan
- School of Medicine, Faculty of Medicine and Dentistry, University College London, London, Greater London, UK
| | - Thomas O Millner
- Division of Neuropathology, Queen Square Institute of Neurology, University College London, London, Greater London, UK
- Blizard Institute, Faculty of Medicine and Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, Greater London, UK
| | - Sebastian Brandner
- Division of Neuropathology, Queen Square Institute of Neurology, University College London, London, Greater London, UK
| | - Sascha Brückmann
- Institut für Pathologie, University Hospital Carl Gustav Carus, Dresden, Saxony, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, North Rhine-Westphalia, Germany
- Clinical Artificial Intelligence, Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Saxony, Germany
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Saxony, Germany
- Pathology & Data Analytics, Faculty of Medicine and Health, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, West Yorkshire, UK
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Baden- Württemberg, Germany
| |
Collapse
|