1
|
Ahmad Z, Al-Thelaya K, Alzubaidi M, Joad F, Gilal NU, Mifsud W, Boughorbel S, Pintore G, Gobbetti E, Schneider J, Agus M. HistoMSC: Density and topology analysis for AI-based visual annotation of histopathology whole slide images. Comput Biol Med 2025; 190:109991. [PMID: 40120181 DOI: 10.1016/j.compbiomed.2025.109991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/20/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
We introduce an end-to-end framework for the automated visual annotation of histopathology whole slide images. Our method integrates deep learning models to achieve precise localization and classification of cell nuclei with spatial data aggregation to extend classes of sparsely distributed nuclei across the entire slide. We introduce a novel and cost-effective approach to localization, leveraging a U-Net architecture and a ResNet-50 backbone. The performance is boosted through color normalization techniques, helping achieve robustness under color variations resulting from diverse scanners and staining reagents. The framework is complemented by a YOLO detection architecture, augmented with generative methods. For classification, we use context patches around each nucleus, fed to various deep architectures. Sparse nuclei-level annotations are then aggregated using kernel density estimation, followed by color-coding and isocontouring. This reduces visual clutter and provides per-pixel probabilities with respect to pathology taxonomies. Finally, we use Morse-Smale theory to generate abstract annotations, highlighting extrema in the density functions and potential spatial interactions in the form of abstract graphs. Thus, our visualization allows for exploration at scales ranging from individual nuclei to the macro-scale. We tested the effectiveness of our framework in an assessment by six pathologists using various neoplastic cases. Our results demonstrate the robustness and usefulness of the proposed framework in aiding histopathologists in their analysis and interpretation of whole slide images.
Collapse
Affiliation(s)
- Zahoor Ahmad
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Khaled Al-Thelaya
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mahmood Alzubaidi
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Faaiz Joad
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Nauman Ullah Gilal
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | | | - Sabri Boughorbel
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | | | | | - Jens Schneider
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Marco Agus
- Division of Information and Computing Technology, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
2
|
Gupta V, Sehrawat TS, Pinzani M, Strazzabosco M. Portal Fibrosis and the Ductular Reaction: Pathophysiological Role in the Progression of Liver Disease and Translational Opportunities. Gastroenterology 2025; 168:675-690. [PMID: 39251168 PMCID: PMC11885590 DOI: 10.1053/j.gastro.2024.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/11/2024]
Abstract
A consistent feature of chronic liver diseases and the hallmark of pathologic repair is the so-called "ductular reaction." This is a histologic abnormality characterized by an expansion of dysmorphic cholangiocytes inside and around portal spaces infiltrated by inflammatory, mesenchymal, and vascular cells. The ductular reaction is a highly regulated response based on the reactivation of morphogenetic signaling mechanisms and a complex crosstalk among a multitude of cell types. The nature and mechanism of these exchanges determine the difference between healthy regenerative liver repair and pathologic repair. An orchestrated signaling among cell types directs mesenchymal cells to deposit a specific extracellular matrix with distinct physical and biochemical properties defined as portal fibrosis. Progression of fibrosis leads to vast architectural and vascular changes known as "liver cirrhosis." The signals regulating the ecology of this microenvironment are just beginning to be addressed. Contrary to the tumor microenvironment, immune modulation inside this "benign" microenvironment is scarcely known. One of the reasons for this is that both the ductular reaction and portal fibrosis have been primarily considered a manifestation of cholestatic liver disease, whereas this phenomenon is also present, albeit with distinctive features, in all chronic human liver diseases. Novel human-derived cellular models and progress in "omics" technologies are increasing our knowledge at a fast pace. Most importantly, this knowledge is on the edge of generating new diagnostic and therapeutic advances. Here, we will critically review the latest advances, in terms of mechanisms, pathophysiology, and treatment prospects. In addition, we will delineate future avenues of research, including innovative translational opportunities.
Collapse
Affiliation(s)
- Vikas Gupta
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Tejasav S Sehrawat
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Massimo Pinzani
- UCL Institute for Liver & Digestive Health, Royal Free Hospital, London, United Kingdom; University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies, Palermo, Italy
| | - Mario Strazzabosco
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
3
|
Acs B, Fend F, Guettier C, L'Imperio V, Montezuma D, Zerbe N, Zlobec I. Debating the pros and cons of computational pathology at the European Congress of Pathology (ECP) 2024. Virchows Arch 2025:10.1007/s00428-025-04084-8. [PMID: 40131426 DOI: 10.1007/s00428-025-04084-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Affiliation(s)
- Balazs Acs
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| | - Falko Fend
- Institute of Pathology and Neuropathology, Tübingen University Hospital and Eberhard Karls University Tübingen, Tübingen, Germany
| | - Catherine Guettier
- Department of Pathology, Hopital Bicêtre, Assistance Publique- Hôpitaux de Paris, Le Kremlin-Bicêtre, France
- Faculté de Médecine, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Pathology, Italy
| | - Diana Montezuma
- Research & Development Unit, IMP Diagnostics, Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPO)@RISE (Health Research Network), Portuguese Oncology Institute of Porto/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC Raquel Seruca), Porto, Portugal
| | - Norman Zerbe
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin, Germany
- Institute of Pathology, Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Inti Zlobec
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, Bern, Switzerland
| |
Collapse
|
4
|
Bessen JL, Alexander M, Foroughi O, Brathwaite R, Baser E, Lee LC, Perez O, Gustavsen G. Perspectives on Reducing Barriers to the Adoption of Digital and Computational Pathology Technology by Clinical Labs. Diagnostics (Basel) 2025; 15:794. [PMID: 40218144 PMCID: PMC11988507 DOI: 10.3390/diagnostics15070794] [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: 02/17/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Digital and computational pathology (DP/CP) tools have the potential to improve the efficiency and accuracy of the anatomic pathology workflow; however, current adoption among US hospital and reference labs remains low. Methods: To better understand the current utilization of DP/CP technology and barriers to widespread adoption, we conducted a survey among 63 anatomic pathologists and lab directors within the US health system. Results: The survey results indicated that current use cases for DP/CP involve streamlining traditional manual pathology and that labs would have substantial difficulty providing AI-guided image analysis if it were required by physicians today. Among potential catalysts for the broader adoption of DP/CP, pathologists identified clinical guidelines as a key resource for anatomic pathology, whose endorsement of DP/CP would be highly impactful for reducing current barriers. Conclusions: Expanded access to DP/CP may ultimately benefit all major stakeholders-patients, physicians, clinical laboratory professionals, care settings, and payers-and will therefore require collaboration across these groups.
Collapse
Affiliation(s)
| | | | | | | | - Emre Baser
- AstraZeneca, Gaithersburg, MD 20878, USA
| | | | - Omar Perez
- AstraZeneca, Gaithersburg, MD 20878, USA
| | | |
Collapse
|
5
|
Macaulay DO, Han W, Zarella MD, Garcia CA, Tavolara TE. Enhancing HER2 testing in breast cancer: predicting fluorescence in situ hybridization (FISH) scores from immunohistochemistry images via deep learning. J Pathol Clin Res 2025; 11:e70024. [PMID: 40050230 PMCID: PMC11884934 DOI: 10.1002/2056-4538.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 03/10/2025]
Abstract
Breast cancer affects millions globally, necessitating precise biomarker testing for effective treatment. HER2 testing is crucial for guiding therapy, particularly with novel antibody-drug conjugates (ADCs) like trastuzumab deruxtecan, which shows promise for breast cancers with low HER2 expression. Current HER2 testing methods, including immunohistochemistry (IHC) and in situ hybridization (ISH), have limitations. IHC, a semi-quantitative assay, is prone to interobserver variability. While ISH provides higher precision than IHC, it remains more resource-intensive in terms of cost and workflow. However, turnaround time is typically faster than that of other advanced molecular methods such as next-generation sequencing. We adapted the clustering-constrained-attention multiple-instance deep learning model to improve IHC testing and reduce dependence on reflex fluorescence ISH (FISH) tests. Using 5,731 HER2 IHC images, including 592 cases with FISH testing, we trained two models: one for predicting HER2 scores from IHC images and another for predicting FISH scores from equivocal cases. The HER2 IHC score prediction model achieved 91% ± 0.01 overall accuracy and a receiver operating characteristic (ROC) area under the curve (AUC) of 0.98 ± 0.01. The FISH score prediction model had an ROC AUC of 0.84 ± 0.07, with sensitivity at 0.37 ± 0.13 and specificity at 0.96 ± 0.03. External validation on cases from 203 institutions showed similar performance. The HER2 IHC model maintained a 91% ± 0.01 accuracy and an ROC AUC of 0.98 ± 0.01, while the FISH model had an ROC AUC of 0.75 ± 0.03, with sensitivity at 0.28 ± 0.04 and specificity at 0.93 ± 0.01. Our model advances HER2 scoring by reducing subjectivity and variability in current scoring methods. Despite lower accuracy and sensitivity in the FISH prediction model, it may be a beneficial option for settings where reflex FISH testing is unavailable or prohibitive. With high specificity, our model can serve as an effective screening tool, enhancing breast cancer diagnosis and treatment selection.
Collapse
Affiliation(s)
- Daniel O Macaulay
- Division of Computational Pathology and AI, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Wenchao Han
- Division of Computational Pathology and AI, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Mark D Zarella
- Division of Computational Pathology and AI, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Chris A Garcia
- Division of Computational Pathology and AI, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| | - Thomas E Tavolara
- Division of Computational Pathology and AI, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| |
Collapse
|
6
|
Liang X, Deng M, Zhu Z, Zhang W, Li Y, Luo J, Wang H, Wu S, Chen R, Wang G, Wu H, Shen C, Hu G, Zhang K, Sun Q, Wang Z. A novel approach for estimating postmortem intervals under varying temperature conditions using pathology images and artificial intelligence models. Int J Legal Med 2025:10.1007/s00414-025-03447-9. [PMID: 40019556 DOI: 10.1007/s00414-025-03447-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 03/01/2025]
Abstract
Estimating the postmortem interval (PMI) is a critical yet complex task in forensic investigations, with accurate and timely determination playing a key role in case resolution and legal outcomes. Traditional methods often suffer from environmental variability and subjective biases, emphasizing the need for more reliable and objective approaches. In this study, we present a novel predictive model for PMI estimation, introduced here for the first time, that leverages pathological tissue images and artificial intelligence (AI). The model is designed to perform under three temperature conditions: 25 °C, 37 °C, and 4 °C. Using a ResNet50 neural network, patch-level images were analyzed to extract deep learning-derived features, which were integrated with machine learning algorithms for whole slide image (WSI) classification. The model achieved strong performance, with micro and macro AUC values of at least 0.949 at the patch-level and 0.800 at the WSI-level in both training and testing sets. In external validation, micro and macro AUC values at the patch-level exceeded 0.960. These results highlight the potential of AI to improve the accuracy and efficiency of PMI estimation. As AI technology continues to advance, this approach holds promise for enhancing forensic investigations and supporting more precise case resolutions.
Collapse
Affiliation(s)
- Xinggong Liang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Mingyan Deng
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Zhengyang Zhu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Wanqing Zhang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuqian Li
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jianliang Luo
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Han Wang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Shuo Wu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Run Chen
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Gongji Wang
- College of Forensic Medicine, Kunming Medical University, Kunming, Yunnan, 650500, People's Republic of China
| | - Hao Wu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chen Shen
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Gengwang Hu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Kai Zhang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Qinru Sun
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China.
| | - Zhenyuan Wang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
7
|
Ge Y, Leng J, Tang Z, Wang K, U K, Zhang SM, Han S, Zhang Y, Xiang J, Yang S, Liu X, Song Y, Wang X, Li Y, Zhao J. Deep Learning-Enabled Integration of Histology and Transcriptomics for Tissue Spatial Profile Analysis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0568. [PMID: 39830364 PMCID: PMC11739434 DOI: 10.34133/research.0568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025]
Abstract
Spatially resolved transcriptomics enable comprehensive measurement of gene expression at subcellular resolution while preserving the spatial context of the tissue microenvironment. While deep learning has shown promise in analyzing SCST datasets, most efforts have focused on sequence data and spatial localization, with limited emphasis on leveraging rich histopathological insights from staining images. We introduce GIST, a deep learning-enabled gene expression and histology integration for spatial cellular profiling. GIST employs histopathology foundation models pretrained on millions of histology images to enhance feature extraction and a hybrid graph transformer model to integrate them with transcriptome features. Validated with datasets from human lung, breast, and colorectal cancers, GIST effectively reveals spatial domains and substantially improves the accuracy of segmenting the microenvironment after denoising transcriptomics data. This enhancement enables more accurate gene expression analysis and aids in identifying prognostic marker genes, outperforming state-of-the-art deep learning methods with a total improvement of up to 49.72%. GIST provides a generalizable framework for integrating histology with spatial transcriptome analysis, revealing novel insights into spatial organization and functional dynamics.
Collapse
Affiliation(s)
- Yongxin Ge
- School of Big Data and Software Engineering,
Chongqing University, Chongqing, China
| | - Jiake Leng
- School of Big Data and Software Engineering,
Chongqing University, Chongqing, China
| | - Ziyang Tang
- Department of Computer and Information Technology,
Purdue University, West Lafayette, IN, USA
| | - Kanran Wang
- Radiation Oncology Center,
Chongqing University Cancer Hospital, Chongqing, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment,
Chongqing University Cancer Hospital, Chongqing, China
| | - Kaicheng U
- Tri-Institutional Computational Biology & Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sophia Meixuan Zhang
- College of Agriculture and Life Sciences,
Cornell University, Ithaca, NY, USA
- Harvard College,
Harvard University, Cambridge, MA, USA
| | - Sen Han
- Division of Genetics, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Yiyan Zhang
- Department of Biostatistics,
University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics,
University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jinxi Xiang
- Department of Radiation Oncology,
Stanford University School of Medicine, Stanford, CA, USA
| | - Sen Yang
- Department of Radiation Oncology,
Stanford University School of Medicine, Stanford, CA, USA
| | - Xiang Liu
- Department of Biostatistics and Health Data Science,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Song
- Department of Neurosurgery,
Chongqing University Three Gorges Hospital, Chongqing, China
| | - Xiyue Wang
- Department of Radiation Oncology,
Stanford University School of Medicine, Stanford, CA, USA
| | - Yuchen Li
- Department of Radiation Oncology,
Stanford University School of Medicine, Stanford, CA, USA
| | - Junhan Zhao
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biomedical Informatics,
Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
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
|
9
|
Shah RM, Shah KM, Bahar P, James CA. Preparing Physicians of the Future: Incorporating Data Science into Medical Education. MEDICAL SCIENCE EDUCATOR 2024; 34:1565-1570. [PMID: 39758456 PMCID: PMC11699019 DOI: 10.1007/s40670-024-02137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 01/07/2025]
Abstract
The recent excitement surrounding artificial intelligence (AI) in health care underscores the importance of physician engagement with new technologies. Future clinicians must develop a strong understanding of data science (DS) to further enhance patient care. However, DS remains largely absent from medical school curricula, even though it is recognized as vital by medical students and residents alike. Here, we evaluate the current DS landscape in medical education and illustrate its impact in medicine through examples in pathology classification and sepsis detection. We also explore reasons for the exclusion of DS and propose solutions to integrate it into existing medical education frameworks.
Collapse
Affiliation(s)
- Rishi M. Shah
- Department of Applied Mathematics, Yale College, New Haven, CT USA
| | - Kavya M. Shah
- Department of Clinical Neurosciences, University of Cambridge, Hills Road, Cambridge, England CB2 0QQ UK
| | - Piroz Bahar
- University of Michigan Medical School, Ann Arbor, MI USA
| | - Cornelius A. James
- Department of Internal Medicine, Pediatrics, and Learning Health Sciences, University of Michigan Medical School, Ann Arbor, MI USA
| |
Collapse
|
10
|
Vigdorovits A, Olteanu GE, Pascalau AV, Pirlog R, Berindan-Neagoe I, Pop OL. Novel Immunohistochemical Profiling of Small-Cell Lung Cancer: Correlations Between Tumor Subtypes and Immune Microenvironment. Diagnostics (Basel) 2024; 14:2660. [PMID: 39682568 DOI: 10.3390/diagnostics14232660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Small-cell lung cancer (SCLC) is a highly aggressive malignancy with an emerging molecular classification based on the expression of the transcription factors ASCL1, NEUROD1, and POU2F3. This study aimed to explore the relationship between these novel subtypes and the tumor immune microenvironment (TIME), particularly CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs). METHODS In 51 cases of patients with SCLC, immunohistochemical (IHC) stains for ASCL1, NEUROD1, POU2F3, CD56, Ki67, CD8, and CD4 were performed. H-scores for the novel transcription factors were calculated to determine tumor subtype. CD8+ and CD4+ TIL counts were averaged across 10 high-power fields. The Kruskal-Wallis test and subsequent post hoc Dunn tests were used to determine the differences in transcription factor expression and TILs across subtypes. RESULTS In our cohort, 68.62% of our cases were SCLC-A, 9.80% were SCLC-N, 7.84% were SCLC-P, and 13.72% were SCLC-I. Significant differences were observed in the expression of ASCL1, NEUROD1, and POU2F3 across subtypes. CD8+ TILs were more abundant in SCLC-P and SCLC-I. CD8+ TILs were negatively correlated with ASCL1 expression (p < 0.05) and positively correlated with POU2F3 expression (p < 0.005). CONCLUSIONS This study highlights the need to integrate the novel SCLC classification with data regarding the TIME to better inform patient prognosis and treatment.
Collapse
Affiliation(s)
- Alon Vigdorovits
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| | - Gheorghe-Emilian Olteanu
- British Columbia Cancer, Department of Pathology, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | | | - Radu Pirlog
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, 94010 Créteil, France
- INSERM U955, Université Paris Est Créteil, 94010 Créteil, France
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Doctoral School, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ovidiu-Laurean Pop
- Department of Morphological Disciplines, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
11
|
Prassas I, Clarke B, Youssef T, Phlamon J, Dimitrakopoulos L, Rofaeil A, Yousef GM. Computational pathology: an evolving concept. Clin Chem Lab Med 2024; 62:2148-2155. [PMID: 38646706 DOI: 10.1515/cclm-2023-1124] [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: 10/24/2023] [Accepted: 04/10/2024] [Indexed: 04/23/2024]
Abstract
The initial enthusiasm about computational pathology (CP) and artificial intelligence (AI) was that they will replace pathologists entirely on the way to fully automated diagnostics. It is becoming clear that currently this is not the immediate model to pursue. On top of the legal and regulatory complexities surrounding its implementation, the majority of tested machine learning (ML)-based predictive algorithms do not display the exquisite performance needed to render them unequivocal, standalone decision makers for matters with direct implications to human health. We are thus moving into a different model of "computer-assisted diagnostics", where AI is there to provide support, rather than replacing, the pathologist. Herein we focus on the practical aspects of CP, from a pathologist perspective. There is a wide range of potential applications where CP can enhance precision of pathology diagnosis, tailor prognostic and predictive information, as well as save time. There are, however, a number of potential limitations for CP that currently hinder their wider adoption in the clinical setting. We address the key necessary steps towards clinical implementation of computational pathology, discuss the significant obstacles that hinders its adoption in the clinical context and summarize some proposed solutions. We conclude that the advancement of CP in the clinic is a promising resource-intensive endeavour that requires broad and inclusive collaborations between academia, industry, and regulatory bodies.
Collapse
Affiliation(s)
- Ioannis Prassas
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Blaise Clarke
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Timothy Youssef
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
| | - Juliana Phlamon
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
| | | | - Andrew Rofaeil
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
| | - George M Yousef
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Chen S, Ding P, Guo H, Meng L, Zhao Q, Li C. Applications of artificial intelligence in digital pathology for gastric cancer. Front Oncol 2024; 14:1437252. [PMID: 39529836 PMCID: PMC11551048 DOI: 10.3389/fonc.2024.1437252] [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: 05/23/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer is one of the most common cancers and is one of the leading causes of cancer-related deaths in worldwide. Early diagnosis and treatment are essential for a positive outcome. The integration of artificial intelligence in the pathology field is increasingly widespread, including histopathological images analysis. In recent years, the application of digital pathology technology emerged as a potential solution to enhance the understanding and management of gastric cancer. Through sophisticated image analysis algorithms, artificial intelligence technologies facilitate the accuracy and sensitivity of gastric cancer diagnosis and treatment and personalized therapeutic strategies. This review aims to evaluate the current landscape and future potential of artificial intelligence in transforming gastric cancer pathology, so as to provide ideas for future research.
Collapse
Affiliation(s)
- Sheng Chen
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
| | - Ping’an Ding
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Honghai Guo
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Lingjiao Meng
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Qun Zhao
- The Third Department of Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, Hebei, China
| | - Cong Li
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
13
|
Kapoor DU, Saini PK, Sharma N, Singh A, Prajapati BG, Elossaily GM, Rashid S. AI illuminates paths in oral cancer: transformative insights, diagnostic precision, and personalized strategies. EXCLI JOURNAL 2024; 23:1091-1116. [PMID: 39391057 PMCID: PMC11464865 DOI: 10.17179/excli2024-7253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Oral cancer retains one of the lowest survival rates worldwide, despite recent therapeutic advancements signifying a tenacious challenge in healthcare. Artificial intelligence exhibits noteworthy potential in escalating diagnostic and treatment procedures, offering promising advancements in healthcare. This review entails the traditional imaging techniques for the oral cancer treatment. The role of artificial intelligence in prognosis of oral cancer including predictive modeling, identification of prognostic factors and risk stratification also discussed significantly in this review. The review also encompasses the utilization of artificial intelligence such as automated image analysis, computer-aided detection and diagnosis integration of machine learning algorithms for oral cancer diagnosis and treatment. The customizing treatment approaches for oral cancer through artificial intelligence based personalized medicine is also part of this review. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Devesh U. Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli-394601, Gujarat, India
| | - Pushpendra Kumar Saini
- Department of Pharmaceutics, Sri Balaji College of Pharmacy, Jaipur, Rajasthan-302013, India
| | - Narendra Sharma
- Department of Pharmaceutics, Sri Balaji College of Pharmacy, Jaipur, Rajasthan-302013, India
| | - Ankul Singh
- Faculty of Pharmacy, Department of Pharmacology, Dr MGR Educational and Research Institute, Velapanchavadi, Chennai-77, Tamil Nadu, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva-384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
14
|
Zhang DY, Venkat A, Khasawneh H, Sali R, Zhang V, Pei Z. Implementation of Digital Pathology and Artificial Intelligence in Routine Pathology Practice. J Transl Med 2024; 104:102111. [PMID: 39053633 DOI: 10.1016/j.labinv.2024.102111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
The advent of affordable technology has significantly influenced the practice of digital pathology, leading to its growing adoption within the pathology community. This review article aimed to outline the latest developments in digital pathology, the cutting-edge advancements in artificial intelligence (AI) applications within this field, and the pertinent United States regulatory frameworks. The content is based on a thorough analysis of original research articles and official United States Federal guidelines. Findings from our review indicate that several Food and Drug Administration-approved digital scanners and image management systems are establishing a solid foundation for the seamless integration of advanced technologies into everyday pathology workflows, which may reduce device and operational costs in the future. AI is particularly transforming the way morphologic diagnoses are automated, notably in cancers like prostate and colorectal, within screening initiatives, albeit challenges such as data privacy issues and algorithmic biases remain. The regulatory environment, shaped by standards from the Food and Drug Administration, Centers for Medicare & Medicaid Services/Clinical Laboratory Improvement Amendments, and College of American Pathologists, is evolving to accommodate these innovations while ensuring safety and reliability. Centers for Medicare & Medicaid Services/Clinical Laboratory Improvement Amendments have issued policies to allow pathologists to review and render diagnoses using digital pathology remotely. Moreover, the introduction of new digital pathology Current Procedural Terminology codes designed to complement existing pathology Current Procedural Terminology codes is facilitating reimbursement processes. Overall, these advancements are heralding a new era in pathology that promises enhanced diagnostic precision and efficiency through digital and AI technologies, potentially improving patient care as well as bolstering educational and research activities.
Collapse
Affiliation(s)
- David Y Zhang
- Department of Computation, NovinoAI, Fort Lauderdale, Florida; Department of Veterans Affairs New York Harbor Healthcare System, New York, New York.
| | - Arsha Venkat
- School of Medicine, New York Medical College, New York, New York
| | - Hamdi Khasawneh
- King Hussein School of Computing Sciences, Princess Sumaya University for Technology, Amman, Jordan
| | - Rasoul Sali
- Department of Computation, NovinoAI, Fort Lauderdale, Florida; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Valerio Zhang
- Department of Computation, NovinoAI, Fort Lauderdale, Florida
| | - Zhiheng Pei
- Department of Veterans Affairs New York Harbor Healthcare System, New York, New York; Department of Pathology, New York University School of Medicine, New York, New York; Department of Medicine, New York University School of Medicine, New York, New York.
| |
Collapse
|
15
|
Sanchez DF, Oliveira P. Pathology of Squamous Cell Carcinoma of the Penis: Back to Square One. Urol Clin North Am 2024; 51:313-325. [PMID: 38925734 DOI: 10.1016/j.ucl.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The landscape of squamous cell carcinoma of the penis (SCC-P) has undergone a significant transformation since the new World Health Organization classification of genitourinary cancers and recent European Association of Urology/American Association of Clinical Oncology guidelines. These changes emphasize the necessity to categorize SCC-P into 2 groups based on its association with human papillomavirus (HPV) infection. This shift has major implications, considering that prior knowledge was derived from a mix of both groups. Given the distinct prognosis, treatment options, and staging systems observed for HPV-associated tumors in other body areas, the question now arises: will similar patterns emerge for SCC-P?
Collapse
Affiliation(s)
- Diego F Sanchez
- Translational Oncogenomics Group, Manchester Cancer Research Centre & CRUK-MI, Wilmslow Road, Manchester M20 4GJ, UK.
| | - Pedro Oliveira
- Department of Pathology, Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
16
|
Ma M, Zeng X, Qu L, Sheng X, Ren H, Chen W, Li B, You Q, Xiao L, Wang Y, Dai M, Zhang B, Lu C, Sheng W, Huang D. Advancing Automatic Gastritis Diagnosis: An Interpretable Multilabel Deep Learning Framework for the Simultaneous Assessment of Multiple Indicators. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1538-1549. [PMID: 38762117 DOI: 10.1016/j.ajpath.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/17/2024] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
The evaluation of morphologic features, such as inflammation, gastric atrophy, and intestinal metaplasia, is crucial for diagnosing gastritis. However, artificial intelligence analysis for nontumor diseases like gastritis is limited. Previous deep learning models have omitted important morphologic indicators and cannot simultaneously diagnose gastritis indicators or provide interpretable labels. To address this, an attention-based multi-instance multilabel learning network (AMMNet) was developed to simultaneously achieve the multilabel diagnosis of activity, atrophy, and intestinal metaplasia with only slide-level weak labels. To evaluate AMMNet's real-world performance, a diagnostic test was designed to observe improvements in junior pathologists' diagnostic accuracy and efficiency with and without AMMNet assistance. In this study of 1096 patients from seven independent medical centers, AMMNet performed well in assessing activity [area under the curve (AUC), 0.93], atrophy (AUC, 0.97), and intestinal metaplasia (AUC, 0.93). The false-negative rates of these indicators were only 0.04, 0.08, and 0.18, respectively, and junior pathologists had lower false-negative rates with model assistance (0.15 versus 0.10). Furthermore, AMMNet reduced the time required per whole slide image from 5.46 to 2.85 minutes, enhancing diagnostic efficiency. In block-level clustering analysis, AMMNet effectively visualized task-related patches within whole slide images, improving interpretability. These findings highlight AMMNet's effectiveness in accurately evaluating gastritis morphologic indicators on multicenter data sets. Using multi-instance multilabel learning strategies to support routine diagnostic pathology deserves further evaluation.
Collapse
Affiliation(s)
- Mengke Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Xixi Zeng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Linhao Qu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Xia Sheng
- Department of Pathology, Minhang Hospital, Fudan University, Shanghai, China
| | - Hongzheng Ren
- Department of Pathology, Gongli Hospital, Naval Medical University, Shanghai, China
| | - Weixiang Chen
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Pathology, Shanghai Xu-Hui Central Hospital, Shanghai, China
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Li Xiao
- Department of Pathology, Huadong Hospital, Shanghai, China
| | - Yi Wang
- Information Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mei Dai
- Information Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Boqiang Zhang
- Shanghai Foremost Medical Technology Co. Ltd., Shanghai, China
| | - Changqing Lu
- Shanghai Foremost Medical Technology Co. Ltd., Shanghai, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China.
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Rizk PA, Gonzalez MR, Galoaa BM, Girgis AG, Van Der Linden L, Chang CY, Lozano-Calderon SA. Machine Learning-Assisted Decision Making in Orthopaedic Oncology. JBJS Rev 2024; 12:01874474-202407000-00005. [PMID: 38991098 DOI: 10.2106/jbjs.rvw.24.00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
» Artificial intelligence is an umbrella term for computational calculations that are designed to mimic human intelligence and problem-solving capabilities, although in the future, this may become an incomplete definition. Machine learning (ML) encompasses the development of algorithms or predictive models that generate outputs without explicit instructions, assisting in clinical predictions based on large data sets. Deep learning is a subset of ML that utilizes layers of networks that use various inter-relational connections to define and generalize data.» ML algorithms can enhance radiomics techniques for improved image evaluation and diagnosis. While ML shows promise with the advent of radiomics, there are still obstacles to overcome.» Several calculators leveraging ML algorithms have been developed to predict survival in primary sarcomas and metastatic bone disease utilizing patient-specific data. While these models often report exceptionally accurate performance, it is crucial to evaluate their robustness using standardized guidelines.» While increased computing power suggests continuous improvement of ML algorithms, these advancements must be balanced against challenges such as diversifying data, addressing ethical concerns, and enhancing model interpretability.
Collapse
Affiliation(s)
- Paul A Rizk
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marcos R Gonzalez
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bishoy M Galoaa
- Interdisciplinary Science & Engineering Complex (ISEC), Northeastern University, Boston, Massachusetts
| | - Andrew G Girgis
- Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Lotte Van Der Linden
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Connie Y Chang
- Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Santiago A Lozano-Calderon
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Bian C, Ashton G, Grant M, Rodriguez VP, Martin IP, Tsakiroglou AM, Cook M, Fergie M. Integrating Spatial and Morphological Characteristics into Melanoma Prognosis: A Computational Approach. Cancers (Basel) 2024; 16:2026. [PMID: 38893146 PMCID: PMC11171264 DOI: 10.3390/cancers16112026] [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: 04/19/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
In this study, the prognostic value of cellular morphology and spatial configurations in melanoma has been examined, aiming to complement traditional prognostic indicators like mitotic activity and tumor thickness. Through a computational pipeline using machine learning and deep learning methods, we quantified nuclei sizes within different spatial regions and analyzed their prognostic significance using univariate and multivariate Cox models. Nuclei sizes in the invasive band demonstrated a significant hazard ratio (HR) of 1.1 (95% CI: 1.03, 1.18). Similarly, the nuclei sizes of tumor cells and Ki67 S100 co-positive cells in the invasive band achieved HRs of 1.07 (95% CI: 1.02, 1.13) and 1.09 (95% CI: 1.04, 1.16), respectively. Our findings reveal that nuclei sizes, particularly in the invasive band, are potentially prognostic factors. Correlation analyses further demonstrated a meaningful relationship between cellular morphology and tumor progression, notably showing that nuclei size within the invasive band correlates substantially with tumor thickness. These results suggest the potential of integrating spatial and morphological analyses into melanoma prognostication.
Collapse
Affiliation(s)
- Chang Bian
- The Division of Informatics, Imaging and Data Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Garry Ashton
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Megan Grant
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Valeria Pavet Rodriguez
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Isabel Peset Martin
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Anna Maria Tsakiroglou
- The Division of Informatics, Imaging and Data Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Martin Cook
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
- Royal Surrey County Hospital, Guildford GU2 7XX, UK
| | - Martin Fergie
- The Division of Informatics, Imaging and Data Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
19
|
Ivanova M, Pescia C, Trapani D, Venetis K, Frascarelli C, Mane E, Cursano G, Sajjadi E, Scatena C, Cerbelli B, d’Amati G, Porta FM, Guerini-Rocco E, Criscitiello C, Curigliano G, Fusco N. Early Breast Cancer Risk Assessment: Integrating Histopathology with Artificial Intelligence. Cancers (Basel) 2024; 16:1981. [PMID: 38893102 PMCID: PMC11171409 DOI: 10.3390/cancers16111981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Effective risk assessment in early breast cancer is essential for informed clinical decision-making, yet consensus on defining risk categories remains challenging. This paper explores evolving approaches in risk stratification, encompassing histopathological, immunohistochemical, and molecular biomarkers alongside cutting-edge artificial intelligence (AI) techniques. Leveraging machine learning, deep learning, and convolutional neural networks, AI is reshaping predictive algorithms for recurrence risk, thereby revolutionizing diagnostic accuracy and treatment planning. Beyond detection, AI applications extend to histological subtyping, grading, lymph node assessment, and molecular feature identification, fostering personalized therapy decisions. With rising cancer rates, it is crucial to implement AI to accelerate breakthroughs in clinical practice, benefiting both patients and healthcare providers. However, it is important to recognize that while AI offers powerful automation and analysis tools, it lacks the nuanced understanding, clinical context, and ethical considerations inherent to human pathologists in patient care. Hence, the successful integration of AI into clinical practice demands collaborative efforts between medical experts and computational pathologists to optimize patient outcomes.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Cristian Scatena
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Bruna Cerbelli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy;
| | - Giulia d’Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| | - Francesca Maria Porta
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
20
|
Hu S, Xiao Q, Gao R, Qin J, Nie J, Chen Y, Lou J, Ding M, Pan Y, Wang S. Identification of BGN positive fibroblasts as a driving factor for colorectal cancer and development of its related prognostic model combined with machine learning. BMC Cancer 2024; 24:516. [PMID: 38654221 PMCID: PMC11041013 DOI: 10.1186/s12885-024-12251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Numerous studies have indicated that cancer-associated fibroblasts (CAFs) play a crucial role in the progression of colorectal cancer (CRC). However, there are still many unknowns regarding the exact role of CAF subtypes in CRC. METHODS The data for this study were obtained from bulk, single-cell, and spatial transcriptomic sequencing data. Bioinformatics analysis, in vitro experiments, and machine learning methods were employed to investigate the functional characteristics of CAF subtypes and construct prognostic models. RESULTS Our study demonstrates that Biglycan (BGN) positive cancer-associated fibroblasts (BGN + Fib) serve as a driver in colorectal cancer (CRC). The proportion of BGN + Fib increases gradually with the progression of CRC, and high infiltration of BGN + Fib is associated with poor prognosis in terms of overall survival (OS) and recurrence-free survival (RFS) in CRC. Downregulation of BGN expression in cancer-associated fibroblasts (CAFs) significantly reduces migration and proliferation of CRC cells. Among 101 combinations of 10 machine learning algorithms, the StepCox[both] + plsRcox combination was utilized to develop a BGN + Fib derived risk signature (BGNFRS). BGNFRS was identified as an independent adverse prognostic factor for CRC OS and RFS, outperforming 92 previously published risk signatures. A Nomogram model constructed based on BGNFRS and clinical-pathological features proved to be a valuable tool for predicting CRC prognosis. CONCLUSION In summary, our study identified BGN + Fib as drivers of CRC, and the derived BGNFRS was effective in predicting the OS and RFS of CRC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- School of Medicine, Southeast University, 210009, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, 210006, Nanjing, Jiangsu, China
| | - Qianni Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China
| | - Rui Gao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China
| | - Jian Qin
- School of Medicine, Southeast University, 210009, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, 210006, Nanjing, Jiangsu, China
| | - Junjie Nie
- School of Medicine, Southeast University, 210009, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, 210006, Nanjing, Jiangsu, China
| | - Yuhan Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China
| | - Jinwei Lou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China
| | - Muzi Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, 210006, Nanjing, Jiangsu, China.
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, 211100, Nanjing, Jiangsu, China.
| | - Shukui Wang
- School of Medicine, Southeast University, 210009, Nanjing, Jiangsu, China.
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, No. 68, Changle Road, 210006, Nanjing, Jiangsu, China.
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 211122, Nanjing, Jiangsu, China.
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, 211100, Nanjing, Jiangsu, China.
| |
Collapse
|
21
|
Cai X, Zhang H, Wang Y, Zhang J, Li T. Digital pathology-based artificial intelligence models for differential diagnosis and prognosis of sporadic odontogenic keratocysts. Int J Oral Sci 2024; 16:16. [PMID: 38403665 PMCID: PMC10894880 DOI: 10.1038/s41368-024-00287-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/24/2023] [Accepted: 02/01/2024] [Indexed: 02/27/2024] Open
Abstract
Odontogenic keratocyst (OKC) is a common jaw cyst with a high recurrence rate. OKC combined with basal cell carcinoma as well as skeletal and other developmental abnormalities is thought to be associated with Gorlin syndrome. Moreover, OKC needs to be differentiated from orthokeratinized odontogenic cyst and other jaw cysts. Because of the different prognosis, differential diagnosis of several cysts can contribute to clinical management. We collected 519 cases, comprising a total of 2 157 hematoxylin and eosin-stained images, to develop digital pathology-based artificial intelligence (AI) models for the diagnosis and prognosis of OKC. The Inception_v3 neural network was utilized to train and test models developed from patch-level images. Finally, whole slide image-level AI models were developed by integrating deep learning-generated pathology features with several machine learning algorithms. The AI models showed great performance in the diagnosis (AUC = 0.935, 95% CI: 0.898-0.973) and prognosis (AUC = 0.840, 95%CI: 0.751-0.930) of OKC. The advantages of multiple slides model for integrating of histopathological information are demonstrated through a comparison with the single slide model. Furthermore, the study investigates the correlation between AI features generated by deep learning and pathological findings, highlighting the interpretative potential of AI models in the pathology. Here, we have developed the robust diagnostic and prognostic models for OKC. The AI model that is based on digital pathology shows promise potential for applications in odontogenic diseases of the jaw.
Collapse
Affiliation(s)
- Xinjia Cai
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Heyu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yanjin Wang
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| | - Jianyun Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing, China.
| | - Tiejun Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing, China.
| |
Collapse
|
22
|
Brancato V, Esposito G, Coppola L, Cavaliere C, Mirabelli P, Scapicchio C, Borgheresi R, Neri E, Salvatore M, Aiello M. Standardizing digital biobanks: integrating imaging, genomic, and clinical data for precision medicine. J Transl Med 2024; 22:136. [PMID: 38317237 PMCID: PMC10845786 DOI: 10.1186/s12967-024-04891-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
Advancements in data acquisition and computational methods are generating a large amount of heterogeneous biomedical data from diagnostic domains such as clinical imaging, pathology, and next-generation sequencing (NGS), which help characterize individual differences in patients. However, this information needs to be available and suitable to promote and support scientific research and technological development, supporting the effective adoption of the precision medicine approach in clinical practice. Digital biobanks can catalyze this process, facilitating the sharing of curated and standardized imaging data, clinical, pathological and molecular data, crucial to enable the development of a comprehensive and personalized data-driven diagnostic approach in disease management and fostering the development of computational predictive models. This work aims to frame this perspective, first by evaluating the state of standardization of individual diagnostic domains and then by identifying challenges and proposing a possible solution towards an integrative approach that can guarantee the suitability of information that can be shared through a digital biobank. Our analysis of the state of the art shows the presence and use of reference standards in biobanks and, generally, digital repositories for each specific domain. Despite this, standardization to guarantee the integration and reproducibility of the numerical descriptors generated by each domain, e.g. radiomic, pathomic and -omic features, is still an open challenge. Based on specific use cases and scenarios, an integration model, based on the JSON format, is proposed that can help address this problem. Ultimately, this work shows how, with specific standardization and promotion efforts, the digital biobank model can become an enabling technology for the comprehensive study of diseases and the effective development of data-driven technologies at the service of precision medicine.
Collapse
Affiliation(s)
| | - Giuseppina Esposito
- Bio Check Up S.R.L, 80121, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | | | | | - Peppino Mirabelli
- UOS Laboratori di Ricerca e Biobanca, AORN Santobono-Pausilipon, Via Teresa Ravaschieri, 8, 80122, Naples, Italy
| | - Camilla Scapicchio
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | - Rita Borgheresi
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | - Emanuele Neri
- Academic Radiology, Department of Translational Research, University of Pisa, via Roma, 67, 56126, Pisa, Italy
| | | | | |
Collapse
|
23
|
Jones JL, Poulsom R, Coates PJ. Recent Advances in Pathology: the 2023 Annual Review Issue of The Journal of Pathology. J Pathol 2023; 260:495-497. [PMID: 37580852 DOI: 10.1002/path.6192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/16/2023]
Abstract
The 2023 Annual Review Issue of The Journal of Pathology, Recent Advances in Pathology, contains 12 invited reviews on topics of current interest in pathology. This year, our subjects include immuno-oncology and computational pathology approaches for diagnostic and research applications in human disease. Reviews on the tissue microenvironment include the effects of apoptotic cell-derived exosomes, how understanding the tumour microenvironment predicts prognosis, and the growing appreciation of the diverse functions of fibroblast subtypes in health and disease. We also include up-to-date reviews of modern aspects of the molecular basis of malignancies, and our final review covers new knowledge of vascular and lymphatic regeneration in cardiac disease. All of the reviews contained in this issue are written by expert groups of authors selected to discuss the recent progress in their particular fields and all articles are freely available online (https://pathsocjournals.onlinelibrary.wiley.com/journal/10969896). © 2023 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Richard Poulsom
- The Pathological Society of Great Britain and Ireland, London, UK
| | - Philip J Coates
- Research Center for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|