1
|
Cannarozzi AL, Biscaglia G, Parente P, Latiano TP, Gentile A, Ciardiello D, Massimino L, Di Brina ALP, Guerra M, Tavano F, Ungaro F, Bossa F, Perri F, Latiano A, Palmieri O. Artificial intelligence and whole slide imaging, a new tool for the microsatellite instability prediction in colorectal cancer: Friend or foe? Crit Rev Oncol Hematol 2025; 210:104694. [PMID: 40064251 DOI: 10.1016/j.critrevonc.2025.104694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common and second most deadly cancer worldwide. Despite advances in screening and treatment, CRC is heterogeneous and the response to therapy varies significantly, limiting personalized treatment options. Certain molecular biomarkers, including microsatellite instability (MSI), are critical in planning personalized treatment, although only a subset of patients may benefit. Currently, the primary methods for assessing MSI status include immunohistochemistry (IHC) for DNA mismatch repair proteins (MMRs), polymerase chain reaction (PCR)-based molecular testing, or next-generation sequencing (NGS). However, these techniques have limitations, are expensive and time-consuming, and often result in inter-method inconsistencies. Deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H) are critical predictive biomarkers of response to immune checkpoint inhibitor (ICI) therapy and MSI testing is recommended to identify patients who may benefit. There is a pressing need for a more robust, reliable, and cost-effective approach that accurately assesses MSI status. Recent advances in computational pathology, in particular the development of technologies that digitally scan whole slide images (WSI) at high resolution, as well as new approaches to artificial intelligence (AI) in medicine, are increasingly gaining ground. This review aims to provide an overview of the latest findings on WSI and advances in AI methods for predicting MSI status, summarize their applications in CRC, and discuss their strengths and limitations in daily clinical practice.
Collapse
Affiliation(s)
- Anna Lucia Cannarozzi
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Giuseppe Biscaglia
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo 71013, Italy.
| | - Tiziana Pia Latiano
- Oncology Unit, Fondazione Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo 71013, Italy.
| | - Annamaria Gentile
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan.
| | - Luca Massimino
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Anna Laura Pia Di Brina
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Maria Guerra
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesca Tavano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Federica Ungaro
- Gastroenterology and Digestive Endoscopy Department, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Fabrizio Bossa
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Francesco Perri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Anna Latiano
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - Orazio Palmieri
- Division of Gastroenterology, Fondazione IRCCS - Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
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
|
Marra A, Morganti S, Pareja F, Campanella G, Bibeau F, Fuchs T, Loda M, Parwani A, Scarpa A, Reis-Filho JS, Curigliano G, Marchiò C, Kather JN. Artificial intelligence entering the pathology arena in oncology: current applications and future perspectives. Ann Oncol 2025:S0923-7534(25)00112-7. [PMID: 40307127 DOI: 10.1016/j.annonc.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/19/2025] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Artificial intelligence (AI) is rapidly transforming the fields of pathology and oncology, offering novel opportunities for advancing diagnosis, prognosis, and treatment of cancer. METHODS Through a systematic review-based approach, the representatives from the European Society for Medical Oncology (ESMO) Precision Oncology Working Group (POWG) and international experts identified studies in pathology and oncology that applied AI-based algorithms for tumour diagnosis, molecular biomarker detection, and cancer prognosis assessment. These findings were synthesised to provide a comprehensive overview of current AI applications and future directions in cancer pathology. RESULTS The integration of AI tools in digital pathology is markedly improving the accuracy and efficiency of image analysis, allowing for automated tumour detection and classification, identification of prognostic molecular biomarkers, and prediction of treatment response and patient outcomes. Several barriers for the adoption of AI in clinical workflows, such as data availability, explainability, and regulatory considerations, still persist. There are currently no prognostic or predictive AI-based biomarkers supported by level IA or IB evidence. The ongoing advancements in AI algorithms, particularly foundation models, generalist models and transformer-based deep learning, offer immense promise for the future of cancer research and care. AI is also facilitating the integration of multi-omics data, leading to more precise patient stratification and personalised treatment strategies. CONCLUSIONS The application of AI in pathology is poised to not only enhance the accuracy and efficiency of cancer diagnosis and prognosis but also facilitate the development of personalised treatment strategies. Although barriers to implementation remain, ongoing research and development in this field coupled with addressing ethical and regulatory considerations will likely lead to a future where AI plays an integral role in cancer management and precision medicine. The continued evolution and adoption of AI in pathology and oncology are anticipated to reshape the landscape of cancer care, heralding a new era of precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- A Marra
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - S Morganti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Department of Medicine, Harvard Medical School, Boston, USA; Gerstner Center for Cancer Diagnostics, Broad Institute of MIT and Harvard, Boston, USA
| | - F Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Campanella
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - F Bibeau
- Department of Pathology, University Hospital of Besançon, Besancon, France
| | - T Fuchs
- Hasso Plattner Institute for Digital Health, Mount Sinai Medical School, New York, USA; Department of AI and Human Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, USA; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, USA
| | - A Parwani
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, USA
| | - A Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University of Verona, Verona, Italy
| | - J S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - C Marchiò
- Candiolo Cancer Institute, FPO IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - J N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany; Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
4
|
Holowatyj AN, Overman MJ, Votanopoulos KI, Lowy AM, Wagner P, Washington MK, Eng C, Foo WC, Goldberg RM, Hosseini M, Idrees K, Johnson DB, Shergill A, Ward E, Zachos NC, Shelton D. Defining a 'cells to society' research framework for appendiceal tumours. Nat Rev Cancer 2025; 25:293-315. [PMID: 39979656 DOI: 10.1038/s41568-024-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/22/2025]
Abstract
Tumours of the appendix - a vestigial digestive organ attached to the colon - are rare. Although we estimate that around 3,000 new appendiceal cancer cases are diagnosed annually in the USA, the challenges of accurately diagnosing and identifying this tumour type suggest that this number may underestimate true population incidence. In the current absence of disease-specific screening and diagnostic imaging modalities, or well-established risk factors, the incidental discovery of appendix tumours is often prompted by acute presentations mimicking appendicitis or when the tumour has already spread into the abdominal cavity - wherein the potential misclassification of appendiceal tumours as malignancies of the colon and ovaries also increases. Notwithstanding these diagnostic difficulties, our understanding of appendix carcinogenesis has advanced in recent years. However, there persist considerable challenges to accelerating the pace of research discoveries towards the path to improved treatments and cures for patients with this group of orphan malignancies. The premise of this Expert Recommendation article is to discuss the current state of the field, to delineate unique challenges for the study of appendiceal tumours, and to propose key priority research areas that will deliver a more complete picture of appendix carcinogenesis and metastasis. The Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation Scientific Think Tank delivered a consensus of core research priorities for appendiceal tumours that are poised to be ground-breaking and transformative for scientific discovery and innovation. On the basis of these six research areas, here, we define the first 'cells to society' research framework for appendix tumours.
Collapse
Affiliation(s)
- Andreana N Holowatyj
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Patrick Wagner
- Division of Surgical Oncology, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Mary K Washington
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cathy Eng
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mojgan Hosseini
- Department of Pathology, University of California, San Diego, San Diego, CA, USA
| | - Kamran Idrees
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ardaman Shergill
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Erin Ward
- Section of Surgical Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicholas C Zachos
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deborah Shelton
- Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation, Springfield, PA, USA
| |
Collapse
|
5
|
Chen L, Xiao H, Jiang J, Li B, Liu W, Huang W. The KMeansGraphMIL Model: A Weakly Supervised Multiple Instance Learning Model for Predicting Colorectal Cancer Tumor Mutational Burden. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:671-679. [PMID: 39800053 DOI: 10.1016/j.ajpath.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 01/15/2025]
Abstract
Colorectal cancer (CRC) is one of the top three most lethal malignancies worldwide, posing a significant threat to human health. Recently proposed immunotherapy checkpoint blockade treatments have proven effective for CRC, but their use depends on measuring specific biomarkers in patients. Among these biomarkers, tumor mutational burden (TMB) has emerged as a novel indicator, traditionally requiring next-generation sequencing for measurement, which is time-consuming, labor intensive, and costly. To provide an economical and rapid way to predict patients' TMB, the KMeansGraphMIL model was proposed based on weakly supervised multiple-instance learning. Compared with previous weakly supervised multiple-instance learning models, KMeansGraphMIL leveraged both the similarity of image patch feature vectors and the spatial relationships between patches. This approach improved the model's area under the receiver operating characteristic curve to 0.8334 and significantly increased the recall to 0.7556. Thus, this study presents an economical and rapid framework for predicting CRC TMB, offering the potential for physicians to quickly develop treatment plans and saving patients substantial time and money.
Collapse
Affiliation(s)
- Linghao Chen
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Huiling Xiao
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jiale Jiang
- Department of Medical Imaging, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Bing Li
- Department of Medical Imaging, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Weixiang Liu
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen, China.
| | - Wensheng Huang
- Radiology Department, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
6
|
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
|
7
|
Cai Y, Chen X, Chen J, Liao J, Han M, Lin D, Hong X, Hu H, Hu J. Deep learning-assisted colonoscopy images for prediction of mismatch repair deficiency in colorectal cancer. Surg Endosc 2025; 39:859-867. [PMID: 39623175 DOI: 10.1007/s00464-024-11426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Deficient mismatch repair or microsatellite instability is a major predictive biomarker for the efficacy of immune checkpoint inhibitors of colorectal cancer. However, routine testing has not been uniformly implemented due to cost and resource constraints. METHODS We developed and validated a deep learning-based classifiers to detect mismatch repair-deficient status from routine colonoscopy images. We obtained the colonoscopy images from the imaging database at Endoscopic Center of the Sixth Affiliated Hospital, Sun Yat-sen University. Colonoscopy images from a prospective trial (Neoadjuvant PD-1 blockade by toripalimab with or without celecoxib in mismatch repair-deficient or microsatellite instability-high locally advanced colorectal cancer) were used to test the model. RESULTS A total of 5226 eligible images from 892 tumors from the consecutive patients were utilized to develop and validate the deep learning model. 2105 colorectal cancer images from 306 tumors were randomly selected to form model development dataset with a class-balanced approach. 3121 images of 488 proficient mismatch repair tumors and 98 deficient mismatch repair tumors were used to form the independent dataset. The model achieved an AUROC of 0.948 (95% CI 0.919-0.977) on the test dataset. On the independent validation dataset, the AUROC was 0.807 (0.760-0.854), and the NPV in was 94.2% (95% CI 0.918-0.967). On the prospective trial dataset, the model identified 29 tumors among the 33 deficient mismatch repair tumors (87.88%). CONCLUSIONS The model achieved a high NPV in detecting deficient mismatch repair colorectal cancers. This model might serve as an automatic screening tool.
Collapse
Affiliation(s)
- Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
| | - Xijie Chen
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Gastric Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Junguo Chen
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - James Liao
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Ming Han
- Guangzhou Aptiligent Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Dezheng Lin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoling Hong
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
| | - Jiancong Hu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Chen W, Zheng K, Yuan W, Jia Z, Wu Y, Duan X, Yang W, Wen Z, Zhong L, Liu X. A CT-based deep learning for segmenting tumors and predicting microsatellite instability in patients with colorectal cancers: a multicenter cohort study. LA RADIOLOGIA MEDICA 2025; 130:214-225. [PMID: 39586941 DOI: 10.1007/s11547-024-01909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE To develop and validate deep learning (DL) models using preoperative contrast-enhanced CT images for tumor auto-segmentation and microsatellite instability (MSI) prediction in colorectal cancer (CRC). MATERIALS AND METHODS Patients with CRC who underwent surgery or biopsy between January 2018 and April 2023 were retrospectively enrolled. Mismatch repair protein expression was determined via immunohistochemistry or fluorescence multiplex polymerase chain reaction-capillary electrophoresis. Manually delineated tumor contours using arterial and venous phase CT images by three abdominal radiologists are served as ground truth. Tumor auto-segmentation used nnU-Net. MSI prediction employed ViT or convolutional neural networks models, trained and validated with arterial and venous phase images (image model) or combined clinical-pathological factors (combined model). The segmentation model was evaluated using patch coverage ratio, Dice coefficient, recall, precision, and F1-score. The predictive models' efficacy was assessed using areas under the curves and decision curve analysis. RESULTS Overall, 2180 patients (median age: 61 years ± 17 [SD]; 1285 males) were divided into training (n = 1159), validation (n = 289), and independent external test (n = 732) groups. High-level MSI status was present in 435 patients (20%). In the external test set, the segmentation model performed well in the arterial phase, with patch coverage ratio, Dice coefficient, recall, precision, and F1-score values of 0.87, 0.71, 0.72, 0.74, and 0.71, respectively. For MSI prediction, the combined models outperformed the clinical model (AUC = 0.83 and 0.82 vs 0.67, p < 0.001) and two image models (AUC = 0.75 and 0.77, p < 0.001). Decision curve analysis confirmed the higher net benefit of the combined model compared to the other models across probability thresholds ranging from 0.1 to 0.45. CONCLUSION DL enhances tumor segmentation efficiency and, when integrated with contrast-enhanced CT and clinicopathological factors, exhibits good diagnostic performance in predicting MSI in CRC.
Collapse
Affiliation(s)
- Weicui Chen
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Kaiyi Zheng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenjing Yuan
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Ziqi Jia
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Yuankui Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaohui Duan
- Radiology Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Wei Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhibo Wen
- Radiology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Liming Zhong
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xian Liu
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
9
|
Hezi H, Gelber M, Balabanov A, Maruvka YE, Freiman M. CIMIL-CRC: A clinically-informed multiple instance learning framework for patient-level colorectal cancer molecular subtypes classification from H&E stained images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 259:108513. [PMID: 39581068 DOI: 10.1016/j.cmpb.2024.108513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND AND OBJECTIVE Treatment approaches for colorectal cancer (CRC) are highly dependent on the molecular subtype, as immunotherapy has shown efficacy in cases with microsatellite instability (MSI) but is ineffective for the microsatellite stable (MSS) subtype. There is promising potential in utilizing deep neural networks (DNNs) to automate the differentiation of CRC subtypes by analyzing hematoxylin and eosin (H&E) stained whole-slide images (WSIs). Due to the extensive size of WSIs, multiple instance learning (MIL) techniques are typically explored. However, existing MIL methods focus on identifying the most representative image patches for classification, which may result in the loss of critical information. Additionally, these methods often overlook clinically relevant information, like the tendency for MSI class tumors to predominantly occur on the proximal (right side) colon. METHODS We introduce 'CIMIL-CRC', a DNN framework that: (1) solves the MSI/MSS MIL problem by efficiently combining a pre-trained feature extraction model with principal component analysis (PCA) to aggregate information from all patches, and (2) integrates clinical priors, particularly the tumor location within the colon, into the model to enhance patient-level classification accuracy. We assessed our CIMIL-CRC method using the average area under the receiver operating characteristic curve (AUROC) from a 5-fold cross-validation experimental setup for model development on the TCGA-CRC-DX cohort, contrasting it with a baseline patch-level classification, a MIL-only approach, and a clinically-informed patch-level classification approach. RESULTS Our CIMIL-CRC outperformed all methods (AUROC: 0.92±0.002 (95% CI 0.91-0.92), vs. 0.79±0.02 (95% CI 0.76-0.82), 0.86±0.01 (95% CI 0.85-0.88), and 0.87±0.01 (95% CI 0.86-0.88), respectively). The improvement was statistically significant. To the best of our knowledge, this is the best result achieved for MSI/MSS classification on this dataset. CONCLUSION Our CIMIL-CRC method holds promise for offering insights into the key representations of histopathological images and suggests a straightforward implementation.
Collapse
Affiliation(s)
- Hadar Hezi
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Matan Gelber
- Faculty of Electrical and Computer Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexander Balabanov
- Faculty of Electrical and Computer Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yosef E Maruvka
- Faculty of Food Engineering and Biotechnology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Moti Freiman
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
10
|
Trahearn N, Sakr C, Banerjee A, Lee SH, Baker A, Kocher HM, Angerilli V, Morano F, Bergamo F, Maddalena G, Intini R, Cremolini C, Caravagna G, Graham T, Pietrantonio F, Lonardi S, Fassan M, Sottoriva A. Computational pathology applied to clinical colorectal cancer cohorts identifies immune and endothelial cell spatial patterns predictive of outcome. J Pathol 2025; 265:198-210. [PMID: 39788558 PMCID: PMC11717494 DOI: 10.1002/path.6378] [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: 07/16/2024] [Revised: 10/04/2024] [Accepted: 11/06/2024] [Indexed: 01/12/2025]
Abstract
Colorectal cancer (CRC) is a histologically heterogeneous disease with variable clinical outcome. The role the tumour microenvironment (TME) plays in determining tumour progression is complex and not fully understood. To improve our understanding, it is critical that the TME is studied systematically within clinically annotated patient cohorts with long-term follow-up. Here we studied the TME in three clinical cohorts of metastatic CRC with diverse molecular subtype and treatment history. The MISSONI cohort included cases with microsatellite instability that received immunotherapy (n = 59, 24 months median follow-up). The BRAF cohort included BRAF V600E mutant microsatellite stable (MSS) cancers (n = 141, 24 months median follow-up). The VALENTINO cohort included RAS/RAF WT MSS cases who received chemotherapy and anti-EGFR therapy (n = 175, 32 months median follow-up). Using a Deep learning cell classifier, trained upon >38,000 pathologist annotations, to detect eight cell types within H&E-stained sections of CRC, we quantified the spatial tissue organisation and colocalisation of cell types across these cohorts. We found that the ratio of infiltrating endothelial cells to cancer cells, a possible marker of vascular invasion, was an independent predictor of progression-free survival (PFS) in the BRAF+MISSONI cohort (p = 0.033, HR = 1.44, CI = 1.029-2.01). In the VALENTINO cohort, this pattern was also an independent PFS predictor in TP53 mutant patients (p = 0.009, HR = 0.59, CI = 0.40-0.88). Tumour-infiltrating lymphocytes were an independent predictor of PFS in BRAF+MISSONI (p = 0.016, HR = 0.36, CI = 0.153-0.83). Elevated tumour-infiltrating macrophages were predictive of improved PFS in the MISSONI cohort (p = 0.031). We validated our cell classification using highly multiplexed immunofluorescence for 17 markers applied to the same sections that were analysed by the classifier (n = 26 cases). These findings uncovered important microenvironmental factors that underpin treatment response across and within CRC molecular subtypes, while providing an atlas of the distribution of 180 million cells in 375 clinically annotated CRC patients. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Nicholas Trahearn
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
- UCL Cancer InstituteUCLLondon, UK
| | - Chirine Sakr
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
| | | | - Seung Hyun Lee
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
- Systems Oncology group, Cancer Research UK Manchester InstituteThe University of ManchesterManchesterUK
| | - Ann‐Marie Baker
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Hemant M Kocher
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | | | | | | | - Giulia Maddalena
- Veneto Institute of Oncology IOV‐IRCCSPaduaItaly
- Department of Surgical, Oncological and Gastroenterological SciencesUniversity of PaduaPaduaItaly
| | | | | | | | - Trevor Graham
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | | | - Sara Lonardi
- Veneto Institute of Oncology IOV‐IRCCSPaduaItaly
| | - Matteo Fassan
- Department of Medicine (DIMED)University of PaduaPaduaItaly
- Veneto Institute of Oncology IOV‐IRCCSPaduaItaly
| | - Andrea Sottoriva
- Centre for Evolution and CancerThe Institute of Cancer ResearchLondonUK
- Computational Biology Research CentreHuman TechnopoleMilanItaly
| |
Collapse
|
11
|
Fountzilas E, Pearce T, Baysal MA, Chakraborty A, Tsimberidou AM. Convergence of evolving artificial intelligence and machine learning techniques in precision oncology. NPJ Digit Med 2025; 8:75. [PMID: 39890986 PMCID: PMC11785769 DOI: 10.1038/s41746-025-01471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025] Open
Abstract
The confluence of new technologies with artificial intelligence (AI) and machine learning (ML) analytical techniques is rapidly advancing the field of precision oncology, promising to improve diagnostic approaches and therapeutic strategies for patients with cancer. By analyzing multi-dimensional, multiomic, spatial pathology, and radiomic data, these technologies enable a deeper understanding of the intricate molecular pathways, aiding in the identification of critical nodes within the tumor's biology to optimize treatment selection. The applications of AI/ML in precision oncology are extensive and include the generation of synthetic data, e.g., digital twins, in order to provide the necessary information to design or expedite the conduct of clinical trials. Currently, many operational and technical challenges exist related to data technology, engineering, and storage; algorithm development and structures; quality and quantity of the data and the analytical pipeline; data sharing and generalizability; and the incorporation of these technologies into the current clinical workflow and reimbursement models.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Panorama, Thessaloniki, Greece
| | | | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA.
| |
Collapse
|
12
|
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
|
13
|
Wang CW, Liu TC, Lai PJ, Muzakky H, Wang YC, Yu MH, Wu CH, Chao TK. Ensemble transformer-based multiple instance learning to predict pathological subtypes and tumor mutational burden from histopathological whole slide images of endometrial and colorectal cancer. Med Image Anal 2025; 99:103372. [PMID: 39461079 DOI: 10.1016/j.media.2024.103372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/30/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
In endometrial cancer (EC) and colorectal cancer (CRC), in addition to microsatellite instability, tumor mutational burden (TMB) has gradually gained attention as a genomic biomarker that can be used clinically to determine which patients may benefit from immune checkpoint inhibitors. High TMB is characterized by a large number of mutated genes, which encode aberrant tumor neoantigens, and implies a better response to immunotherapy. Hence, a part of EC and CRC patients associated with high TMB may have higher chances to receive immunotherapy. TMB measurement was mainly evaluated by whole-exome sequencing or next-generation sequencing, which was costly and difficult to be widely applied in all clinical cases. Therefore, an effective, efficient, low-cost and easily accessible tool is urgently needed to distinguish the TMB status of EC and CRC patients. In this study, we present a deep learning framework, namely Ensemble Transformer-based Multiple Instance Learning with Self-Supervised Learning Vision Transformer feature encoder (ETMIL-SSLViT), to predict pathological subtype and TMB status directly from the H&E stained whole slide images (WSIs) in EC and CRC patients, which is helpful for both pathological classification and cancer treatment planning. Our framework was evaluated on two different cancer cohorts, including an EC cohort with 918 histopathology WSIs from 529 patients and a CRC cohort with 1495 WSIs from 594 patients from The Cancer Genome Atlas. The experimental results show that the proposed methods achieved excellent performance and outperforming seven state-of-the-art (SOTA) methods in cancer subtype classification and TMB prediction on both cancer datasets. Fisher's exact test further validated that the associations between the predictions of the proposed models and the actual cancer subtype or TMB status are both extremely strong (p<0.001). These promising findings show the potential of our proposed methods to guide personalized treatment decisions by accurately predicting the EC and CRC subtype and the TMB status for effective immunotherapy planning for EC and CRC patients.
Collapse
Affiliation(s)
- Ching-Wei Wang
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Tzu-Chien Liu
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Po-Jen Lai
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Hikam Muzakky
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Yu-Chi Wang
- Department of Gynecology and Obstetrics, Tri-Service General Hospital, Taipei, 114202, Taiwan; Department of Gynecology and Obstetrics, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Mu-Hsien Yu
- Department of Gynecology and Obstetrics, Tri-Service General Hospital, Taipei, 114202, Taiwan; Department of Gynecology and Obstetrics, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chia-Hua Wu
- Department of Pathology, Tri-Service General Hospital, Taipei, 114202, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, Taipei, 114202, Taiwan; Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
14
|
Syrykh C, van den Brand M, Kather JN, Laurent C. Role of artificial intelligence in haematolymphoid diagnostics. Histopathology 2025; 86:58-68. [PMID: 39435690 DOI: 10.1111/his.15327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The advent of digital pathology and the deployment of high-throughput molecular techniques are generating an unprecedented mass of data. Thanks to advances in computational sciences, artificial intelligence (AI) approaches represent a promising avenue for extracting relevant information from complex data structures. From diagnostic assistance to powerful research tools, the potential fields of application of machine learning techniques in pathology are vast and constitute the subject of considerable research work. The aim of this article is to provide an overview of the potential applications of AI in the field of haematopathology and to define the role that these emerging technologies could play in our laboratories in the short to medium term.
Collapse
Affiliation(s)
- Charlotte Syrykh
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
| | - Michiel van den Brand
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Pathology-DNA, Arnhem, The Netherlands
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Camille Laurent
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse CHU Toulouse, Toulouse, France
- INSERM UMR1037, CNRS UMR5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| |
Collapse
|
15
|
Zhang K, Yang X, Wang Y, Yu Y, Huang N, Li G, Li X, Wu JC, Yang S. Artificial intelligence in drug development. Nat Med 2025; 31:45-59. [PMID: 39833407 DOI: 10.1038/s41591-024-03434-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025]
Abstract
Drug development is a complex and time-consuming endeavor that traditionally relies on the experience of drug developers and trial-and-error experimentation. The advent of artificial intelligence (AI) technologies, particularly emerging large language models and generative AI, is poised to redefine this paradigm. The integration of AI-driven methodologies into the drug development pipeline has already heralded subtle yet meaningful enhancements in both the efficiency and effectiveness of this process. Here we present an overview of recent advancements in AI applications across the entire drug development workflow, encompassing the identification of disease targets, drug discovery, preclinical and clinical studies, and post-market surveillance. Lastly, we critically examine the prevailing challenges to highlight promising future research directions in AI-augmented drug development.
Collapse
Affiliation(s)
- Kang Zhang
- Eye Hospital and Institute for Advanced Study on Eye Health and Diseases, Institute for clinical Data Science, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, Wenzhou Medical University, Wenzhou, China.
| | - Xin Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yunfang Yu
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute for AI in Medicine and faculty of Medicine, Macau University of Science and Technology, Macau, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Niu Huang
- National Institute of Biological Sciences, Beijing, China
| | - Gen Li
- Eye Hospital and Institute for Advanced Study on Eye Health and Diseases, Institute for clinical Data Science, Wenzhou Medical University, Wenzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Eye and Vision Innovation Center, Eye Valley, Wenzhou, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, Wenzhou Medical University, Wenzhou, China
| | - Joseph C Wu
- Cardiovascular Research Institute, Stanford University, Stanford, CA, USA
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Tafenzi HA, Essaadi I, Belbaraka R. Digital Oncology in Morocco: Embracing Artificial Intelligence in a New Era. JCO Glob Oncol 2025; 11:e2400583. [PMID: 39787448 DOI: 10.1200/go-24-00583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Affiliation(s)
- Hassan Abdelilah Tafenzi
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | - Ismail Essaadi
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
- Medical Oncology Department, Avicenna Military Hospital of Marrakech, Marrakech, Morocco
| | - Rhizlane Belbaraka
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
17
|
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
|
18
|
Zhang Y, Huang Y, Xu M, Zhuang J, Zhou Z, Zheng S, Zhu B, Guan G, Chen H, Liu X. Pathomics-based machine learning models for predicting pathological complete response and prognosis in locally advanced rectal cancer patients post-neoadjuvant chemoradiotherapy: insights from two independent institutional studies. BMC Cancer 2024; 24:1580. [PMID: 39725903 DOI: 10.1186/s12885-024-13328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Accurate prediction of pathological complete response (pCR) and disease-free survival (DFS) in locally advanced rectal cancer (LARC) patients undergoing neoadjuvant chemoradiotherapy (NCRT) is essential for formulating effective treatment plans. This study aimed to construct and validate the machine learning (ML) models to predict pCR and DFS using pathomics. METHOD A retrospective analysis was conducted on 294 patients who received NCRT from two independent institutions. Pathomics from pre-NCRT H&E stains were extracted, and five ML models were developed and validated across two centers using ROC, Kaplan-Meier, time-dependent ROC, and nomogram analyses. RESULT Among the five ML models, the Xgboost (XGB) model demonstrated superior performance in predicting pCR, achieving an AUC of 1.000 (p < 0.001) on the internal data-set and an AUC of 0.950 (p = 0.001) on the external data-set.The XGB model effectively differentiated between high-risk and low-risk prognosis patients across all five centers: internal dataset (DFS, p = 0.002; OS, p = 0.004) and external dataset (DFS, p = 0.074; OS, p = 0.224).Furthermore, the COX regression demonstrated that the tumor length (HR = 1.230, 95%CI: 1.050-1.440, p = 0.010), post-NCRT CEA (HR = 1.716, 95%CI: 1.031- 2.858, p = 0.038), and XGB model score (HR = 0.128, 95%CI: 0.026-0.636, p = 0.012) were independent predictors of DFS after NCRT in the internal data-set.Using COX regression, the nomogram model and time-dependent AUC analysis demonstrated strong predictive discrimination for DFS in LARC patients across two independent institutions. CONCLUSION The ML model based on pathomics demonstrated effective prediction of pCR and prognosis in LARC patients. Further validation in larger cohorts is warranted to confirm the findings of this study.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meifang Xu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiazheng Zhuang
- Department of Gastrointestinal Surgery, The Quanzhou First Hospital Affiliated of Fujian Medical University, Quanzhou, China
| | - Zhibo Zhou
- Fujian Medical University, Fuzhou, China
| | | | - Bingwang Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Hong Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| |
Collapse
|
19
|
Crasta KC, Faggioli F. Editorial for Special Issue "Cellular Senescence: Recent Cellular Advances and Discoveries". Biomedicines 2024; 12:2796. [PMID: 39767702 PMCID: PMC11673480 DOI: 10.3390/biomedicines12122796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cellular senescence has emerged as a fascinating frontier in biological research and now presents profound implications across diverse fields, from aging research to cancer therapy [...].
Collapse
Affiliation(s)
- Karen Carmelina Crasta
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore 117456, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Francesca Faggioli
- IRCCS Humanitas Research Hospital, Rozzano, 220089 Milan, Italy
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, 20132 Milan, Italy
| |
Collapse
|
20
|
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
|
21
|
Rai HM, Yoo J, Razaque A. Comparative analysis of machine learning and deep learning models for improved cancer detection: A comprehensive review of recent advancements in diagnostic techniques. EXPERT SYSTEMS WITH APPLICATIONS 2024; 255:124838. [DOI: 10.1016/j.eswa.2024.124838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
22
|
Kong F, Wang X, Xiang J, Yang S, Wang X, Yue M, Zhang J, Zhao J, Han X, Dong Y, Zhu B, Wang F, Liu Y. Federated attention consistent learning models for prostate cancer diagnosis and Gleason grading. Comput Struct Biotechnol J 2024; 23:1439-1449. [PMID: 38623561 PMCID: PMC11016961 DOI: 10.1016/j.csbj.2024.03.028] [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: 01/14/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/17/2024] Open
Abstract
Artificial intelligence (AI) holds significant promise in transforming medical imaging, enhancing diagnostics, and refining treatment strategies. However, the reliance on extensive multicenter datasets for training AI models poses challenges due to privacy concerns. Federated learning provides a solution by facilitating collaborative model training across multiple centers without sharing raw data. This study introduces a federated attention-consistent learning (FACL) framework to address challenges associated with large-scale pathological images and data heterogeneity. FACL enhances model generalization by maximizing attention consistency between local clients and the server model. To ensure privacy and validate robustness, we incorporated differential privacy by introducing noise during parameter transfer. We assessed the effectiveness of FACL in cancer diagnosis and Gleason grading tasks using 19,461 whole-slide images of prostate cancer from multiple centers. In the diagnosis task, FACL achieved an area under the curve (AUC) of 0.9718, outperforming seven centers with an average AUC of 0.9499 when categories are relatively balanced. For the Gleason grading task, FACL attained a Kappa score of 0.8463, surpassing the average Kappa score of 0.7379 from six centers. In conclusion, FACL offers a robust, accurate, and cost-effective AI training model for prostate cancer pathology while maintaining effective data safeguards.
Collapse
Affiliation(s)
- Fei Kong
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xiyue Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | | | - Sen Yang
- AI Lab, Tencent, Shenzhen, 518057, China
| | - Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Meng Yue
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Jun Zhang
- AI Lab, Tencent, Shenzhen, 518057, China
| | - Junhan Zhao
- Massachusetts General Hospital, Boston, MA, 02114, United States
- Harvard T.H. Chan School of Public Health, Boston, MA, 02115, United States
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, United States
| | - Xiao Han
- AI Lab, Tencent, Shenzhen, 518057, China
| | - Yuhan Dong
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Biyue Zhu
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| |
Collapse
|
23
|
Lauck KC, Cho SW, DaCunha M, Wuennenberg J, Aasi S, Alam M, Arron ST, Bar A, Brodland DG, Cerci FB, Cohen JL, Coldiron B, Council ML, Harmon CB, Hruza G, Läuchli S, Moody BR, Wysong AS, Zitelli JA, Tolkachjov SN. The utility of artificial intelligence platforms for patient-generated questions in Mohs micrographic surgery: a multi-national, blinded expert panel evaluation. Int J Dermatol 2024; 63:1592-1598. [PMID: 39123288 DOI: 10.1111/ijd.17382] [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: 06/07/2024] [Revised: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Artificial intelligence (AI) and large language models (LLMs) transform how patients inform themselves. LLMs offer potential as educational tools, but their quality depends upon the information generated. Current literature examining AI as an informational tool in dermatology has been limited in evaluating AI's multifaceted roles and diversity of opinions. Here, we evaluate LLMs as a patient-educational tool for Mohs micrographic surgery (MMS) in and out of the clinic utilizing an international expert panel. METHODS The most common patient MMS questions were extracted from Google and transposed into two LLMs and Google's search engine. 15 MMS surgeons evaluated the generated responses, examining their appropriateness as a patient-facing informational platform, sufficiency of response in a clinical environment, and accuracy of content generated. Validated scales were employed to assess the comprehensibility of each response. RESULTS The majority of reviewers deemed all LLM responses appropriate. 75% of responses were rated as mostly accurate or higher. ChatGPT had the highest mean accuracy. The majority of the panel deemed 33% of responses sufficient for clinical practice. The mean comprehensibility scores for all platforms indicated a required 10th-grade reading level. CONCLUSIONS LLM-generated responses were rated as appropriate patient informational sources and mostly accurate in their content. However, these platforms may not provide sufficient information to function in a clinical environment, and complex comprehensibility may represent a barrier to utilization. As the popularity of these platforms increases, it is important for dermatologists to be aware of these limitations.
Collapse
Affiliation(s)
- Kyle C Lauck
- Baylor University Medical Center, Dallas, TX, USA
| | - Seo Won Cho
- Texas A&M College of Medicine, Dallas, TX, USA
| | - Matthew DaCunha
- The Skin Cancer Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - Sumaira Aasi
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | - Murad Alam
- Department of Dermatology, Feinberg School of Medicine, Chicago, IL, USA
| | | | - Anna Bar
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - David G Brodland
- Zitelli & Brodland Skin Cancer Center, University of Pittsburgh Medical Center Shadyside Hospital, Pittsburgh, PA, USA
| | - Felipe B Cerci
- Division of Dermatology, Hospital Universitário Evangélico Mackenzie, Curitiba, Brazil
| | - Joel L Cohen
- AboutSkin Dermatology and DermSurgery, Irvine, CO, USA
| | - Brett Coldiron
- The Skin Cancer Center, University of Cincinnati, Cincinnati, OH, USA
| | - M Laurin Council
- Division of Dermatology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | | | - George Hruza
- Department of Dermatology, Saint-Louis University, Saint-Louis, MO, USA
| | | | | | - Ashley S Wysong
- Department of Dermatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - John A Zitelli
- Zitelli & Brodland Skin Cancer Center, University of Pittsburgh Medical Center Shadyside Hospital, Pittsburgh, PA, USA
| | - Stanislav N Tolkachjov
- Baylor University Medical Center, Dallas, TX, USA
- Texas A&M College of Medicine, Dallas, TX, USA
- Epiphany Dermatology, Dallas, TX, USA
- Department of Dermatology, University of Texas at Southwestern, Dallas, TX, USA
| |
Collapse
|
24
|
D’Orsi L, Capasso B, Lamacchia G, Pizzichini P, Ferranti S, Liverani A, Fontana C, Panunzi S, De Gaetano A, Lo Presti E. Recent Advances in Artificial Intelligence to Improve Immunotherapy and the Use of Digital Twins to Identify Prognosis of Patients with Solid Tumors. Int J Mol Sci 2024; 25:11588. [PMID: 39519142 PMCID: PMC11546512 DOI: 10.3390/ijms252111588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
To date, the public health system has been impacted by the increasing costs of many diagnostic and therapeutic pathways due to limited resources. At the same time, we are constantly seeking to improve these paths through approaches aimed at personalized medicine. To achieve the required levels of diagnostic and therapeutic precision, it is necessary to integrate data from different sources and simulation platforms. Today, artificial intelligence (AI), machine learning (ML), and predictive computer models are more efficient at guiding decisions regarding better therapies and medical procedures. The evolution of these multiparametric and multimodal systems has led to the creation of digital twins (DTs). The goal of our review is to summarize AI applications in discovering new immunotherapies and developing predictive models for more precise immunotherapeutic decision-making. The findings from this literature review highlight that DTs, particularly predictive mathematical models, will be pivotal in advancing healthcare outcomes. Over time, DTs will indeed bring the benefits of diagnostic precision and personalized treatment to a broader spectrum of patients.
Collapse
Affiliation(s)
- Laura D’Orsi
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
| | - Biagio Capasso
- Department of General Surgery, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (B.C.); (S.F.)
| | - Giuseppe Lamacchia
- General Surgery Unit, Regina Apostolorum Hospital, Via S. Francesco d’Assisi, 50, 00041 Albano Laziale, RM, Italy; (G.L.); (A.L.)
| | - Paolo Pizzichini
- Department of Intensive Care Unit, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (P.P.); (C.F.)
| | - Sergio Ferranti
- Department of General Surgery, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (B.C.); (S.F.)
| | - Andrea Liverani
- General Surgery Unit, Regina Apostolorum Hospital, Via S. Francesco d’Assisi, 50, 00041 Albano Laziale, RM, Italy; (G.L.); (A.L.)
| | - Costantino Fontana
- Department of Intensive Care Unit, Policlinico Militare di Roma “Celio”, Piazza Celimontana, 50, 00184 Rome, RM, Italy; (P.P.); (C.F.)
| | - Simona Panunzi
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
| | - Andrea De Gaetano
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti”, BioMatLab, Via dei Taurini, 19, 00185 Rome, RM, Italy; (L.D.); (S.P.); (A.D.G.)
- National Research Council of Italy, Institute for Biomedical Research and Innovation (CNR-IRIB), Via Ugo La Malfa, 153, 90146 Palermo, PA, Italy
- Department of Biomatics, Óbuda University, Bécsi Road 96/B, H-1034 Budapest, Hungary
| | - Elena Lo Presti
- National Research Council of Italy, Institute for Biomedical Research and Innovation (CNR-IRIB), Via Ugo La Malfa, 153, 90146 Palermo, PA, Italy
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Sun L, Zhang R, Gu Y, Huang L, Jin C. Application of Artificial Intelligence in the diagnosis and treatment of colorectal cancer: a bibliometric analysis, 2004-2023. Front Oncol 2024; 14:1424044. [PMID: 39464716 PMCID: PMC11502294 DOI: 10.3389/fonc.2024.1424044] [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: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND An increasing number of studies have turned their lens to the application of Artificial Intelligence (AI) in the diagnosis and treatment of colorectal cancer (CRC). OBJECTIVE To clarify and visualize the basic situation, research hotspots, and development trends of AI in the diagnosis and treatment of CRC, and provide clues for research in the future. METHODS On January 31, 2024, the Web of Science Core Collection (WoSCC) database was searched to screen and export the relevant research published during 2004-2023, and Cite Space, VoSviewer, Bibliometrix were used to visualize the number of publications, countries (regions), institutions, journals, authors, citations, keywords, etc. RESULTS A total of 2715 pieces of literature were included. The number of publications grew slowly until the end of 2016, but rapidly after 2017, till to the peak of 798 in 2023. A total of 92 countries, 3997 organizations, and 15,667 authors were involved in this research. Chinese scholars released the highest number of publications, and the U.S. contributed the highest number of total citations. As to authors, MORI, YUICHI had the highest number of publications, and WANG, PU had the highest number of total citations. According to the analysis of citations and keywords, the current research hotspots are mainly related to "Colonoscopy", "Polyp Segmentation", "Digital Pathology", "Radiomics", "prognosis". CONCLUSION Research on the application of AI in the diagnosis and treatment of CRC has made significant progress and is flourishing across the world. Current research hotspots include AI-assisted early screening and diagnosis, pathology, and staging, and prognosis assessment, and future research is predicted to put weight on multimodal data fusion, personalized treatment, and drug development.
Collapse
Affiliation(s)
- Lamei Sun
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- Department of Traditional Chinese Medicine, Jiangyin Nanzha Community Health Service Center, Wuxi, China
| | - Rong Zhang
- Department of General Surgery, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yidan Gu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Lei Huang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Chunhui Jin
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
28
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
29
|
Ancheta K, Le Calvez S, Williams J. The digital revolution in veterinary pathology. J Comp Pathol 2024; 214:19-31. [PMID: 39241697 DOI: 10.1016/j.jcpa.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/14/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024]
Abstract
For the past two centuries, the use of traditional light microscopy to examine tissues to make diagnoses has remained relatively unchanged. While the fundamental concept of tissue slide analysis has stayed the same, our interaction with the microscope is undergoing significant changes. Digital pathology (DP) has gained momentum in veterinary science and is on the verge of becoming a vital tool in diagnostics, research and education. Many diagnostic laboratories have incorporated DP as a critical part of their workflows. Innovations in DP and whole slide image technology have made telediagnosis (the process of transmitting digital clinical data using telecommunication networks for distant diagnosis) more accessible, leading to improved patient care through streamlining of workflows and greater accessibility of second opinions. The integration of machine learning and artificial intelligence and human-in-the-loop protocols for DP workflows will further the development of computer-aided diagnosis and prognostic tools. Despite its present weaknesses, DP will progressively aid veterinary clinicians and pathologists in delivering more accurate and reliable diagnoses. Consistent incorporation of DP frontline advancements into routine veterinary diagnostic pipelines will assist in improving current tools and help prepare pathologists for the progression of digitalization in the field.
Collapse
Affiliation(s)
- Kenneth Ancheta
- The Royal Veterinary College, Hawkshead Campus, Hawkshead Lane, Hatfield, Hertfordshire AL9 7TA, UK
| | - Sophie Le Calvez
- IDEXX Laboratories Ltd, Grange House, Sandbeck Way, Wetherby, Yorkshire LS22 7DN, UK
| | - Jonathan Williams
- The Royal Veterinary College, Hawkshead Campus, Hawkshead Lane, Hatfield, Hertfordshire AL9 7TA, UK.
| |
Collapse
|
30
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
31
|
Nowak M, Jabbar F, Rodewald AK, Gneo L, Tomasevic T, Harkin A, Iveson T, Saunders M, Kerr R, Oein K, Maka N, Hay J, Edwards J, Tomlinson I, Sansom O, Kelly C, Pezzella F, Kerr D, Easton A, Domingo E, Koelzer VH, Church DN. Single-cell AI-based detection and prognostic and predictive value of DNA mismatch repair deficiency in colorectal cancer. Cell Rep Med 2024; 5:101727. [PMID: 39293403 PMCID: PMC11525017 DOI: 10.1016/j.xcrm.2024.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 09/20/2024]
Abstract
Testing for DNA mismatch repair deficiency (MMRd) is recommended for all colorectal cancers (CRCs). Automating this would enable precision medicine, particularly if providing information on etiology not captured by deep learning (DL) methods. We present AIMMeR, an AI-based method for determination of mismatch repair (MMR) protein expression at a single-cell level in routine pathology samples. AIMMeR shows an area under the receiver-operator curve (AUROC) of 0.98, and specificity of ≥75% at 98% sensitivity against pathologist ground truth in stage II/III in two trial cohorts, with positive predictive value of ≥98% for the commonest pattern of somatic MMRd. Lower agreement with microsatellite instability (MSI) testing (AUROC 0.86) reflects discordance between MMR and MSI PCR rather than AIMMeR misclassification. Analysis of the SCOT trial confirms MMRd prognostic value in oxaliplatin-treated patients; while MMRd does not predict differential benefit of chemotherapy duration, it correlates with difference in relapse by regimen (PInteraction = 0.04). AIMMeR may help reduce pathologist workload and streamline diagnostics in CRC.
Collapse
Affiliation(s)
- Marta Nowak
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland
| | - Faiz Jabbar
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ann-Katrin Rodewald
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland
| | - Luciana Gneo
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Tijana Tomasevic
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrea Harkin
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - Tim Iveson
- University of Southampton, Southampton, UK
| | | | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - Karin Oein
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Noori Maka
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Jennifer Hay
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, UK
| | - Owen Sansom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, UK
| | - Caroline Kelly
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | | | - David Kerr
- Nuffield Department of Clinical and Laboratory Sciences, University of Oxford, Oxford, UK
| | | | - Enric Domingo
- Department of Oncology, University of Oxford, Oxford, UK
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland; Department of Oncology, University of Oxford, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - David N Church
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
32
|
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
|
33
|
Jia P, Yang X, Yang X, Wang T, Xu Y, Ye K. MSIsensor-RNA: Microsatellite Instability Detection for Bulk and Single-cell Gene Expression Data. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae004. [PMID: 39341794 PMCID: PMC12016039 DOI: 10.1093/gpbjnl/qzae004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 10/01/2024]
Abstract
Microsatellite instability (MSI) is an indispensable biomarker in cancer immunotherapy. Currently, MSI scoring methods by high-throughput omics methods have gained popularity and demonstrated better performance than the gold standard method for MSI detection. However, the MSI detection method on expression data, especially single-cell expression data, is still lacking, limiting the scope of clinical application and prohibiting the investigation of MSI at a single-cell level. Herein, we developed MSIsensor-RNA, an accurate, robust, adaptable, and standalone software to detect MSI status based on expression values of MSI-associated genes. We demonstrated the favorable performance and promise of MSIsensor-RNA in both bulk and single-cell gene expression data in multiplatform technologies including RNA sequencing (RNA-seq), microarray, and single-cell RNA-seq. MSIsensor-RNA is a versatile, efficient, and robust method for MSI status detection from both bulk and single-cell gene expression data in clinical studies and applications. MSIsensor-RNA is available at https://github.com/xjtu-omics/msisensor-rna.
Collapse
Affiliation(s)
- Peng Jia
- Department of Gynecology and Obstetrics, Center for Mathematical Medical, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xuanhao Yang
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xiaofei Yang
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Computer Science and Technology, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Tingjie Wang
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- Genome Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yu Xu
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Kai Ye
- Department of Gynecology and Obstetrics, Center for Mathematical Medical, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- Genome Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
34
|
Hezi H, Shats D, Gurevich D, Maruvka YE, Freiman M. Exploring the interplay between colorectal cancer subtypes genomic variants and cellular morphology: A deep-learning approach. PLoS One 2024; 19:e0309380. [PMID: 39255280 PMCID: PMC11386451 DOI: 10.1371/journal.pone.0309380] [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: 02/27/2024] [Accepted: 08/10/2024] [Indexed: 09/12/2024] Open
Abstract
Molecular subtypes of colorectal cancer (CRC) significantly influence treatment decisions. While convolutional neural networks (CNNs) have recently been introduced for automated CRC subtype identification using H&E stained histopathological images, the correlation between CRC subtype genomic variants and their corresponding cellular morphology expressed by their imaging phenotypes is yet to be fully explored. The goal of this study was to determine such correlations by incorporating genomic variants in CNN models for CRC subtype classification from H&E images. We utilized the publicly available TCGA-CRC-DX dataset, which comprises whole slide images from 360 CRC-diagnosed patients (260 for training and 100 for testing). This dataset also provides information on CRC subtype classifications and genomic variations. We trained CNN models for CRC subtype classification that account for potential correlation between genomic variations within CRC subtypes and their corresponding cellular morphology patterns. We assessed the interplay between CRC subtypes' genomic variations and cellular morphology patterns by evaluating the CRC subtype classification accuracy of the different models in a stratified 5-fold cross-validation experimental setup using the area under the ROC curve (AUROC) and average precision (AP) as the performance metrics. The CNN models that account for potential correlation between genomic variations within CRC subtypes and their cellular morphology pattern achieved superior accuracy compared to the baseline CNN classification model that does not account for genomic variations when using either single-nucleotide-polymorphism (SNP) molecular features (AUROC: 0.824±0.02 vs. 0.761±0.04, p<0.05, AP: 0.652±0.06 vs. 0.58±0.08) or CpG-Island methylation phenotype (CIMP) molecular features (AUROC: 0.834±0.01 vs. 0.787±0.03, p<0.05, AP: 0.687±0.02 vs. 0.64±0.05). Combining the CNN models account for variations in CIMP and SNP further improved classification accuracy (AUROC: 0.847±0.01 vs. 0.787±0.03, p = 0.01, AP: 0.68±0.02 vs. 0.64±0.05). The improved accuracy of CNN models for CRC subtype classification that account for potential correlation between genomic variations within CRC subtypes and their corresponding cellular morphology as expressed by H&E imaging phenotypes may elucidate the biological cues impacting cancer histopathological imaging phenotypes. Moreover, considering CRC subtypes genomic variations has the potential to improve the accuracy of deep-learning models in discerning cancer subtype from histopathological imaging data.
Collapse
Affiliation(s)
- Hadar Hezi
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daniel Shats
- Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daniel Gurevich
- Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- Lokey Center for Life Science and Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yosef E Maruvka
- Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
- Lokey Center for Life Science and Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Moti Freiman
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
35
|
Li Y, Yang F, Liu K. Application of digital pathology in liver transplantation. J Hepatol 2024; 81:e112-e113. [PMID: 38527523 DOI: 10.1016/j.jhep.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Affiliation(s)
- Yang Li
- Comprehensive pediatrics & Pulmonary and Critical Care Medicine, Kunming Children's Hospital, No.28, Shulin Street, Kunming 650103, Yunnan Province, China
| | - FengQi Yang
- Comprehensive pediatrics & Pulmonary and Critical Care Medicine, Kunming Children's Hospital, No.28, Shulin Street, Kunming 650103, Yunnan Province, China
| | - Kai Liu
- Comprehensive pediatrics & Pulmonary and Critical Care Medicine, Kunming Children's Hospital, No.28, Shulin Street, Kunming 650103, Yunnan Province, China.
| |
Collapse
|
36
|
Mubarak M, Rashid R, Sapna F, Shakeel S. Expanding role and scope of artificial intelligence in the field of gastrointestinal pathology. Artif Intell Gastroenterol 2024; 5:91550. [DOI: 10.35712/aig.v5.i2.91550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/06/2024] [Accepted: 07/29/2024] [Indexed: 08/08/2024] Open
Abstract
Digital pathology (DP) and its subsidiaries including artificial intelligence (AI) are rapidly making inroads into the area of diagnostic anatomic pathology (AP) including gastrointestinal (GI) pathology. It is poised to revolutionize the field of diagnostic AP. Historically, AP has been slow to adopt digital technology, but this is changing rapidly, with many centers worldwide transitioning to DP. Coupled with advanced techniques of AI such as deep learning and machine learning, DP is likely to transform histopathology from a subjective field to an objective, efficient, and transparent discipline. AI is increasingly integrated into GI pathology, offering numerous advancements and improvements in overall diagnostic accuracy, efficiency, and patient care. Specifically, AI in GI pathology enhances diagnostic accuracy, streamlines workflows, provides predictive insights, integrates multimodal data, supports research, and aids in education and training, ultimately improving patient care and outcomes. This review summarized the latest developments in the role and scope of AI in AP with a focus on GI pathology. The main aim was to provide updates and create awareness among the pathology community.
Collapse
Affiliation(s)
- Muhammed Mubarak
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Rahma Rashid
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Fnu Sapna
- Department of Pathology, Montefiore Medical Center, The University Hospital for Albert Einstein School of Medicine, Bronx, NY 10461, United States
| | - Shaheera Shakeel
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| |
Collapse
|
37
|
Whangbo J, Lee YS, Kim YJ, Kim J, Kim KG. Predicting Mismatch Repair Deficiency Status in Endometrial Cancer through Multi-Resolution Ensemble Learning in Digital Pathology. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1674-1682. [PMID: 38378964 PMCID: PMC11300772 DOI: 10.1007/s10278-024-00997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024]
Abstract
For molecular classification of endometrial carcinoma, testing for mismatch repair (MMR) status is becoming a routine process. Mismatch repair deficiency (MMR-D) is caused by loss of expression in one or more of the 4 major MMR proteins: MLH1, MSH2, MSH6, PHS2. Over 30% of patients with endometrial cancer have MMR-D. Determining the MMR status holds significance as individuals with MMR-D are potential candidates for immunotherapy. Pathological whole slide image (WSI) of endometrial cancer with immunohistochemistry results of MMR proteins were gathered. Color normalization was applied to the tiles using a CycleGAN-based network. The WSI was divided into tiles at three different magnifications (2.5 × , 5 × , and 10 ×). Three distinct networks of the same architecture were employed to include features from all three magnification levels and were stacked for ensemble learning. Three architectures, InceptionResNetV2, EfficientNetB2, and EfficientNetB3 were employed and subjected to comparison. The per-tile results were gathered to classify MMR status in the WSI, and prediction accuracy was evaluated using the following performance metrics: AUC, accuracy, sensitivity, and specificity. The EfficientNetB2 was able to make predictions with an AUC of 0.821, highest among the three architectures, and an overall AUC range of 0.767 - 0.821 was reported across the three architectures. In summary, our study successfully predicted MMR classification from pathological WSIs in endometrial cancer through a multi-resolution ensemble learning approach, which holds the potential to facilitate swift decisions on tailored treatment, such as immunotherapy, in clinical settings.
Collapse
Affiliation(s)
- Jongwook Whangbo
- Department of Computer Science, Wesleyan University, Middletown, Connecticut, USA
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Young Seop Lee
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Young Jae Kim
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
| | - Jisup Kim
- Department of Pathology, Gil Medical Center, Gachon University College of Medicine, 38-13, Dokjeom-Ro 3Beon-Gil, Namdong-Gu, Incheon, Republic of Korea.
| | - Kwang Gi Kim
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea.
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea.
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
38
|
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
|
39
|
Nap R, Aburidi M, Marcia R. Contrastive Pre-Training and Multiple Instance Learning for Predicting Tumor Microsatellite Instability. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-7. [PMID: 40039070 DOI: 10.1109/embc53108.2024.10782037] [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
Accurate classification between tumor MicroSatellite Stability (MSS) and Instability (MSI) is crucial in gastrointestinal (GI) cancer prognosis and treatment. In this paper, we present a novel two-stage weakly supervised methodology, leveraging the synergy of Multiple Instance Learning (MIL) and a unique Contrastive Clustering Network (CCNet), aimed at enhancing MSI prediction in Whole Slide Image (WSI) analysis of GI cancers. In our framework, we utilize a contrastive learning-based feature extractor, coupled with MIL's efficient labeling. Our approach shows notable improvement in MSI classification, outperforming existing methods. Experiments using colorectal cancer and stomach adenocarcinoma datasets demonstrate the model's efficacy and generalizability, marking an advance in computational pathology and cancer diagnostics. Furthermore, we explored the efficacy of transfer learning using our model, examining the performance of pretrained feature extractors from ImageNet and STAD datasets. Our framework outperforms existing methods when pretrained on STAD and transferred to CRC data.Clinical relevance- The potential to enhance the accuracy of gastrointestinal cancer diagnosis and prognosis through advanced machine learning techniques. By leveraging transfer learning and weakly supervised frameworks clinicians can benefit from improved MSI prediction in histopathological images, aiding in personalized treatment strategies and patient outcomes.
Collapse
|
40
|
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
|
41
|
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: 0] [Impact Index Per Article: 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
|
42
|
Ou Q, Lu Z, Cai G, Lai Z, Lin R, Huang H, Zeng D, Wang Z, Luo B, Ouyang W, Liao W. Unraveling the influence of metabolic signatures on immune dynamics for predicting immunotherapy response and survival in cancer. MEDCOMM – FUTURE MEDICINE 2024; 3. [DOI: 10.1002/mef2.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/28/2024] [Indexed: 10/31/2024]
Abstract
AbstractMetabolic reprogramming in cancer significantly impacts immune responses within the tumor microenvironment, but its influence on cancer immunotherapy effectiveness remains uncertain. This study aims to elucidate the prognostic significance of metabolic genes in cancer immunotherapy through a comprehensive analytical approach. Utilizing data from the IMvigor210 trial (n = 348) and validated by retrospective datasets, we performed patient clustering using non‐negative matrix factorization based on metabolism‐related genes. A metabiotic score was developed using a “DeepSurv” neural network to assess correlations with overall survival (OS), progression‐free survival, and immunotherapy response. Validation of the metabolic score and key genes was achieved via comparative gene expression analysis using qPCR. Our analysis identified four distinct metabolic classes with significant variations in OS. Notably, the metabolism‐inactive and hypoxia‐low class demonstrated the most pronounced benefit in terms of OS. The metabolic score predicted immunotherapeutic benefits with high accuracy (AUC: 0.93 at 12 months). SETD3 emerged as a crucial gene, showing strong correlations with improved OS outcomes. This study underscores the importance of metabolic profiling in predicting cancer immunotherapy success. Specifically, patients classified as metabolism‐inactive and hypoxia‐low appear to derive substantial benefits. SETD3 is established as a promising prognostic marker, linking metabolic activity with patient outcomes, advocating for the integration of metabolic profiling into immunotherapy strategies to enhance treatment precision and efficacy.
Collapse
Affiliation(s)
- Qiyun Ou
- Department of Oncology Nanfang Hospital, Southern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Zhiqiang Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Gengyi Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Zijia Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Ruicong Lin
- Faculty of Innovation Engineering Macau University of Science and Technology Taipa China
- School of Computer and Information Engineering Guangzhou Huali College Guangzhou China
| | - Hong Huang
- Clinical Medicine College Guilin Medical University Guilin China
| | - Dongqiang Zeng
- Department of Oncology Nanfang Hospital, Southern Medical University Guangzhou China
| | - Zehua Wang
- Faculty of Medicine Macau University of Science and Technology Taipa China
| | - Baoming Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound in Medicine, Department of Medicine Oncology, Department of Pulmonary and Critical Care Medicine, Sun Yat‐sen Memorial Hospital Sun Yat‐sen University Guangzhou China
| | - Wangjun Liao
- Department of Oncology Nanfang Hospital, Southern Medical University Guangzhou China
| |
Collapse
|
43
|
Hilgers L, Ghaffari Laleh N, West NP, Westwood A, Hewitt KJ, Quirke P, Grabsch HI, Carrero ZI, Matthaei E, Loeffler CML, Brinker TJ, Yuan T, Brenner H, Brobeil A, Hoffmeister M, Kather JN. Automated curation of large-scale cancer histopathology image datasets using deep learning. Histopathology 2024; 84:1139-1153. [PMID: 38409878 DOI: 10.1111/his.15159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/29/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Artificial intelligence (AI) has numerous applications in pathology, supporting diagnosis and prognostication in cancer. However, most AI models are trained on highly selected data, typically one tissue slide per patient. In reality, especially for large surgical resection specimens, dozens of slides can be available for each patient. Manually sorting and labelling whole-slide images (WSIs) is a very time-consuming process, hindering the direct application of AI on the collected tissue samples from large cohorts. In this study we addressed this issue by developing a deep-learning (DL)-based method for automatic curation of large pathology datasets with several slides per patient. METHODS We collected multiple large multicentric datasets of colorectal cancer histopathological slides from the United Kingdom (FOXTROT, N = 21,384 slides; CR07, N = 7985 slides) and Germany (DACHS, N = 3606 slides). These datasets contained multiple types of tissue slides, including bowel resection specimens, endoscopic biopsies, lymph node resections, immunohistochemistry-stained slides, and tissue microarrays. We developed, trained, and tested a deep convolutional neural network model to predict the type of slide from the slide overview (thumbnail) image. The primary statistical endpoint was the macro-averaged area under the receiver operating curve (AUROCs) for detection of the type of slide. RESULTS In the primary dataset (FOXTROT), with an AUROC of 0.995 [95% confidence interval [CI]: 0.994-0.996] the algorithm achieved a high classification performance and was able to accurately predict the type of slide from the thumbnail image alone. In the two external test cohorts (CR07, DACHS) AUROCs of 0.982 [95% CI: 0.979-0.985] and 0.875 [95% CI: 0.864-0.887] were observed, which indicates the generalizability of the trained model on unseen datasets. With a confidence threshold of 0.95, the model reached an accuracy of 94.6% (7331 classified cases) in CR07 and 85.1% (2752 classified cases) for the DACHS cohort. CONCLUSION Our findings show that using the low-resolution thumbnail image is sufficient to accurately classify the type of slide in digital pathology. This can support researchers to make the vast resource of existing pathology archives accessible to modern AI models with only minimal manual annotations.
Collapse
Affiliation(s)
- Lars Hilgers
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Alice Westwood
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Katherine J Hewitt
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Emylou Matthaei
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanwei Yuan
- 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, Heidelberg, Germany
- Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
44
|
Yao L, Li S, Tao Q, Mao Y, Dong J, Lu C, Han C, Qiu B, Huang Y, Huang X, Liang Y, Lin H, Guo Y, Liang Y, Chen Y, Lin J, Chen E, Jia Y, Chen Z, Zheng B, Ling T, Liu S, Tong T, Cao W, Zhang R, Chen X, Liu Z. Deep learning for colorectal cancer detection in contrast-enhanced CT without bowel preparation: a retrospective, multicentre study. EBioMedicine 2024; 104:105183. [PMID: 38848616 PMCID: PMC11192791 DOI: 10.1016/j.ebiom.2024.105183] [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: 10/27/2023] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Contrast-enhanced CT scans provide a means to detect unsuspected colorectal cancer. However, colorectal cancers in contrast-enhanced CT without bowel preparation may elude detection by radiologists. We aimed to develop a deep learning (DL) model for accurate detection of colorectal cancer, and evaluate whether it could improve the detection performance of radiologists. METHODS We developed a DL model using a manually annotated dataset (1196 cancer vs 1034 normal). The DL model was tested using an internal test set (98 vs 115), two external test sets (202 vs 265 in 1, and 252 vs 481 in 2), and a real-world test set (53 vs 1524). We compared the detection performance of the DL model with radiologists, and evaluated its capacity to enhance radiologists' detection performance. FINDINGS In the four test sets, the DL model had the area under the receiver operating characteristic curves (AUCs) ranging between 0.957 and 0.994. In both the internal test set and external test set 1, the DL model yielded higher accuracy than that of radiologists (97.2% vs 86.0%, p < 0.0001; 94.9% vs 85.3%, p < 0.0001), and significantly improved the accuracy of radiologists (93.4% vs 86.0%, p < 0.0001; 93.6% vs 85.3%, p < 0.0001). In the real-world test set, the DL model delivered sensitivity comparable to that of radiologists who had been informed about clinical indications for most cancer cases (94.3% vs 96.2%, p > 0.99), and it detected 2 cases that had been missed by radiologists. INTERPRETATION The developed DL model can accurately detect colorectal cancer and improve radiologists' detection performance, showing its potential as an effective computer-aided detection tool. FUNDING This study was supported by National Science Fund for Distinguished Young Scholars of China (No. 81925023); Regional Innovation and Development Joint Fund of National Natural Science Foundation of China (No. U22A20345); National Natural Science Foundation of China (No. 82072090 and No. 82371954); Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application (No. 2022B1212010011); High-level Hospital Construction Project (No. DFJHBF202105).
Collapse
Affiliation(s)
- Lisha Yao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Suyun Li
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; School of Medicine, South Medical University, Guangzhou, China
| | - Quan Tao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yun Mao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Dong
- Department of Radiology, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), The Third Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Cheng Lu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chu Han
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bingjiang Qiu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Sciences), Guangzhou, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Xin Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; School of Medicine, Shantou University Medical College, Shantou, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China; School of Medicine, South Medical University, Guangzhou, China
| | - Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Yongmei Guo
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yingying Liang
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yizhou Chen
- Department of Radiology, Puning People's Hospital, Southern Medical University, Jieyang, China
| | - Jie Lin
- Department of Radiology, Puning People's Hospital, Southern Medical University, Jieyang, China
| | - Enyan Chen
- Department of Radiology, Puning People's Hospital, Southern Medical University, Jieyang, China
| | - Yanlian Jia
- Department of Radiology, Liaobu Hospital of Guangdong, Dongguan, China
| | - Zhihong Chen
- Institute of Computing Science and Technology, Guangzhou University, Guangzhou, China
| | - Bochi Zheng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Tong Ling
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ruiping Zhang
- Department of Radiology, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), The Third Affiliated Hospital of Shanxi Medical University, Taiyuan, China.
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
45
|
Duwe G, Mercier D, Wiesmann C, Kauth V, Moench K, Junker M, Neumann CCM, Haferkamp A, Dengel A, Höfner T. Challenges and perspectives in use of artificial intelligence to support treatment recommendations in clinical oncology. Cancer Med 2024; 13:e7398. [PMID: 38923826 PMCID: PMC11196383 DOI: 10.1002/cam4.7398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Artificial intelligence (AI) promises to be the next revolutionary step in modern society. Yet, its role in all fields of industry and science need to be determined. One very promising field is represented by AI-based decision-making tools in clinical oncology leading to more comprehensive, personalized therapy approaches. In this review, the authors provide an overview on all relevant technical applications of AI in oncology, which are required to understand the future challenges and realistic perspectives for decision-making tools. In recent years, various applications of AI in medicine have been developed focusing on the analysis of radiological and pathological images. AI applications encompass large amounts of complex data supporting clinical decision-making and reducing errors by objectively quantifying all aspects of the data collected. In clinical oncology, almost all patients receive a treatment recommendation in a multidisciplinary cancer conference at the beginning and during their treatment periods. These highly complex decisions are based on a large amount of information (of the patients and of the various treatment options), which need to be analyzed and correctly classified in a short time. In this review, the authors describe the technical and medical requirements of AI to address these scientific challenges in a multidisciplinary manner. Major challenges in the use of AI in oncology and decision-making tools are data security, data representation, and explainability of AI-based outcome predictions, in particular for decision-making processes in multidisciplinary cancer conferences. Finally, limitations and potential solutions are described and compared for current and future research attempts.
Collapse
Affiliation(s)
- Gregor Duwe
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Dominique Mercier
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Crispin Wiesmann
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Verena Kauth
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Kerstin Moench
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Markus Junker
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Christopher C. M. Neumann
- Department of Hematology, Oncology and Tumor ImmunologyCharité‐Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu BerlinBerlinGermany
| | - Axel Haferkamp
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Andreas Dengel
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Thomas Höfner
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
- Department of Urology, Ordensklinikum Linz ElisabethinenLinzAustria
| |
Collapse
|
46
|
Gustav M, Reitsam NG, Carrero ZI, Loeffler CML, van Treeck M, Yuan T, West NP, Quirke P, Brinker TJ, Brenner H, Favre L, Märkl B, Stenzinger A, Brobeil A, Hoffmeister M, Calderaro J, Pujals A, Kather JN. Deep learning for dual detection of microsatellite instability and POLE mutations in colorectal cancer histopathology. NPJ Precis Oncol 2024; 8:115. [PMID: 38783059 PMCID: PMC11116442 DOI: 10.1038/s41698-024-00592-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/14/2024] [Indexed: 05/25/2024] Open
Abstract
In the spectrum of colorectal tumors, microsatellite-stable (MSS) tumors with DNA polymerase ε (POLE) mutations exhibit a hypermutated profile, holding the potential to respond to immunotherapy similarly to their microsatellite-instable (MSI) counterparts. Yet, due to their rarity and the associated testing costs, systematic screening for these mutations is not commonly pursued. Notably, the histopathological phenotype resulting from POLE mutations is theorized to resemble that of MSI. This resemblance not only could facilitate their detection by a transformer-based Deep Learning (DL) system trained on MSI pathology slides, but also indicates the possibility for MSS patients with POLE mutations to access enhanced treatment options, which might otherwise be overlooked. To harness this potential, we trained a Deep Learning classifier on a large dataset with the ground truth for microsatellite status and subsequently validated its capabilities for MSI and POLE detection across three external cohorts. Our model accurately identified MSI status in both the internal and external resection cohorts using pathology images alone. Notably, with a classification threshold of 0.5, over 75% of POLE driver mutant patients in the external resection cohorts were flagged as "positive" by a DL system trained on MSI status. In a clinical setting, deploying this DL model as a preliminary screening tool could facilitate the efficient identification of clinically relevant MSI and POLE mutations in colorectal tumors, in one go.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Titus J Brinker
- Digital Biomarkers for Oncology, 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
| | - Loëtitia Favre
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Anaïs Pujals
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 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
|
47
|
Schüffler P, Steiger K, Mogler C. [Artificial intelligence for pathology-how, where, and why?]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:198-202. [PMID: 38472382 PMCID: PMC11045628 DOI: 10.1007/s00292-024-01314-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Artificial intelligence promises many innovations and simplifications in pathology, but also raises just as many questions and uncertainties. In this article, we provide a brief overview of the current status, the goals already achieved by existing algorithms, and the remaining challenges.
Collapse
Affiliation(s)
- Peter Schüffler
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland.
- TUM School of Computation, Information and Technology, Technische Universität München, München, Deutschland.
- Munich Center for Machine Learning (MCML), München, Deutschland.
| | - Katja Steiger
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland
| | - Carolin Mogler
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland
| |
Collapse
|
48
|
Lotter W, Hassett MJ, Schultz N, Kehl KL, Van Allen EM, Cerami E. Artificial Intelligence in Oncology: Current Landscape, Challenges, and Future Directions. Cancer Discov 2024; 14:711-726. [PMID: 38597966 PMCID: PMC11131133 DOI: 10.1158/2159-8290.cd-23-1199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/29/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Artificial intelligence (AI) in oncology is advancing beyond algorithm development to integration into clinical practice. This review describes the current state of the field, with a specific focus on clinical integration. AI applications are structured according to cancer type and clinical domain, focusing on the four most common cancers and tasks of detection, diagnosis, and treatment. These applications encompass various data modalities, including imaging, genomics, and medical records. We conclude with a summary of existing challenges, evolving solutions, and potential future directions for the field. SIGNIFICANCE AI is increasingly being applied to all aspects of oncology, where several applications are maturing beyond research and development to direct clinical integration. This review summarizes the current state of the field through the lens of clinical translation along the clinical care continuum. Emerging areas are also highlighted, along with common challenges, evolving solutions, and potential future directions for the field.
Collapse
Affiliation(s)
- William Lotter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael J. Hassett
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth L. Kehl
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M. Van Allen
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
49
|
van Diest PJ, Flach RN, van Dooijeweert C, Makineli S, Breimer GE, Stathonikos N, Pham P, Nguyen TQ, Veta M. Pros and cons of artificial intelligence implementation in diagnostic pathology. Histopathology 2024; 84:924-934. [PMID: 38433288 DOI: 10.1111/his.15153] [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: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/19/2024] [Indexed: 03/05/2024]
Abstract
The rapid introduction of digital pathology has greatly facilitated development of artificial intelligence (AI) models in pathology that have shown great promise in assisting morphological diagnostics and quantitation of therapeutic targets. We are now at a tipping point where companies have started to bring algorithms to the market, and questions arise whether the pathology community is ready to implement AI in routine workflow. However, concerns also arise about the use of AI in pathology. This article reviews the pros and cons of introducing AI in diagnostic pathology.
Collapse
Affiliation(s)
- Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rachel N Flach
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Oncological Urology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Seher Makineli
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerben E Breimer
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul Pham
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Mitko Veta
- Department of Oncological Urology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
50
|
Zhao S, Yan CY, Lv H, Yang JC, You C, Li ZA, Ma D, Xiao Y, Hu J, Yang WT, Jiang YZ, Xu J, Shao ZM. Deep learning framework for comprehensive molecular and prognostic stratifications of triple-negative breast cancer. FUNDAMENTAL RESEARCH 2024; 4:678-689. [PMID: 38933195 PMCID: PMC11197495 DOI: 10.1016/j.fmre.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most challenging breast cancer subtype. Molecular stratification and target therapy bring clinical benefit for TNBC patients, but it is difficult to implement comprehensive molecular testing in clinical practice. Here, using our multi-omics TNBC cohort (N = 425), a deep learning-based framework was devised and validated for comprehensive predictions of molecular features, subtypes and prognosis from pathological whole slide images. The framework first incorporated a neural network to decompose the tissue on WSIs, followed by a second one which was trained based on certain tissue types for predicting different targets. Multi-omics molecular features were analyzed including somatic mutations, copy number alterations, germline mutations, biological pathway activities, metabolomics features and immunotherapy biomarkers. It was shown that the molecular features with therapeutic implications can be predicted including the somatic PIK3CA mutation, germline BRCA2 mutation and PD-L1 protein expression (area under the curve [AUC]: 0.78, 0.79 and 0.74 respectively). The molecular subtypes of TNBC can be identified (AUC: 0.84, 0.85, 0.93 and 0.73 for the basal-like immune-suppressed, immunomodulatory, luminal androgen receptor, and mesenchymal-like subtypes respectively) and their distinctive morphological patterns were revealed, which provided novel insights into the heterogeneity of TNBC. A neural network integrating image features and clinical covariates stratified patients into groups with different survival outcomes (log-rank P < 0.001). Our prediction framework and neural network models were externally validated on the TNBC cases from TCGA (N = 143) and appeared robust to the changes in patient population. For potential clinical translation, we built a novel online platform, where we modularized and deployed our framework along with the validated models. It can realize real-time one-stop prediction for new cases. In summary, using only pathological WSIs, our proposed framework can enable comprehensive stratifications of TNBC patients and provide valuable information for therapeutic decision-making. It had the potential to be clinically implemented and promote the personalized management of TNBC.
Collapse
Affiliation(s)
- Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Chao-Yang Yan
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jing-Cheng Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Greater Bay Area Institute of Precision Medicine, Guangzhou 511466, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zi-Ang Li
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jia Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wen-Tao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jun Xu
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing 210044, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| |
Collapse
|