1
|
Liao Y, Chen X, Hu S, Chen B, Zhuo X, Xu H, Wu X, Zeng X, Zeng H, Zhang D, Zhi Y, Zhao L. Artificial Intelligence for Predicting HER2 Status of Gastric Cancer Based on Whole-Slide Histopathology Images: A Retrospective Multicenter Study. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2408451. [PMID: 39792693 DOI: 10.1002/advs.202408451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/23/2024] [Revised: 12/15/2024] [Indexed: 01/12/2025]
Abstract
Human epidermal growth factor receptor 2 (HER2) positive gastric cancer (GC) shows a robust response to the combined therapy based HER2-targeted therapy. The application of these therapies is highly dependent on the evaluation of tumor HER2 status. However, there are many risks and challenges in HER2 assessment in GC. Therefore, an economically viable and readily available instrument is requisite for distinguishing HER2 status among patients diagnosed with GC. The study has innovatively developed a deep learning model, HER2Net, which can predict the HER2 status by quantitatively calculating the proportion of HER2 high-expression regions. The HER2Net is trained on an internal training set derived from 531 hematoxylin & eosin (H&E) whole slide images (WSI) of 520 patients. Subsequently, the performance of HER2Net is validated on an internal test set from 115 H&E WSI of 111 patients and an external multi-center test set from 102 H&E WSI of 101 patients. The HER2Net achieves an accuracy of 0.9043 on the internal test set, and an accuracy of 0.8922 on an external test set from multiple institutes. This discovery indicates that the HER2Net can potentially offer a novel methodology for the identification of HER2-positive GC.
Collapse
Affiliation(s)
- Yuhan Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xinhua Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shupeng Hu
- School of Computer Science, University of Manchester, Manchester, M13 9PL, UK
| | - Bing Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xinghua Zhuo
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaojin Wu
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Xiaofeng Zeng
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Huimin Zeng
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| | - Donghui Zhang
- Department of Pathology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Yunfei Zhi
- Department of Gastroenterology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pathology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528399, China
| |
Collapse
|
2
|
Cheng X, Zhao W, Ren D, Xia X, Lu S, Chen D, Wang X, Li Q, Lu Q, Gu Y, Bian X, Yu P, Dong W. RNA transcription assisted universal CRISPR/Cas12a system for programmable analysis of multiple colorectal cancer-associated microRNAs. Talanta 2025; 282:126960. [PMID: 39362038 DOI: 10.1016/j.talanta.2024.126960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Accurate analysis of multiple microRNA (miRNA) levels is significantly valuable for early diagnosis of colorectal cancer noninvasively considering the miRNA expression is highly relevant to the occurrence and progression of cancer. However, the low abundance and high sequence homology of miRNAs make their precise determination extremely challenging. Here, we developed a universal and programmable diagnostic strategy allowing for analyzing multiple colorectal cancer-associated miRNAs. The system combined sequentially programmable rolling circle transcription (RCT) and the CRISPR/Cas12a system with high trans-cleavage activity to achieve highly sensitive and specific detection of four target miRNAs. Owing to the remarkable performance of universal RCT-Cas12a strategy, this biosensor could detect miR-21, miR-17, miR-31 and miR-92a with a LOD of 2.1, 1.6, 3.7 and 1.0 pM, respectively. This strategy had a unique advantage in distinguishing human normal colon epithelial cells lines (NCM460) from human colon cancer cells (HT29). In particular, the designed system exhibited superior analytical capability in distinguishing paracancerous and colorectal cancer tissues from patients undergoing colorectal cancer surgery. This arbitrarily programmable, scalable, fast and specific strategy potentially offered an attractive alternative to handle varied challenges encountered with CRISPR-based systems, and held immense promise in scientific research and clinical applications.
Collapse
Affiliation(s)
- Xia Cheng
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenchen Zhao
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China; School of Clinical Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Dandan Ren
- Anhui Provincial Engineering Research Center for Dental materials and application, Wannan Medical College,Wuhu, Anhui, 241002, China
| | - Xinyi Xia
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Sijia Lu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Daixi Chen
- School of Clinical Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Xiaohong Wang
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China
| | - Qijun Li
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China
| | - Qi Lu
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China
| | - Yan Gu
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China
| | - Xiaolan Bian
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ping Yu
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wenpei Dong
- Department of General Surgery, Huadong Hospital, Fudan University, No.221 West Yan'an Road, Shanghai, 200040, China.
| |
Collapse
|
3
|
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] [Academic Contribution 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
|
4
|
de Back TR, van Hooff SR, Sommeijer DW, Vermeulen L. Transcriptomic subtyping of gastrointestinal malignancies. Trends Cancer 2024; 10:842-856. [PMID: 39019673 DOI: 10.1016/j.trecan.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/25/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Gastrointestinal (GI) cancers are highly heterogeneous at multiple levels. Tumor heterogeneity can be captured by molecular profiling, such as genetic, epigenetic, proteomic, and transcriptomic classification. Transcriptomic subtyping has the advantage of combining genetic and epigenetic information, cancer cell-intrinsic properties, and the tumor microenvironment (TME). Unsupervised transcriptomic subtyping systems of different GI malignancies have gained interest because they reveal shared biological features across cancers and bear prognostic and predictive value. Importantly, transcriptomic subtypes accurately reflect complex phenotypic states varying not only per tumor region, but also throughout disease progression, with consequences for clinical management. Here, we discuss methodologies of transcriptomic subtyping, proposed taxonomies for GI malignancies, and the challenges posed to clinical implementation, highlighting opportunities for future transcriptomic profiling efforts to optimize clinical impact.
Collapse
Affiliation(s)
- Tim R de Back
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sander R van Hooff
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Dirkje W Sommeijer
- Flevohospital, Department of Internal Medicine, Hospitaalweg 1, 1315 RA, Almere, The Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Asadi-Aghbolaghi M, Darbandsari A, Zhang A, Contreras-Sanz A, Boschman J, Ahmadvand P, Köbel M, Farnell D, Huntsman DG, Churg A, Black PC, Wang G, Gilks CB, Farahani H, Bashashati A. Learning generalizable AI models for multi-center histopathology image classification. NPJ Precis Oncol 2024; 8:151. [PMID: 39030380 PMCID: PMC11271637 DOI: 10.1038/s41698-024-00652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2023] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Investigation of histopathology slides by pathologists is an indispensable component of the routine diagnosis of cancer. Artificial intelligence (AI) has the potential to enhance diagnostic accuracy, improve efficiency, and patient outcomes in clinical pathology. However, variations in tissue preparation, staining protocols, and histopathology slide digitization could result in over-fitting of deep learning models when trained on the data from only one center, thereby underscoring the necessity to generalize deep learning networks for multi-center use. Several techniques, including the use of grayscale images, color normalization techniques, and Adversarial Domain Adaptation (ADA) have been suggested to generalize deep learning algorithms, but there are limitations to their effectiveness and discriminability. Convolutional Neural Networks (CNNs) exhibit higher sensitivity to variations in the amplitude spectrum, whereas humans predominantly rely on phase-related components for object recognition. As such, we propose Adversarial fourIer-based Domain Adaptation (AIDA) which applies the advantages of a Fourier transform in adversarial domain adaptation. We conducted a comprehensive examination of subtype classification tasks in four cancers, incorporating cases from multiple medical centers. Specifically, the datasets included multi-center data for 1113 ovarian cancer cases, 247 pleural cancer cases, 422 bladder cancer cases, and 482 breast cancer cases. Our proposed approach significantly improved performance, achieving superior classification results in the target domain, surpassing the baseline, color augmentation and normalization techniques, and ADA. Furthermore, extensive pathologist reviews suggested that our proposed approach, AIDA, successfully identifies known histotype-specific features. This superior performance highlights AIDA's potential in addressing generalization challenges in deep learning models for multi-center histopathology datasets.
Collapse
Affiliation(s)
| | - Amirali Darbandsari
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Allen Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Jeffrey Boschman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Pouya Ahmadvand
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Churg
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Gang Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Hossein Farahani
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ali Bashashati
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
6
|
Wang M, Yan X, Dong Y, Li X, Gao B. Machine learning and multi-omics data reveal driver gene-based molecular subtypes in hepatocellular carcinoma for precision treatment. PLoS Comput Biol 2024; 20:e1012113. [PMID: 38728362 PMCID: PMC11230636 DOI: 10.1371/journal.pcbi.1012113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/02/2024] [Revised: 07/08/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
The heterogeneity of Hepatocellular Carcinoma (HCC) poses a barrier to effective treatment. Stratifying highly heterogeneous HCC into molecular subtypes with similar features is crucial for personalized anti-tumor therapies. Although driver genes play pivotal roles in cancer progression, their potential in HCC subtyping has been largely overlooked. This study aims to utilize driver genes to construct HCC subtype models and unravel their molecular mechanisms. Utilizing a novel computational framework, we expanded the initially identified 96 driver genes to 1192 based on mutational aspects and an additional 233 considering driver dysregulation. These genes were subsequently employed as stratification markers for further analyses. A novel multi-omics subtype classification algorithm was developed, leveraging mutation and expression data of the identified stratification genes. This algorithm successfully categorized HCC into two distinct subtypes, CLASS A and CLASS B, demonstrating significant differences in survival outcomes. Integrating multi-omics and single-cell data unveiled substantial distinctions between these subtypes regarding transcriptomics, mutations, copy number variations, and epigenomics. Moreover, our prognostic model exhibited excellent predictive performance in training and external validation cohorts. Finally, a 10-gene classification model for these subtypes identified TTK as a promising therapeutic target with robust classification capabilities. This comprehensive study provides a novel perspective on HCC stratification, offering crucial insights for a deeper understanding of its pathogenesis and the development of promising treatment strategies.
Collapse
Affiliation(s)
- Meng Wang
- Faculty of Environment and Life of Beijing University of Technology, Beijing, China
| | - Xinyue Yan
- Faculty of Environment and Life of Beijing University of Technology, Beijing, China
| | - Yanan Dong
- Faculty of Environment and Life of Beijing University of Technology, Beijing, China
| | - Xiaoqin Li
- Faculty of Environment and Life of Beijing University of Technology, Beijing, China
| | - Bin Gao
- Faculty of Environment and Life of Beijing University of Technology, Beijing, China
| |
Collapse
|
7
|
Mayer RS, Kinzler MN, Stoll AK, Gretser S, Ziegler PK, Saborowski A, Reis H, Vogel A, Wild PJ, Flinner N. [The model transferability of AI in digital pathology : Potential and reality]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:124-132. [PMID: 38372762 PMCID: PMC10901943 DOI: 10.1007/s00292-024-01299-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Accepted: 12/18/2023] [Indexed: 02/20/2024]
Abstract
OBJECTIVE Artificial intelligence (AI) holds the potential to make significant advancements in pathology. However, its actual implementation and certification for practical use are currently limited, often due to challenges related to model transferability. In this context, we investigate the factors influencing transferability and present methods aimed at enhancing the utilization of AI algorithms in pathology. MATERIALS AND METHODS Various convolutional neural networks (CNNs) and vision transformers (ViTs) were trained using datasets from two institutions, along with the publicly available TCGA-MIBC dataset. These networks conducted predictions in urothelial tissue and intrahepatic cholangiocarcinoma (iCCA). The objective was to illustrate the impact of stain normalization, the influence of various artifacts during both training and testing, as well as the effects of the NoisyEnsemble method. RESULTS We were able to demonstrate that stain normalization of slides from different institutions has a significant positive effect on the inter-institutional transferability of CNNs and ViTs (respectively +13% and +10%). In addition, ViTs usually achieve a higher accuracy in the external test (here +1.5%). Similarly, we showcased how artifacts in test data can negatively affect CNN predictions and how incorporating these artifacts during training leads to improvements. Lastly, NoisyEnsembles of CNNs (better than ViTs) were shown to enhance transferability across different tissues and research questions (+7% Bladder, +15% iCCA). DISCUSSION It is crucial to be aware of the transferability challenge: achieving good performance during development does not necessarily translate to good performance in real-world applications. The inclusion of existing methods to enhance transferability, such as stain normalization and NoisyEnsemble, and their ongoing refinement, is of importance.
Collapse
Affiliation(s)
- Robin S Mayer
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
| | - Maximilian N Kinzler
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
- Universitätsklinikum, Medizinische Klinik 1, Goethe-Universität Frankfurt, Frankfurt am Main, Deutschland
| | - Alexandra K Stoll
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Deutschland
| | - Steffen Gretser
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
| | - Paul K Ziegler
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
| | - Anna Saborowski
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Henning Reis
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
| | - Arndt Vogel
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Deutschland
| | - Peter J Wild
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Deutschland
- Wildlab, University Hospital Frankfurt MVZ GmbH, Frankfurt am Main, Deutschland
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Deutschland
- University Cancer Center (UCT) Frankfurt-Marburg, Frankfurt am Main, Deutschland
| | - Nadine Flinner
- Universitätsklinikum, Dr. Senckenbergisches Institut für Pathologie, Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Deutschland.
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Deutschland.
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Deutschland.
- University Cancer Center (UCT) Frankfurt-Marburg, Frankfurt am Main, Deutschland.
| |
Collapse
|
8
|
Chaves J, Libânio D, Pimentel-Nunes P. Management of high risk T1 gastric adenocarcinoma following endoscopic resection. Best Pract Res Clin Gastroenterol 2024; 68:101887. [PMID: 38522891 DOI: 10.1016/j.bpg.2024.101887] [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] [Academic Contribution Register] [Received: 08/14/2023] [Accepted: 01/23/2024] [Indexed: 03/26/2024]
Abstract
Endoscopic submucosal dissection has revolutionized the treatment of early gastric cancer. However, cases that do not meet the curability criteria have a higher risk of lymph node metastasis and salvage surgery is still considered the next treatment approach to increase the chance of cure. Nevertheless, not all high-risk resections entail the same level of risk, emphasizing the utmost importance of individualized stratification for further treatment. In this review, we aim to examine the current evidence concerning the management following a high-risk non-curative resection, highlighting the existing approaches, while also presenting upcoming strategies that attempt to improve patient outcomes, minimize adverse events, and provide a tailored management.
Collapse
Affiliation(s)
- Jéssica Chaves
- Department of Gastroenterology, Porto Comprehensive Cancer Center, Porto, Portugal.
| | - Diogo Libânio
- Department of Gastroenterology, Porto Comprehensive Cancer Center, Porto, Portugal; MEDCIDS- Department of Community, Medicine, Health Information and Decision, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Pedro Pimentel-Nunes
- Gastroenterology, Unilabs, Portugal; Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
9
|
Ahn JC, Shah VH. Artificial intelligence in gastroenterology and hepatology. ARTIFICIAL INTELLIGENCE IN CLINICAL PRACTICE 2024:443-464. [DOI: 10.1016/b978-0-443-15688-5.00016-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2025]
|
10
|
Kim GJ, Lee T, Ahn S, Uh Y, Kim SH. Efficient diagnosis of IDH-mutant gliomas: 1p/19qNET assesses 1p/19q codeletion status using weakly-supervised learning. NPJ Precis Oncol 2023; 7:94. [PMID: 37717080 PMCID: PMC10505231 DOI: 10.1038/s41698-023-00450-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/24/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023] Open
Abstract
Accurate identification of molecular alterations in gliomas is crucial for their diagnosis and treatment. Although, fluorescence in situ hybridization (FISH) allows for the observation of diverse and heterogeneous alterations, it is inherently time-consuming and challenging due to the limitations of the molecular method. Here, we report the development of 1p/19qNET, an advanced deep-learning network designed to predict fold change values of 1p and 19q chromosomes and classify isocitrate dehydrogenase (IDH)-mutant gliomas from whole-slide images. We trained 1p/19qNET on next-generation sequencing data from a discovery set (DS) of 288 patients and utilized a weakly-supervised approach with slide-level labels to reduce bias and workload. We then performed validation on an independent validation set (IVS) comprising 385 samples from The Cancer Genome Atlas, a comprehensive cancer genomics resource. 1p/19qNET outperformed traditional FISH, achieving R2 values of 0.589 and 0.547 for the 1p and 19q arms, respectively. As an IDH-mutant glioma classifier, 1p/19qNET attained AUCs of 0.930 and 0.837 in the DS and IVS, respectively. The weakly-supervised nature of 1p/19qNET provides explainable heatmaps for the results. This study demonstrates the successful use of deep learning for precise determination of 1p/19q codeletion status and classification of IDH-mutant gliomas as astrocytoma or oligodendroglioma. 1p/19qNET offers comparable results to FISH and provides informative spatial information. This approach has broader applications in tumor classification.
Collapse
Affiliation(s)
- Gi Jeong Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University Graduate School, Seoul, Republic of Korea
| | - Tonghyun Lee
- Department of Artificial Intelligence, Yonsei University College of Computing, Seoul, Republic of Korea
| | - Sangjeong Ahn
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youngjung Uh
- Department of Artificial Intelligence, Yonsei University College of Computing, Seoul, Republic of Korea.
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Wang Z, Liu Y, Niu X. Application of artificial intelligence for improving early detection and prediction of therapeutic outcomes for gastric cancer in the era of precision oncology. Semin Cancer Biol 2023; 93:83-96. [PMID: 37116818 DOI: 10.1016/j.semcancer.2023.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Gastric cancer is a leading contributor to cancer incidence and mortality globally. Recently, artificial intelligence approaches, particularly machine learning and deep learning, are rapidly reshaping the full spectrum of clinical management for gastric cancer. Machine learning is formed from computers running repeated iterative models for progressively improving performance on a particular task. Deep learning is a subtype of machine learning on the basis of multilayered neural networks inspired by the human brain. This review summarizes the application of artificial intelligence algorithms to multi-dimensional data including clinical and follow-up information, conventional images (endoscope, histopathology, and computed tomography (CT)), molecular biomarkers, etc. to improve the risk surveillance of gastric cancer with established risk factors; the accuracy of diagnosis, and survival prediction among established gastric cancer patients; and the prediction of treatment outcomes for assisting clinical decision making. Therefore, artificial intelligence makes a profound impact on almost all aspects of gastric cancer from improving diagnosis to precision medicine. Despite this, most established artificial intelligence-based models are in a research-based format and often have limited value in real-world clinical practice. With the increasing adoption of artificial intelligence in clinical use, we anticipate the arrival of artificial intelligence-powered gastric cancer care.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning, China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning, China.
| | - Xing Niu
- China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
12
|
Choi S, Kim S. Artificial Intelligence in the Pathology of Gastric Cancer. J Gastric Cancer 2023; 23:410-427. [PMID: 37553129 PMCID: PMC10412971 DOI: 10.5230/jgc.2023.23.e25] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Recent advances in artificial intelligence (AI) have provided novel tools for rapid and precise pathologic diagnosis. The introduction of digital pathology has enabled the acquisition of scanned slide images that are essential for the application of AI. The application of AI for improved pathologic diagnosis includes the error-free detection of potentially negligible lesions, such as a minute focus of metastatic tumor cells in lymph nodes, the accurate diagnosis of potentially controversial histologic findings, such as very well-differentiated carcinomas mimicking normal epithelial tissues, and the pathological subtyping of the cancers. Additionally, the utilization of AI algorithms enables the precise decision of the score of immunohistochemical markers for targeted therapies, such as human epidermal growth factor receptor 2 and programmed death-ligand 1. Studies have revealed that AI assistance can reduce the discordance of interpretation between pathologists and more accurately predict clinical outcomes. Several approaches have been employed to develop novel biomarkers from histologic images using AI. Moreover, AI-assisted analysis of the cancer microenvironment showed that the distribution of tumor-infiltrating lymphocytes was related to the response to the immune checkpoint inhibitor therapy, emphasizing its value as a biomarker. As numerous studies have demonstrated the significance of AI-assisted interpretation and biomarker development, the AI-based approach will advance diagnostic pathology.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
13
|
Tolkach Y, Wolgast LM, Damanakis A, Pryalukhin A, Schallenberg S, Hulla W, Eich ML, Schroeder W, Mukhopadhyay A, Fuchs M, Klein S, Bruns C, Büttner R, Gebauer F, Schömig-Markiefka B, Quaas A. Artificial intelligence for tumour tissue detection and histological regression grading in oesophageal adenocarcinomas: a retrospective algorithm development and validation study. Lancet Digit Health 2023; 5:e265-e275. [PMID: 37100542 DOI: 10.1016/s2589-7500(23)00027-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND Oesophageal adenocarcinoma and adenocarcinoma of the oesophagogastric junction are among the most common malignant epithelial tumours. Most patients receive neoadjuvant therapy before complete tumour resection. Histological assessment after resection includes identification of residual tumour tissue and areas of regressive tumour, data which are used to calculate a clinically relevant regression score. We developed an artificial intelligence (AI) algorithm for tumour tissue detection and tumour regression grading in surgical specimens from patients with oesophageal adenocarcinoma or adenocarcinoma of the oesophagogastric junction. METHODS We used one training cohort and four independent test cohorts to develop, train, and validate a deep learning tool. The material consisted of histological slides from surgically resected specimens from patients with oesophageal adenocarcinoma and adenocarcinoma of the oesophagogastric junction from three pathology institutes (two in Germany, one in Austria) and oesophageal cancer cohort of The Cancer Genome Atlas (TCGA). All slides were from neoadjuvantly treated patients except for those from the TCGA cohort, who were neoadjuvant-therapy naive. Data from training cohort and test cohort cases were extensively manually annotated for 11 tissue classes. A convolutional neural network was trained on the data using a supervised principle. First, the tool was formally validated using manually annotated test datasets. Next, tumour regression grading was assessed in a retrospective cohort of post-neoadjuvant therapy surgical specimens. The grading of the algorithm was compared with that of a group of 12 board-certified pathologists from one department. To further validate the tool, three pathologists processed whole resection cases with and without AI assistance. FINDINGS Of the four test cohorts, one included 22 manually annotated histological slides (n=20 patients), one included 62 sides (n=15), one included 214 slides (n=69), and the final one included 22 manually annotated histological slides (n=22). In the independent test cohorts the AI tool had high patch-level accuracy for identifying both tumour and regression tissue. When we validated the concordance of the AI tool against analyses by a group of pathologists (n=12), agreement was 63·6% (quadratic kappa 0·749; p<0·0001) at case level. The AI-based regression grading triggered true reclassification of resected tumour slides in seven cases (including six cases who had small tumour regions that were initially missed by pathologists). Use of the AI tool by three pathologists increased interobserver agreement and substantially reduced diagnostic time per case compared with working without AI assistance. INTERPRETATION Use of our AI tool in the diagnostics of oesophageal adenocarcinoma resection specimens by pathologists increased diagnostic accuracy, interobserver concordance, and significantly reduced assessment time. Prospective validation of the tool is required. FUNDING North Rhine-Westphalia state, Federal Ministry of Education and Research of Germany, and the Wilhelm Sander Foundation.
Collapse
Affiliation(s)
- Yuri Tolkach
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany.
| | - Lisa Marie Wolgast
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexander Damanakis
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexey Pryalukhin
- Institute of Pathology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Simon Schallenberg
- Institute of Pathology, University Hospital Berlin-Charité, Berlin, Germany
| | - Wolfgang Hulla
- Institute of Pathology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Marie-Lisa Eich
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Wolfgang Schroeder
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | | | - Moritz Fuchs
- Technical University Darmstadt, Darmstadt, Germany
| | - Sebastian Klein
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Birgid Schömig-Markiefka
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
14
|
Xiao Y, Wang S, Ling R, Song Y. Application of artificial neural network algorithm in pathological diagnosis and prognosis prediction of digestive tract malignant tumors. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:243-248. [PMID: 37283110 DOI: 10.3724/zdxbyxb-2022-0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/08/2023]
Abstract
The application of artificial neural network algorithm in pathological diagnosis of gastrointestinal malignant tumors has become a research hotspot. In the previous studies, the algorithm research mainly focused on the model development based on convolutional neural networks, while only a few studies used the combination of convolutional neural networks and recurrent neural networks. The research contents included classical histopathological diagnosis and molecular typing of malignant tumors, and the prediction of patient prognosis by utilizing artificial neural networks. This article reviews the research progress on artificial neural network algorithm in the pathological diagnosis and prognosis prediction of digestive tract malignant tumors.
Collapse
Affiliation(s)
- Ya Xiao
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China.
| | - Shuyang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai 200032, China
| | - Ren Ling
- Shanghai Laizi Software Technology Co. Ltd., Shanghai 201499, China
| | - Yufei Song
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo 315046, Zhejiang Province, China.
| |
Collapse
|
15
|
Yavuz A, Alpsoy A, Gedik EO, Celik MY, Bassorgun CI, Unal B, Elpek GO. Artificial intelligence applications in predicting the behavior of gastrointestinal cancers in pathology. Artif Intell Gastroenterol 2022; 3:142-162. [DOI: 10.35712/aig.v3.i5.142] [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] [Academic Contribution Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Recent research has provided a wealth of data supporting the application of artificial intelligence (AI)-based applications in routine pathology practice. Indeed, it is clear that these methods can significantly support an accurate and rapid diagnosis by eliminating errors, increasing reliability, and improving workflow. In addition, the effectiveness of AI in the pathological evaluation of prognostic parameters associated with behavior, course, and treatment in many types of tumors has also been noted. Regarding gastrointestinal system (GIS) cancers, the contribution of AI methods to pathological diagnosis has been investigated in many studies. On the other hand, studies focusing on AI applications in evaluating parameters to determine tumor behavior are relatively few. For this purpose, the potential of AI models has been studied over a broad spectrum, from tumor subtyping to the identification of new digital biomarkers. The capacity of AI to infer genetic alterations of cancer tissues from digital slides has been demonstrated. Although current data suggest the merit of AI-based approaches in assessing tumor behavior in GIS cancers, a wide range of challenges still need to be solved, from laboratory infrastructure to improving the robustness of algorithms, before incorporating AI applications into real-life GIS pathology practice. This review aims to present data from AI applications in evaluating pathological parameters related to the behavior of GIS cancer with an overview of the opportunities and challenges encountered in implementing AI in pathology.
Collapse
Affiliation(s)
- Aysen Yavuz
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Anil Alpsoy
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Elif Ocak Gedik
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | | | | | - Betul Unal
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Gulsum Ozlem Elpek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| |
Collapse
|
16
|
Deep learning model to predict Epstein-Barr virus associated gastric cancer in histology. Sci Rep 2022; 12:18466. [PMID: 36323712 PMCID: PMC9630260 DOI: 10.1038/s41598-022-22731-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2022] [Accepted: 10/18/2022] [Indexed: 11/20/2022] Open
Abstract
The detection of Epstein-Barr virus (EBV) in gastric cancer patients is crucial for clinical decision making, as it is related with specific treatment responses and prognoses. Despite its importance, the limited medical resources preclude universal EBV testing. Herein, we propose a deep learning-based EBV prediction method from H&E-stained whole-slide images (WSI). Our model was developed using 319 H&E stained WSI (26 EBV positive; TCGA dataset) from the Cancer Genome Atlas, and 108 WSI (8 EBV positive; ISH dataset) from an independent institution. Our deep learning model, EBVNet consists of two sequential components: a tumor classifier and an EBV classifier. We visualized the learned representation by the classifiers using UMAP. We externally validated the model using 60 additional WSI (7 being EBV positive; HGH dataset). We compared the model's performance with those of four pathologists. EBVNet achieved an AUPRC of 0.65, whereas the four pathologists yielded a mean AUPRC of 0.41. Moreover, EBVNet achieved an negative predictive value, sensitivity, specificity, precision, and F1-score of 0.98, 0.86, 0.92, 0.60, and 0.71, respectively. Our proposed model is expected to contribute to prescreen patients for confirmatory testing, potentially to save test-related cost and labor.
Collapse
|
17
|
Mayer RS, Gretser S, Heckmann LE, Ziegler PK, Walter B, Reis H, Bankov K, Becker S, Triesch J, Wild PJ, Flinner N. How to learn with intentional mistakes: NoisyEnsembles to overcome poor tissue quality for deep learning in computational pathology. Front Med (Lausanne) 2022; 9:959068. [PMID: 36106328 PMCID: PMC9464871 DOI: 10.3389/fmed.2022.959068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
There is a lot of recent interest in the field of computational pathology, as many algorithms are introduced to detect, for example, cancer lesions or molecular features. However, there is a large gap between artificial intelligence (AI) technology and practice, since only a small fraction of the applications is used in routine diagnostics. The main problems are the transferability of convolutional neural network (CNN) models to data from other sources and the identification of uncertain predictions. The role of tissue quality itself is also largely unknown. Here, we demonstrated that samples of the TCGA ovarian cancer (TCGA-OV) dataset from different tissue sources have different quality characteristics and that CNN performance is linked to this property. CNNs performed best on high-quality data. Quality control tools were partially able to identify low-quality tiles, but their use did not increase the performance of the trained CNNs. Furthermore, we trained NoisyEnsembles by introducing label noise during training. These NoisyEnsembles could improve CNN performance for low-quality, unknown datasets. Moreover, the performance increases as the ensemble become more consistent, suggesting that incorrect predictions could be discarded efficiently to avoid wrong diagnostic decisions.
Collapse
Affiliation(s)
- Robin S. Mayer
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Steffen Gretser
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Lara E. Heckmann
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Paul K. Ziegler
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Britta Walter
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Henning Reis
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Katrin Bankov
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Sven Becker
- Department of Gynecology and Obstetrics, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
| | - Peter J. Wild
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
- Wildlab, University Hospital Frankfurt MVZ GmbH, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt-Marburg, Frankfurt am Main, Germany
| | - Nadine Flinner
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- University Cancer Center (UCT) Frankfurt-Marburg, Frankfurt am Main, Germany
| |
Collapse
|
18
|
Angerilli V, Pennelli G, Galuppini F, Realdon S, Fantin A, Savarino E, Farinati F, Mastracci L, Luchini C, Fassan M. Molecular subtyping of gastroesophageal dysplasia heterogeneity according to TCGA/ACRG classes. Virchows Arch 2022; 481:545-552. [PMID: 35925389 PMCID: PMC9534804 DOI: 10.1007/s00428-022-03392-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/07/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022]
Abstract
Gastric adenocarcinoma has recently been classified into several subtypes on the basis of molecular profiling, which has been successfully reproduced by immunohistochemistry (IHC) and in situ hybridization (ISH). A series of 73 gastroesophageal dysplastic lesions (37 gastric dysplasia and 36 Barrett dysplasia; 44 low-grade dysplasia and 29 high-grade dysplasia) was investigated for mismatch repair proteins, E-cadherin, p53, and EBER status, to reproduce The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG) molecular clustering. Overall, the dysplastic lesions were classified as follows: according to TCGA classification, EBV, 0/73 (0%), MSI, 6/73 (8.2%), GS, 4/73 (5.5%), CIN, 63/73 (86.3%); according to ACRG molecular subtyping, MSI, 6/73 (8.2%), MSS/EMT, 4/73 (5.5%), MSS/TP53−, 33/73 (45.2%), MSS/TP53+, 30/73 (41.1%). A positive association was found between MSS/TP53− and Barrett dysplasia (p = 0.0004), between MSS/TP53+ and LG dysplasia (p = 0.001) and between MSS/TP53+ and gastric dysplasia (p = 0.0018). Gastroesophageal dysplastic lesions proved to be heterogenous in terms of TCGA/ACRG classes, but with a different distribution from that of cancers, with no EBV-positive cases, an increasing presence of mismatch repair deficiency from low grade to high grade lesions, and a prevalence of p53 aberrations in Barrett dysplasia. The present study further demonstrated that gastroesophageal dysplastic lesions may be characterized by alterations in predictive/prognostic biomarkers, and this should be considered in routine diagnostic.
Collapse
Affiliation(s)
- Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, via Gabelli 61, 35121, Padua, Italy
| | - Gianmaria Pennelli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, via Gabelli 61, 35121, Padua, Italy
| | - Francesca Galuppini
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, via Gabelli 61, 35121, Padua, Italy
| | | | | | - Edoardo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Luca Mastracci
- Department of Surgical Science and Integrated Diagnostics (DISC), University of Genova, Genoa, Italy
| | - Claudio Luchini
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, via Gabelli 61, 35121, Padua, Italy. .,Istituto Oncologico Veneto-IOV-IRCCS, Padua, Italy.
| |
Collapse
|