1
|
Rehman ZU, Ahmad Fauzi MF, Wan Ahmad WSHM, Abas FS, Cheah PL, Chiew SF, Looi LM. Review of In Situ Hybridization (ISH) Stain Images Using Computational Techniques. Diagnostics (Basel) 2024; 14:2089. [PMID: 39335767 PMCID: PMC11430898 DOI: 10.3390/diagnostics14182089] [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: 07/30/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements in medical imaging have greatly enhanced the application of computational techniques in digital pathology, particularly for the classification of breast cancer using in situ hybridization (ISH) imaging. HER2 amplification, a key prognostic marker in 20-25% of breast cancers, can be assessed through alterations in gene copy number or protein expression. However, challenges persist due to the heterogeneity of nuclear regions and complexities in cancer biomarker detection. This review examines semi-automated and fully automated computational methods for analyzing ISH images with a focus on HER2 gene amplification. Literature from 1997 to 2023 is analyzed, emphasizing silver-enhanced in situ hybridization (SISH) and its integration with image processing and machine learning techniques. Both conventional machine learning approaches and recent advances in deep learning are compared. The review reveals that automated ISH analysis in combination with bright-field microscopy provides a cost-effective and scalable solution for routine pathology. The integration of deep learning techniques shows promise in improving accuracy over conventional methods, although there are limitations related to data variability and computational demands. Automated ISH analysis can reduce manual labor and increase diagnostic accuracy. Future research should focus on refining these computational methods, particularly in handling the complex nature of HER2 status evaluation, and integrate best practices to further enhance clinical adoption of these techniques.
Collapse
Affiliation(s)
- Zaka Ur Rehman
- Faculty of Engineering, Multimedia University, Cyberjaya 63100, Malaysia
| | | | - Wan Siti Halimatul Munirah Wan Ahmad
- Faculty of Engineering, Multimedia University, Cyberjaya 63100, Malaysia
- Institute for Research, Development and Innovation (IRDI), IMU University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Fazly Salleh Abas
- Faculty of Engineering and Technology, Multimedia University, Bukit Beruang, Melaka 75450, Malaysia
| | - Phaik Leng Cheah
- Department of Pathology, University Malaya-Medical Center, Kuala Lumpur 50603, Malaysia
| | - Seow Fan Chiew
- Department of Pathology, University Malaya-Medical Center, Kuala Lumpur 50603, Malaysia
| | - Lai-Meng Looi
- Department of Pathology, University Malaya-Medical Center, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
2
|
Jian Z, Song T, Zhang Z, Ai Z, Zhao H, Tang M, Liu K. An Improved Nested U-Net Network for Fluorescence In Situ Hybridization Cell Image Segmentation. SENSORS (BASEL, SWITZERLAND) 2024; 24:928. [PMID: 38339644 PMCID: PMC10857237 DOI: 10.3390/s24030928] [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: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024]
Abstract
Fluorescence in situ hybridization (FISH) is a powerful cytogenetic method used to precisely detect and localize nucleic acid sequences. This technique is proving to be an invaluable tool in medical diagnostics and has made significant contributions to biology and the life sciences. However, the number of cells is large and the nucleic acid sequences are disorganized in the FISH images taken using the microscope. Processing and analyzing images is a time-consuming and laborious task for researchers, as it can easily tire the human eyes and lead to errors in judgment. In recent years, deep learning has made significant progress in the field of medical imaging, especially the successful application of introducing the attention mechanism. The attention mechanism, as a key component of deep learning, improves the understanding and interpretation of medical images by giving different weights to different regions of the image, enabling the model to focus more on important features. To address the challenges in FISH image analysis, we combined medical imaging with deep learning to develop the SEAM-Unet++ automated cell contour segmentation algorithm with integrated attention mechanism. The significant advantage of this algorithm is that it improves the accuracy of cell contours in FISH images. Experiments have demonstrated that by introducing the attention mechanism, our method is able to segment cells that are adherent to each other more efficiently.
Collapse
Affiliation(s)
| | | | | | | | | | - Man Tang
- School of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan 430200, China; (Z.J.); (T.S.); (Z.Z.); (Z.A.); (H.Z.)
| | - Kan Liu
- School of Electronic and Electrical Engineering, Wuhan Textile University, Wuhan 430200, China; (Z.J.); (T.S.); (Z.Z.); (Z.A.); (H.Z.)
| |
Collapse
|
3
|
Lee S, Kim G, Lee J, Lee AC, Kwon S. Mapping cancer biology in space: applications and perspectives on spatial omics for oncology. Mol Cancer 2024; 23:26. [PMID: 38291400 PMCID: PMC10826015 DOI: 10.1186/s12943-024-01941-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024] Open
Abstract
Technologies to decipher cellular biology, such as bulk sequencing technologies and single-cell sequencing technologies, have greatly assisted novel findings in tumor biology. Recent findings in tumor biology suggest that tumors construct architectures that influence the underlying cancerous mechanisms. Increasing research has reported novel techniques to map the tissue in a spatial context or targeted sampling-based characterization and has introduced such technologies to solve oncology regarding tumor heterogeneity, tumor microenvironment, and spatially located biomarkers. In this study, we address spatial technologies that can delineate the omics profile in a spatial context, novel findings discovered via spatial technologies in oncology, and suggest perspectives regarding therapeutic approaches and further technological developments.
Collapse
Affiliation(s)
- Sumin Lee
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Meteor Biotech,, Co. Ltd, Seoul, 08826, Republic of Korea
| | - Gyeongjun Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - JinYoung Lee
- Division of Engineering Science, University of Toronto, Toronto, Ontario, ON, M5S 3H6, Canada
| | - Amos C Lee
- Meteor Biotech,, Co. Ltd, Seoul, 08826, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Sunghoon Kwon
- Department of Electrical and Computer Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
4
|
Ahn JS, Shin S, Yang SA, Park EK, Kim KH, Cho SI, Ock CY, Kim S. Artificial Intelligence in Breast Cancer Diagnosis and Personalized Medicine. J Breast Cancer 2023; 26:405-435. [PMID: 37926067 PMCID: PMC10625863 DOI: 10.4048/jbc.2023.26.e45] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Breast cancer is a significant cause of cancer-related mortality in women worldwide. Early and precise diagnosis is crucial, and clinical outcomes can be markedly enhanced. The rise of artificial intelligence (AI) has ushered in a new era, notably in image analysis, paving the way for major advancements in breast cancer diagnosis and individualized treatment regimens. In the diagnostic workflow for patients with breast cancer, the role of AI encompasses screening, diagnosis, staging, biomarker evaluation, prognostication, and therapeutic response prediction. Although its potential is immense, its complete integration into clinical practice is challenging. Particularly, these challenges include the imperatives for extensive clinical validation, model generalizability, navigating the "black-box" conundrum, and pragmatic considerations of embedding AI into everyday clinical environments. In this review, we comprehensively explored the diverse applications of AI in breast cancer care, underlining its transformative promise and existing impediments. In radiology, we specifically address AI in mammography, tomosynthesis, risk prediction models, and supplementary imaging methods, including magnetic resonance imaging and ultrasound. In pathology, our focus is on AI applications for pathologic diagnosis, evaluation of biomarkers, and predictions related to genetic alterations, treatment response, and prognosis in the context of breast cancer diagnosis and treatment. Our discussion underscores the transformative potential of AI in breast cancer management and emphasizes the importance of focused research to realize the full spectrum of benefits of AI in patient care.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - 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
|
5
|
Wang CW, Chu KL, Muzakky H, Lin YJ, Chao TK. Efficient Convolution Network to Assist Breast Cancer Diagnosis and Target Therapy. Cancers (Basel) 2023; 15:3991. [PMID: 37568809 PMCID: PMC10416960 DOI: 10.3390/cancers15153991] [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: 07/14/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths among women worldwide, and early detection and treatment has been shown to significantly reduce fatality rates from severe illness. Moreover, determination of the human epidermal growth factor receptor-2 (HER2) gene amplification by Fluorescence in situ hybridization (FISH) and Dual in situ hybridization (DISH) is critical for the selection of appropriate breast cancer patients for HER2-targeted therapy. However, visual examination of microscopy is time-consuming, subjective and poorly reproducible due to high inter-observer variability among pathologists and cytopathologists. The lack of consistency in identifying carcinoma-like nuclei has led to divergences in the calculation of sensitivity and specificity. This manuscript introduces a highly efficient deep learning method with low computing cost. The experimental results demonstrate that the proposed framework achieves high precision and recall on three essential clinical applications, including breast cancer diagnosis and human epidermal receptor factor 2 (HER2) amplification detection on FISH and DISH slides for HER2 target therapy. Furthermore, the proposed method outperforms the majority of the benchmark methods in terms of IoU by a significant margin (p<0.001) on three essential clinical applications. Importantly, run time analysis shows that the proposed method obtains excellent segmentation results with notably reduced time for Artificial intelligence (AI) training (16.93%), AI inference (17.25%) and memory usage (18.52%), making the proposed framework feasible for practical clinical usage.
Collapse
Affiliation(s)
- Ching-Wei Wang
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan; (K.-L.C.); (H.M.)
| | - Kai-Lin Chu
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan; (K.-L.C.); (H.M.)
| | - Hikam Muzakky
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan; (K.-L.C.); (H.M.)
| | - Yi-Jia Lin
- Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan;
- Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, Taipei 11490, Taiwan;
- Institute of Pathology and Parasitology, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
6
|
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: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar 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
|
7
|
Wang CW, Khalil MA, Lin YJ, Lee YC, Chao TK. Detection of ERBB2 and CEN17 signals in fluorescent in situ hybridization and dual in situ hybridization for guiding breast cancer HER2 target therapy. Artif Intell Med 2023; 141:102568. [PMID: 37295903 DOI: 10.1016/j.artmed.2023.102568] [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: 11/29/2022] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 06/12/2023]
Abstract
The overexpression of the human epidermal growth factor receptor 2 (HER2) is a predictive biomarker in therapeutic effects for metastatic breast cancer. Accurate HER2 testing is critical for determining the most suitable treatment for patients. Fluorescent in situ hybridization (FISH) and dual in situ hybridization (DISH) have been recognized as FDA-approved methods to determine HER2 overexpression. However, analysis of HER2 overexpression is challenging. Firstly, the boundaries of cells are often unclear and blurry, with large variations in cell shapes and signals, making it challenging to identify the precise areas of HER2-related cells. Secondly, the use of sparsely labeled data, where some unlabeled HER2-related cells are classified as background, can significantly confuse fully supervised AI learning and result in unsatisfactory model outcomes. In this study, we present a weakly supervised Cascade R-CNN (W-CRCNN) model to automatically detect HER2 overexpression in HER2 DISH and FISH images acquired from clinical breast cancer samples. The experimental results demonstrate that the proposed W-CRCNN achieves excellent results in identification of HER2 amplification in three datasets, including two DISH datasets and a FISH dataset. For the FISH dataset, the proposed W-CRCNN achieves an accuracy of 0.970±0.022, precision of 0.974±0.028, recall of 0.917±0.065, F1-score of 0.943±0.042 and Jaccard Index of 0.899±0.073. For DISH datasets, the proposed W-CRCNN achieves an accuracy of 0.971±0.024, precision of 0.969±0.015, recall of 0.925±0.020, F1-score of 0.947±0.036 and Jaccard Index of 0.884±0.103 for dataset 1, and an accuracy of 0.978±0.011, precision of 0.975±0.011, recall of 0.918±0.038, F1-score of 0.946±0.030 and Jaccard Index of 0.884±0.052 for dataset 2, respectively. In comparison with the benchmark methods, the proposed W-CRCNN significantly outperforms all the benchmark approaches in identification of HER2 overexpression in FISH and DISH datasets (p<0.05). With the high degree of accuracy, precision and recall , the results show that the proposed method in DISH analysis for assessment of HER2 overexpression in breast cancer patients has significant potential to assist precision medicine.
Collapse
Affiliation(s)
- Ching-Wei Wang
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan; Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Muhammad-Adil Khalil
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Yi-Jia Lin
- Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan; Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ching Lee
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology, Tri-Service General Hospital, Taipei, Taiwan; Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
8
|
Arshadi A, Tolomeo D, Venuto S, Storlazzi CT. Advancements in Focal Amplification Detection in Tumor/Liquid Biopsies and Emerging Clinical Applications. Genes (Basel) 2023; 14:1304. [PMID: 37372484 PMCID: PMC10298061 DOI: 10.3390/genes14061304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Focal amplifications (FAs) are crucial in cancer research due to their significant diagnostic, prognostic, and therapeutic implications. FAs manifest in various forms, such as episomes, double minute chromosomes, and homogeneously staining regions, arising through different mechanisms and mainly contributing to cancer cell heterogeneity, the leading cause of drug resistance in therapy. Numerous wet-lab, mainly FISH, PCR-based assays, next-generation sequencing, and bioinformatics approaches have been set up to detect FAs, unravel the internal structure of amplicons, assess their chromatin compaction status, and investigate the transcriptional landscape associated with their occurrence in cancer cells. Most of them are tailored for tumor samples, even at the single-cell level. Conversely, very limited approaches have been set up to detect FAs in liquid biopsies. This evidence suggests the need to improve these non-invasive investigations for early tumor detection, monitoring disease progression, and evaluating treatment response. Despite the potential therapeutic implications of FAs, such as, for example, the use of HER2-specific compounds for patients with ERBB2 amplification, challenges remain, including developing selective and effective FA-targeting agents and understanding the molecular mechanisms underlying FA maintenance and replication. This review details a state-of-the-art of FA investigation, with a particular focus on liquid biopsies and single-cell approaches in tumor samples, emphasizing their potential to revolutionize the future diagnosis, prognosis, and treatment of cancer patients.
Collapse
Affiliation(s)
| | | | | | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (S.V.)
| |
Collapse
|
9
|
Xue T, Chang H, Ren M, Wang H, Yang Y, Wang B, Lv L, Tang L, Fu C, Fang Q, He C, Zhu X, Zhou X, Bai Q. Deep learning to automatically evaluate HER2 gene amplification status from fluorescence in situ hybridization images. Sci Rep 2023; 13:9746. [PMID: 37328516 PMCID: PMC10275857 DOI: 10.1038/s41598-023-36811-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 06/10/2023] [Indexed: 06/18/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) gene amplification helps identify breast cancer patients who may respond to targeted anti-HER2 therapy. This study aims to develop an automated method for quantifying HER2 fluorescence in situ hybridization (FISH) signals and improve the working efficiency of pathologists. An Aitrox artificial intelligence (AI) model based on deep learning was constructed, and a comparison between the AI model and traditional manual counting was performed. In total, 918 FISH images from 320 consecutive invasive breast cancers were analysed and automatically classified into 5 groups according to the 2018 ASCO/CAP guidelines. The overall classification accuracy was 85.33% (157/184) with a mean average precision of 0.735. In Group 5, the most common group, the consistency was as high as 95.90% (117/122), while the consistency was low in the other groups due to the limited number of cases. The causes of this inconsistency, including clustered HER2 signals, coarse CEP17 signals and some section quality problems, were analysed. The developed AI model is a reliable tool for evaluating HER2 amplification statuses, especially for breast cancer in Group 5; additional cases from multiple centres could further improve the accuracy achieved for other groups.
Collapse
Affiliation(s)
- Tian Xue
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Heng Chang
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Min Ren
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Haochen Wang
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Yu Yang
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Boyang Wang
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Lei Lv
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Licheng Tang
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Chicheng Fu
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Qu Fang
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Chuan He
- Shanghai Aitrox Technology Corporation Limited, Shanghai, China
| | - Xiaoli Zhu
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China.
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Sajjadi E, Guerini-Rocco E, De Camilli E, Pala O, Mazzarol G, Venetis K, Ivanova M, Fusco N. Pathological identification of HER2-low breast cancer: Tips, tricks, and troubleshooting for the optimal test. Front Mol Biosci 2023; 10:1176309. [PMID: 37077201 PMCID: PMC10106673 DOI: 10.3389/fmolb.2023.1176309] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
The introduction of novel anti-HER2 antibody-drug conjugates (ADC) for the treatment of HER2-low breast cancers has transformed the traditional dichotomy of HER2 status to an expanded spectrum. However, the identification of HER2-low (i.e., immunohistochemistry (IHC) score 1 + or IHC score 2+, without gene amplification) tumors is challenged by methodological and analytical variables that might influence the sensitivity and reproducibility of HER2 testing. To open all possible therapeutic opportunities for HER2-low breast cancer patients the implementation of more accurate and reproducible testing strategies is mandatory. Here, we provide an overview of the existing barriers that may trouble HER2-low identification in breast cancer and discuss practical solutions that could enhance HER-low assessment.
Collapse
Affiliation(s)
- Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Oriana Pala
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- *Correspondence: Nicola Fusco,
| |
Collapse
|
11
|
Wu S, Yue M, Zhang J, Li X, Li Z, Zhang H, Wang X, Han X, Cai L, Shang J, Jia Z, Wang X, Li J, Liu Y. The Role of Artificial Intelligence in Accurate Interpretation of HER2 Immunohistochemical Scores 0 and 1+ in Breast Cancer. Mod Pathol 2023; 36:100054. [PMID: 36788100 DOI: 10.1016/j.modpat.2022.100054] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/28/2022] [Accepted: 11/20/2022] [Indexed: 01/11/2023]
Abstract
The new human epidermal growth factor receptor (HER)2-targeting antibody-drug conjugate offers the opportunity to treat patients with HER2-low breast cancer. Distinguishing HER2 immunohistochemical (IHC) scores of 0 and 1+ is not only critical but also challenging owing to HER2 heterogeneity and variability of observers. In this study, we aimed to increase the interpretation accuracy and consistency of HER2 IHC 0 and 1+ evaluation through assistance from an artificial intelligence (AI) algorithm. In addition, we examined the value of our AI algorithm in evaluating HER2 IHC scores in tumors with heterogeneity. AI-assisted interpretation consisted of AI algorithms and an augmenting reality module with a microscope. Fifteen pathologists (5 junior, 5 midlevel, and 5 senior) participated in this multi-institutional 2-round ring study that included 246 infiltrating duct carcinoma cases that were not otherwise specified. In round 1, pathologists analyzed 246 HER2 IHC slides by microscope without AI assistance. After a 2-week washout period, the pathologists read the same slides with AI algorithm assistance and rendered the definitive results by adjusting to the AI algorithm. The accuracy of interpretation accuracy with AI assistance (0.93 vs 0.80), thereby the evaluation precision of HER2 0 and the recall of HER2 1+. In addition, the AI algorithm improved the total consistency (intraclass correlation coefficient = 0.542-0.812), especially in HER2 1+ cases. In cases with heterogeneity, accuracy improved significantly (0.68 to 0.89) and to a similar level as in cases without heterogeneity (accuracy, 0.97). Both accuracy and consistency improved more for junior pathologists than those for the midlevel and senior pathologists. To the best of our knowledge, this is the first study to show that the accuracy and consistency of HER2 IHC 0 and 1+ evaluation and the accuracy of HER2 IHC evaluation in breast cancers with heterogeneity can be significantly improved using AI-assisted interpretation.
Collapse
Affiliation(s)
- Si Wu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Meng Yue
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jun Zhang
- Tencent AI Lab, Nanshan District, Tencent Binhai Building, Shenzhen, Guangdong, China
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, The Emory University School of Medicine, Atlanta, Georgia
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Huina Zhang
- Department of Pathology, University of Rochester Medical Center, Rochester, New York
| | - Xinran Wang
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao Han
- Tencent AI Lab, Nanshan District, Tencent Binhai Building, Shenzhen, Guangdong, China
| | - Lijing Cai
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiuyan Shang
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhanli Jia
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoxiao Wang
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinze Li
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yueping Liu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
12
|
Mitrea DM, Mittasch M, Gomes BF, Klein IA, Murcko MA. Modulating biomolecular condensates: a novel approach to drug discovery. Nat Rev Drug Discov 2022; 21:841-862. [PMID: 35974095 PMCID: PMC9380678 DOI: 10.1038/s41573-022-00505-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 12/12/2022]
Abstract
In the past decade, membraneless assemblies known as biomolecular condensates have been reported to play key roles in many cellular functions by compartmentalizing specific proteins and nucleic acids in subcellular environments with distinct properties. Furthermore, growing evidence supports the view that biomolecular condensates often form by phase separation, in which a single-phase system demixes into a two-phase system consisting of a condensed phase and a dilute phase of particular biomolecules. Emerging understanding of condensate function in normal and aberrant cellular states, and of the mechanisms of condensate formation, is providing new insights into human disease and revealing novel therapeutic opportunities. In this Perspective, we propose that such insights could enable a previously unexplored drug discovery approach based on identifying condensate-modifying therapeutics (c-mods), and we discuss the strategies, techniques and challenges involved.
Collapse
|
13
|
A Soft Label Deep Learning to Assist Breast Cancer Target Therapy and Thyroid Cancer Diagnosis. Cancers (Basel) 2022; 14:cancers14215312. [PMID: 36358732 PMCID: PMC9657740 DOI: 10.3390/cancers14215312] [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: 08/30/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
According to the World Health Organization Report 2022, cancer is the most common cause of death contributing to nearly one out of six deaths worldwide. Early cancer diagnosis and prognosis have become essential in reducing the mortality rate. On the other hand, cancer detection is a challenging task in cancer pathology. Trained pathologists can detect cancer, but their decisions are subjective to high intra- and inter-observer variability, which can lead to poor patient care owing to false-positive and false-negative results. In this study, we present a soft label fully convolutional network (SL-FCN) to assist in breast cancer target therapy and thyroid cancer diagnosis, using four datasets. To aid in breast cancer target therapy, the proposed method automatically segments human epidermal growth factor receptor 2 (HER2) amplification in fluorescence in situ hybridization (FISH) and dual in situ hybridization (DISH) images. To help in thyroid cancer diagnosis, the proposed method automatically segments papillary thyroid carcinoma (PTC) on Papanicolaou-stained fine needle aspiration and thin prep whole slide images (WSIs). In the evaluation of segmentation of HER2 amplification in FISH and DISH images, we compare the proposed method with thirteen deep learning approaches, including U-Net, U-Net with InceptionV5, Ensemble of U-Net with Inception-v4, Inception-Resnet-v2 encoder, and ResNet-34 encoder, SegNet, FCN, modified FCN, YOLOv5, CPN, SOLOv2, BCNet, and DeepLabv3+ with three different backbones, including MobileNet, ResNet, and Xception, on three clinical datasets, including two DISH datasets on two different magnification levels and a FISH dataset. The result on DISH breast dataset 1 shows that the proposed method achieves high accuracy of 87.77 ± 14.97%, recall of 91.20 ± 7.72%, and F1-score of 81.67 ± 17.76%, while, on DISH breast dataset 2, the proposed method achieves high accuracy of 94.64 ± 2.23%, recall of 83.78 ± 6.42%, and F1-score of 85.14 ± 6.61% and, on the FISH breast dataset, the proposed method achieves high accuracy of 93.54 ± 5.24%, recall of 83.52 ± 13.15%, and F1-score of 86.98 ± 9.85%, respectively. Furthermore, the proposed method outperforms most of the benchmark approaches by a significant margin (p <0.001). In evaluation of segmentation of PTC on Papanicolaou-stained WSIs, the proposed method is compared with three deep learning methods, including Modified FCN, U-Net, and SegNet. The experimental result demonstrates that the proposed method achieves high accuracy of 99.99 ± 0.01%, precision of 92.02 ± 16.6%, recall of 90.90 ± 14.25%, and F1-score of 89.82 ± 14.92% and significantly outperforms the baseline methods, including U-Net and FCN (p <0.001). With the high degree of accuracy, precision, and recall, the results show that the proposed method could be used in assisting breast cancer target therapy and thyroid cancer diagnosis with faster evaluation and minimizing human judgment errors.
Collapse
|
14
|
Hossain MS, Syeed MMM, Fatema K, Hossain MS, Uddin MF. Singular Nuclei Segmentation for Automatic HER2 Quantification Using CISH Whole Slide Images. SENSORS (BASEL, SWITZERLAND) 2022; 22:7361. [PMID: 36236459 PMCID: PMC9571354 DOI: 10.3390/s22197361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) quantification is performed routinely for all breast cancer patients to determine their suitability for HER2-targeted therapy. Fluorescence in situ hybridization (FISH) and chromogenic in situ hybridization (CISH) are the US Food and Drug Administration (FDA) approved tests for HER2 quantification in which at least 20 cancer-affected singular nuclei are quantified for HER2 grading. CISH is more advantageous than FISH for cost, time and practical usability. In clinical practice, nuclei suitable for HER2 quantification are selected manually by pathologists which is time-consuming and laborious. Previously, a method was proposed for automatic HER2 quantification using a support vector machine (SVM) to detect suitable singular nuclei from CISH slides. However, the SVM-based method occasionally failed to detect singular nuclei resulting in inaccurate results. Therefore, it is necessary to develop a robust nuclei detection method for reliable automatic HER2 quantification. In this paper, we propose a robust U-net-based singular nuclei detection method with complementary color correction and deconvolution adapted for accurate HER2 grading using CISH whole slide images (WSIs). The efficacy of the proposed method was demonstrated for automatic HER2 quantification during a comparison with the SVM-based approach.
Collapse
Affiliation(s)
- Md Shakhawat Hossain
- Department of CS, American International University-Bangladesh, Dhaka 1229, Bangladesh
- RIoT Research Center, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - M. M. Mahbubul Syeed
- RIoT Research Center, Independent University, Bangladesh, Dhaka 1229, Bangladesh
- Department of CSE, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - Kaniz Fatema
- RIoT Research Center, Independent University, Bangladesh, Dhaka 1229, Bangladesh
- Department of CSE, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - Md Sakir Hossain
- Department of CS, American International University-Bangladesh, Dhaka 1229, Bangladesh
| | - Mohammad Faisal Uddin
- RIoT Research Center, Independent University, Bangladesh, Dhaka 1229, Bangladesh
- Department of CSE, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| |
Collapse
|
15
|
Han Z, Lan J, Wang T, Hu Z, Huang Y, Deng Y, Zhang H, Wang J, Chen M, Jiang H, Lee RG, Gao Q, Du M, Tong T, Chen G. A Deep Learning Quantification Algorithm for HER2 Scoring of Gastric Cancer. Front Neurosci 2022; 16:877229. [PMID: 35706692 PMCID: PMC9190202 DOI: 10.3389/fnins.2022.877229] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer is the third most common cause of cancer-related death in the world. Human epidermal growth factor receptor 2 (HER2) positive is an important subtype of gastric cancer, which can provide significant diagnostic information for gastric cancer pathologists. However, pathologists usually use a semi-quantitative assessment method to assign HER2 scores for gastric cancer by repeatedly comparing hematoxylin and eosin (H&E) whole slide images (WSIs) with their HER2 immunohistochemical WSIs one by one under the microscope. It is a repetitive, tedious, and highly subjective process. Additionally, WSIs have billions of pixels in an image, which poses computational challenges to Computer-Aided Diagnosis (CAD) systems. This study proposed a deep learning algorithm for HER2 quantification evaluation of gastric cancer. Different from other studies that use convolutional neural networks for extracting feature maps or pre-processing on WSIs, we proposed a novel automatic HER2 scoring framework in this study. In order to accelerate the computational process, we proposed to use the re-parameterization scheme to separate the training model from the deployment model, which significantly speedup the inference process. To the best of our knowledge, this is the first study to provide a deep learning quantification algorithm for HER2 scoring of gastric cancer to assist the pathologist's diagnosis. Experiment results have demonstrated the effectiveness of our proposed method with an accuracy of 0.94 for the HER2 scoring prediction.
Collapse
Affiliation(s)
- Zixin Han
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Junlin Lan
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Tao Wang
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Ziwei Hu
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Yuxiu Huang
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Yanglin Deng
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
| | - Hejun Zhang
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianchao Wang
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Musheng Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Haiyan Jiang
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
- College of Electrical Engineering and Automation, Fuzhou University, Fuzhou, China
| | - Ren-Guey Lee
- Department of Electronic Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Qinquan Gao
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
- Imperial Vision Technology, Fuzhou, China
| | - Ming Du
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
| | - Tong Tong
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, China
- Fujian Key Lab of Medical Instrumentation & Pharmaceutical Technology, Fuzhou University, Fuzhou, China
- Imperial Vision Technology, Fuzhou, China
| | - Gang Chen
- College of Electrical Engineering and Automation, Fuzhou University, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicin, Fuzhou, China
| |
Collapse
|
16
|
Aguilera A, Pezoa R, Rodríguez-Delherbe A. A novel ensemble feature selection method for pixel-level segmentation of HER2 overexpression. COMPLEX INTELL SYST 2022. [DOI: 10.1007/s40747-022-00774-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AbstractClassifying histopathology images on a pixel-level requires sets of features able to capture the complex characteristics of the images, like the irregular cell morphology and the color heterogeneity on the tissue aspect. In this context, feature selection becomes a crucial step in the classification process such that it reduces model complexity and computational costs, avoids overfitting, and thereby it improves the model performance. In this study, we propose a new ensemble feature selection method by combining a set of base selectors, classifiers, and rank aggregation methods, aiming to determine from any initial set of handcrafted features, a smaller set of relevant color and texture pixel-level features, subsequently used for segmenting HER2 overexpression on a pixel-level, in breast cancer tissue images. We have been able to significantly reduce the set of initial features, using the proposed ensemble feature selection method. The best results are obtained using $$\chi ^2$$
χ
2
, Random Forest, and Runoff as the based selector, classifier, and aggregation method, respectively. The classification performance of the best model trained on the selected features set results in 0.939 recall, 0.866 specificity, 0.903 accuracy, 0.875 precision, and 0.906 F1-score.
Collapse
|
17
|
Validation of HER2 Status in Whole Genome Sequencing Data of Breast Cancers with the Ploidy-Corrected Copy Number Approach. Mol Diagn Ther 2021; 26:105-116. [PMID: 34932189 PMCID: PMC8766398 DOI: 10.1007/s40291-021-00571-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Human epidermal growth factor receptor 2 (HER2) protein overexpression is one of the most significant biomarkers for breast cancer diagnostics, treatment prediction, and prognostics. The high accessibility of HER2 inhibitors in routine clinical practice directly translates into the diagnostic need for precise and robust marker identification. Even though multigene next-generation sequencing methodologies have slowly taken over the field of single-biomarker molecular tests, the copy number alterations such as amplification of the HER2-coding ERBB2 gene are hard to validate on next-generation sequencing platforms as they are characterized by chromosomal structural heterogeneity, polysomy, and genomic context of ploidy. In our study, we tested the approach of using whole genome sequencing instead of next-generation sequencing panels to determine HER2 status in the clinical set-up. METHODS We used a large dataset of 876 patients with breast cancer whole genomes with curated clinical data and an additional set of 551 patients' external genomic data. We used the decision-tree-based algorithm for optimization of the diagnostic tool for HER2 status assessment by whole genome sequencing. RESULTS The most efficient approach to assess HER2 status in whole genome sequencing data was the ploidy-corrected copy number, utilizing ERBB2 copy number and mean tumor ploidy. The classifier achieved sensitivity of 91.18% and specificity of 98.69% on the internal validation dataset and 89.86% and 96.06% on the external data, which is similar to other next-generation sequencing methods, currently tested in the clinic. CONCLUSIONS We provide evidence that the HER2 status may be reliably determined by whole genome sequencing and is applicable across different laboratory protocols and pipelines. We suggest using the ploidy-corrected copy number for diagnostic purposes.
Collapse
|
18
|
Hinata M, Ushiku T. Detecting immunotherapy-sensitive subtype in gastric cancer using histologic image-based deep learning. Sci Rep 2021; 11:22636. [PMID: 34811485 PMCID: PMC8608814 DOI: 10.1038/s41598-021-02168-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/09/2021] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy is widely used but effective only in a subset of gastric cancers. Epstein–Barr virus (EBV)-positive and microsatellite instability (MSI) / mismatch repair deficient (dMMR) tumors have been reported to be highly responsive to ICIs. However, detecting these subtypes requires costly techniques, such as immunohistochemistry and molecular testing. In the present study, we constructed a histology-based deep learning model that aimed to screen this immunotherapy-sensitive subgroup efficiently. We processed whole slide images of 408 cases of gastric adenocarcinoma, including 108 EBV, 58 MSI/dMMR, and 242 other subtypes. Many images generated by data augmentation of the learning set were used for training convolutional neural networks to establish an automatic detection platform for EBV and MSI/dMMR subtypes, and the test sets of images were used to verify the learning outcome. Our model detected the subgroup (EBV + MSI/dMMR tumors) with high accuracy in test cases with an area under the curve of 0.947 (0.901–0.992). This result was slightly better than when EBV and MSI/dMMR tumors were detected separately. In an external validation cohort including 244 gastric cancers from The Cancer Genome Atlas database, our model showed a favorable result for detecting the “EBV + MSI/dMMR” subgroup with an AUC of 0.870 (0.809–0.931). In addition, a visualization of the trained neural network highlighted intraepithelial lymphocytosis as the ground for prediction, suggesting that this feature is a discriminative characteristic shared by EBV and MSI/dMMR tumors. Histology-based deep learning models are expected to be used for detecting EBV and MSI/dMMR gastric cancers as economical and less time-consuming alternatives, which may help to effectively stratify patients who respond to ICIs.
Collapse
Affiliation(s)
- Munetoshi Hinata
- Department of Pathology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tetsuo Ushiku
- Department of Pathology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
19
|
Xing F, Zhang X, Cornish TC. Artificial intelligence for pathology. Artif Intell Med 2021. [DOI: 10.1016/b978-0-12-821259-2.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
Pandey I, Misra V, Pandey AT, Ramteke PW, Agrawal R. Artificial intelligence technologies empowering identification of novel diagnostic molecular markers in gastric cancer. INDIAN J PATHOL MICR 2021; 64:S63-S68. [PMID: 34135140 DOI: 10.4103/ijpm.ijpm_950_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In recent clinical practice the molecular diagnostics have been significantly empowered and upgraded by the use of Artificial Intelligence and its assisted technologies. The use of Machine leaning and Deep Learning Neural network architectures have brought in a new dimension in clinical oncological research and development. These algorithm based software system with enhanced digital image analysis have emerged into a new branch of digital pathology and contributed immensely towards precision medicine and personal diagnostics. In India, gastric cancer is one of the most common cancers in males as well as in females. Various molecular biomarkers are associated with gastric cancer development and progression of which HER2 protein, a transmembrane tyrosine kinase (TK) receptor of epidermal growth factor receptors (EGFRs) family is of prime importance. The EGF receptor expression in gastric cancer is linked with its prognostics and theragnostics. These expressions are assessed by immunohistochemistry (IHC) and molecular techniques such as Fluorescence in-situ hybridization (FISH), as per recommendations for HER2 targeted immunotherapy. These have motivated the software giants like Google Inc. to produce innovative state of art technologies mimicking human traits such as learning and problem solving skill sets. This field is still under development and is slowly evolving and capturing global importance in recent times. A literature search on PubMed was performed to access updated information for this manuscript.
Collapse
Affiliation(s)
- Ishan Pandey
- Department of Pathology, MotiLal Nehru Medical College, Prayagraj, India
| | - Vatsala Misra
- Department of Pathology, MotiLal Nehru Medical College, Prayagraj, India
| | | | | | - Ranjan Agrawal
- Department of Pathology, Rohilkhand Medical College and Hospital, Bareilly, Uttar Pradesh, India
| |
Collapse
|
21
|
Small but powerful: the promising role of small specimens for biomarker testing. J Am Soc Cytopathol 2020; 9:450-460. [PMID: 32507626 DOI: 10.1016/j.jasc.2020.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/22/2022]
Abstract
Emphasis on the use of small specimens for biomarker testing to provide prognostic and predictive information for guiding clinical management for patients with advanced-stage cancer has been increasing. These biomarker tests include molecular analysis, cytogenetic tests, and immunohistochemical assays. Owing to the limited nature of the cellular material procured in these small specimens, which are collected using minimally invasive techniques (ie, fine needle aspiration and core needle biopsy), pathologists have been required to triage these samples judiciously and provide the clinically relevant genomic information required for patient care. Awareness of the advantages and limitations of these specimen preparations and the specific preanalytic requirements for the testing methods will help pathologists to develop optimal strategies to maximize the chances of effectively using these samples for comprehensive diagnostic and relevant biomarker testing.
Collapse
|
22
|
Deep Learning-based Inaccuracy Compensation in Reconstruction of High Resolution XCT Data. Sci Rep 2020; 10:7682. [PMID: 32376852 PMCID: PMC7203197 DOI: 10.1038/s41598-020-64733-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 04/17/2020] [Indexed: 11/08/2022] Open
Abstract
While X-ray computed tomography (XCT) is pushed further into the micro- and nanoscale, the limitations of various tool components and object motion become more apparent. For high-resolution XCT, it is necessary but practically difficult to align these tool components with sub-micron precision. The aim is to develop a novel reconstruction methodology that considers unavoidable misalignment and object motion during the data acquisition in order to obtain high-quality three-dimensional images and that is applicable for data recovery from incomplete datasets. A reconstruction software empowered by sophisticated correction modules that autonomously estimates and compensates artefacts using gradient descent and deep learning algorithms has been developed and applied. For motion estimation, a novel computer vision methodology coupled with a deep convolutional neural network approach provides estimates for the object motion by tracking features throughout the adjacent projections. The model is trained using the forward projections of simulated phantoms that consist of several simple geometrical features such as sphere, triangle and rectangular. The feature maps extracted by a neural network are used to detect and to classify features done by a support vector machine. For missing data recovery, a novel deep convolutional neural network is used to infer high-quality reconstruction data from incomplete sets of projections. The forward and back projections of simulated geometric shapes from a range of angular ranges are used to train the model. The model is able to learn the angular dependency based on a limited angle coverage and to propose a new set of projections to suppress artefacts. High-quality three-dimensional images demonstrate that it is possible to effectively suppress artefacts caused by thermomechanical instability of tool components and objects resulting in motion, by center of rotation misalignment and by inaccuracy in the detector position without additional computational efforts. Data recovery from incomplete sets of projections result in directly corrected projections instead of suppressing artefacts in the final reconstructed images. The proposed methodology has been proven and is demonstrated for a ball bearing sample. The reconstruction results are compared to prior corrections and benchmarked with a commercially available reconstruction software. Compared to conventional approaches in XCT imaging and data analysis, the proposed methodology for the generation of high-quality three-dimensional X-ray images is fully autonomous. The methodology presented here has been proven for high-resolution micro-XCT and nano-XCT, however, is applicable for all length scales.
Collapse
|
23
|
Han S, Park S, An J, Yang JY, Chung JW, Kim YJ, Kim KO, Park DK, Kwon KA, Lee WK, Nam S, Kim JH. HER2 as a potential biomarker of lymph node metastasis in undifferentiated early gastric cancer. Sci Rep 2020; 10:5270. [PMID: 32210254 PMCID: PMC7093413 DOI: 10.1038/s41598-020-61567-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 02/18/2020] [Indexed: 01/04/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is implicated in several cancers, including gastric cancer. However, limited data are available regarding its clinical significance in early gastric cancer (EGC). We evaluated the clinical significance of HER2 overexpression in patients with EGC. We retrospectively reviewed 727 patients who underwent surgical treatment for EGC between October 2010 and August 2017. HER2 expression was analysed in 680 EGC cases by immunohistochemistry and classified as negative (0 and 1+), equivocal (2+), or positive [overexpression (3+)]. Among patients with differentiated EGC, the number of patients with HER2 overexpression was not significantly different from that of HER2-negative patients in terms of age, sex, tumour size, location, gross type, depth of invasion, presence of lymphovascular invasion (LVI), and presence of lymph node metastasis (LNM). However, in patients with undifferentiated EGC, HER2 overexpression was significantly correlated with LVI and presence of LNM compared with HER2-negative patients. Multivariate analysis indicated HER2 overexpression as a good predictive marker of LNM in patients with undifferentiated EGC. HER2 expression is associated with LNM in undifferentiated EGC. Therefore, the importance of HER2 overexpression in EGC should not be overlooked, and further studies are needed to identify its clinical significance.
Collapse
Affiliation(s)
- Sanghoon Han
- Department of Internal Medicine, Jeju National University School of Medicine, Jeju, Korea
| | - Sungjin Park
- College of Medicine, Gachon University, Incheon, Republic of Korea.,Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Jungsuk An
- Department of Pathology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Jun-Young Yang
- Department of Surgery, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Jun-Won Chung
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Yoon Jae Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Kyoung Oh Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Dong Kyun Park
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Kwang An Kwon
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Woon Kee Lee
- Department of Surgery, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea
| | - Seungyoon Nam
- College of Medicine, Gachon University, Incheon, Republic of Korea. .,Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea. .,Department of Life Sciences, Gachon University, Seongnam, Republic of Korea. .,Gachon Advanced Institute of Health Sciences & Technology, Gachon University, Incheon, Republic of Korea.
| | - Jung Ho Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Republic of Korea. .,Gachon Medical Research Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea.
| |
Collapse
|
24
|
Deep-UV excitation fluorescence microscopy for detection of lymph node metastasis using deep neural network. Sci Rep 2019; 9:16912. [PMID: 31729459 PMCID: PMC6858352 DOI: 10.1038/s41598-019-53405-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Deep-UV (DUV) excitation fluorescence microscopy has potential to provide rapid diagnosis with simple technique comparing to conventional histopathology based on hematoxylin and eosin (H&E) staining. We established a fluorescent staining protocol for DUV excitation fluorescence imaging that has enabled clear discrimination of nucleoplasm, nucleolus, and cytoplasm. Fluorescence images of metastasis-positive/-negative lymph nodes of gastric cancer patients were used for patch-based training with a deep neural network (DNN) based on Inception-v3 architecture. The performance on small patches of the fluorescence images was comparable with that of H&E images. Gradient-weighted class activation mapping analysis revealed the areas where the trained model identified metastatic lesions in the images containing cancer cells. We extended the method to large-size image analysis enabling accurate detection of metastatic lesions. We discuss usefulness of DUV excitation fluorescence imaging with the aid of DNN analysis, which is promising for assisting pathologists in assessment of lymph node metastasis.
Collapse
|