1
|
Bharath E, Raja RV, Kalaivanan K, Deshpande V. Accurate colorectal cancer detection using a random hinge exponential distribution coupled attention network on pathological images. Abdom Radiol (NY) 2025:10.1007/s00261-024-04770-2. [PMID: 39779530 DOI: 10.1007/s00261-024-04770-2] [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: 10/30/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025]
Abstract
Colorectal cancer (CRC) is one of the most common and deadly forms of cancer worldwide, necessitating accurate and early detection to improve treatment outcomes. Traditional diagnostic methods often rely on manual examination of pathological images, which can be time-consuming and prone to human error. This study presents an advanced approach for colorectal cancer detection using a Random Hinge Exponential Distribution coupled Attention Network (RHED-CANet) on pathological images. The input dataset is sourced from the TCGA-CRC-DX cohort and the CRC dataset, both widely recognized for their comprehensive coverage of colorectal cancer cases. Pre-processing and feature extraction are performed using a Modified Square Root Sage-Husa Adaptive Kalman Filter combined with a Spike-Driven Transformer, enhancing noise reduction and feature clarity. Segmentation is achieved through an EfficientNetV2L Inception Transformer, ensuring precise delineation of cancerous regions. The final classification utilizes the RHED-CANet, a network tailored to handle the complexities of pathological data with high accuracy. This methodology achieved remarkable results, with an accuracy of 99.9% and a precision of 99.7%. These performance metrics underscore the method's ability to minimize false positives and enhance diagnostic accuracy. The proposed approach offers significant advantages, including a reduction in diagnostic time and a substantial improvement in detection accuracy, making it a promising tool for clinical applications. Despite its excellent accuracy, the suggested RHED-CANet technique has drawbacks, such as overfitting the TCGA-CRC-DX and CRC datasets by reducing generalizability on other datasets comprising other cancer types or image qualities. The actual application of the techniques in real-time clinical applications may be hampered by this computational load, especially in settings with limited resources, and the model's potential computational complexity due to multiple advanced processing steps. Additionally, the efficiency of training may be impacted by biased inputs, particularly for minor CRC subtypes.
Collapse
Affiliation(s)
- E Bharath
- Department of Artificial Intelligence and Data Science, CK College of Engineering and Technology, Cuddalore, Tamil Nadu, India.
| | - R Vimal Raja
- Department of Computer Science and Engineering, CK College of Engineering and Technology, Cuddalore, Tamil Nadu, India
| | - K Kalaivanan
- Department of Computer Science and Engineering, SRM Institute of Science and Technology, Tiruchirapalli, Tamil Nadu, India
| | - Vivek Deshpande
- Department of Computer Engineering, Vishwakarma Institute of Information Technology, Pune, India
| |
Collapse
|
2
|
Chen W, Zheng K, Yuan W, Jia Z, Wu Y, Duan X, Yang W, Wen Z, Zhong L, Liu X. A CT-based deep learning for segmenting tumors and predicting microsatellite instability in patients with colorectal cancers: a multicenter cohort study. LA RADIOLOGIA MEDICA 2024:10.1007/s11547-024-01909-5. [PMID: 39586941 DOI: 10.1007/s11547-024-01909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE To develop and validate deep learning (DL) models using preoperative contrast-enhanced CT images for tumor auto-segmentation and microsatellite instability (MSI) prediction in colorectal cancer (CRC). MATERIALS AND METHODS Patients with CRC who underwent surgery or biopsy between January 2018 and April 2023 were retrospectively enrolled. Mismatch repair protein expression was determined via immunohistochemistry or fluorescence multiplex polymerase chain reaction-capillary electrophoresis. Manually delineated tumor contours using arterial and venous phase CT images by three abdominal radiologists are served as ground truth. Tumor auto-segmentation used nnU-Net. MSI prediction employed ViT or convolutional neural networks models, trained and validated with arterial and venous phase images (image model) or combined clinical-pathological factors (combined model). The segmentation model was evaluated using patch coverage ratio, Dice coefficient, recall, precision, and F1-score. The predictive models' efficacy was assessed using areas under the curves and decision curve analysis. RESULTS Overall, 2180 patients (median age: 61 years ± 17 [SD]; 1285 males) were divided into training (n = 1159), validation (n = 289), and independent external test (n = 732) groups. High-level MSI status was present in 435 patients (20%). In the external test set, the segmentation model performed well in the arterial phase, with patch coverage ratio, Dice coefficient, recall, precision, and F1-score values of 0.87, 0.71, 0.72, 0.74, and 0.71, respectively. For MSI prediction, the combined models outperformed the clinical model (AUC = 0.83 and 0.82 vs 0.67, p < 0.001) and two image models (AUC = 0.75 and 0.77, p < 0.001). Decision curve analysis confirmed the higher net benefit of the combined model compared to the other models across probability thresholds ranging from 0.1 to 0.45. CONCLUSION DL enhances tumor segmentation efficiency and, when integrated with contrast-enhanced CT and clinicopathological factors, exhibits good diagnostic performance in predicting MSI in CRC.
Collapse
Affiliation(s)
- Weicui Chen
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Kaiyi Zheng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Wenjing Yuan
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Ziqi Jia
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Yuankui Wu
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaohui Duan
- Radiology Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, China
| | - Wei Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhibo Wen
- Radiology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Liming Zhong
- School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Xian Liu
- Radiology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
3
|
Xiaojian Y, Zhanbo Q, Jian C, Zefeng W, Jian L, Jin L, Yuefen P, Shuwen H. Deep learning application in prediction of cancer molecular alterations based on pathological images: a bibliographic analysis via CiteSpace. J Cancer Res Clin Oncol 2024; 150:467. [PMID: 39422817 PMCID: PMC11489169 DOI: 10.1007/s00432-024-05992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The advancements in artificial intelligence (AI) technology for image recognition were propelling molecular pathology research into a new era. OBJECTIVE To summarize the hot spots and research trends in the field of molecular pathology image recognition. METHODS Relevant articles from January 1st, 2010, to August 25th, 2023, were retrieved from the Web of Science Core Collection. Subsequently, CiteSpace was employed for bibliometric and visual analysis, generating diverse network diagrams illustrating keywords, highly cited references, hot topics, and research trends. RESULTS A total of 110 relevant articles were extracted from a pool of 10,205 articles. The overall publication count exhibited a rising trend each year. The leading contributors in terms of institutions, countries, and authors were Maastricht University (11 articles), the United States (38 articles), and Kather Jacob Nicholas (9 articles), respectively. Half of the top ten research institutions, based on publication volume, were affiliated with Germany. The most frequently cited article was authored by Nicolas Coudray et al. accumulating 703 citations. The keyword "Deep learning" had the highest frequency in 2019. Notably, the highlighted keywords from 2022 to 2023 included "microsatellite instability", and there were 21 articles focusing on utilizing algorithms to recognize microsatellite instability (MSI) in colorectal cancer (CRC) pathological images. CONCLUSION The use of DL is expected to provide a new strategy to effectively solve the current problem of time-consuming and expensive molecular pathology detection. Therefore, further research is needed to address issues, such as data quality and standardization, model interpretability, and resource and infrastructure requirements.
Collapse
Affiliation(s)
- Yu Xiaojian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Wang Zefeng
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jin
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Pan Yuefen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
4
|
Nowak M, Jabbar F, Rodewald AK, Gneo L, Tomasevic T, Harkin A, Iveson T, Saunders M, Kerr R, Oein K, Maka N, Hay J, Edwards J, Tomlinson I, Sansom O, Kelly C, Pezzella F, Kerr D, Easton A, Domingo E, Koelzer VH, Church DN. Single-cell AI-based detection and prognostic and predictive value of DNA mismatch repair deficiency in colorectal cancer. Cell Rep Med 2024; 5:101727. [PMID: 39293403 PMCID: PMC11525017 DOI: 10.1016/j.xcrm.2024.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/16/2024] [Accepted: 08/15/2024] [Indexed: 09/20/2024]
Abstract
Testing for DNA mismatch repair deficiency (MMRd) is recommended for all colorectal cancers (CRCs). Automating this would enable precision medicine, particularly if providing information on etiology not captured by deep learning (DL) methods. We present AIMMeR, an AI-based method for determination of mismatch repair (MMR) protein expression at a single-cell level in routine pathology samples. AIMMeR shows an area under the receiver-operator curve (AUROC) of 0.98, and specificity of ≥75% at 98% sensitivity against pathologist ground truth in stage II/III in two trial cohorts, with positive predictive value of ≥98% for the commonest pattern of somatic MMRd. Lower agreement with microsatellite instability (MSI) testing (AUROC 0.86) reflects discordance between MMR and MSI PCR rather than AIMMeR misclassification. Analysis of the SCOT trial confirms MMRd prognostic value in oxaliplatin-treated patients; while MMRd does not predict differential benefit of chemotherapy duration, it correlates with difference in relapse by regimen (PInteraction = 0.04). AIMMeR may help reduce pathologist workload and streamline diagnostics in CRC.
Collapse
Affiliation(s)
- Marta Nowak
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland
| | - Faiz Jabbar
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ann-Katrin Rodewald
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland
| | - Luciana Gneo
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Tijana Tomasevic
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrea Harkin
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | - Tim Iveson
- University of Southampton, Southampton, UK
| | | | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - Karin Oein
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Noori Maka
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Jennifer Hay
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, UK
| | - Owen Sansom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, UK
| | - Caroline Kelly
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, UK
| | | | - David Kerr
- Nuffield Department of Clinical and Laboratory Sciences, University of Oxford, Oxford, UK
| | | | - Enric Domingo
- Department of Oncology, University of Oxford, Oxford, UK
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, Zurich, Zurich, Switzerland; Department of Oncology, University of Oxford, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Oxford, UK; Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - David N Church
- Cancer Genomics and Immunology Group, The Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
5
|
Whangbo J, Lee YS, Kim YJ, Kim J, Kim KG. Predicting Mismatch Repair Deficiency Status in Endometrial Cancer through Multi-Resolution Ensemble Learning in Digital Pathology. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1674-1682. [PMID: 38378964 PMCID: PMC11300772 DOI: 10.1007/s10278-024-00997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024]
Abstract
For molecular classification of endometrial carcinoma, testing for mismatch repair (MMR) status is becoming a routine process. Mismatch repair deficiency (MMR-D) is caused by loss of expression in one or more of the 4 major MMR proteins: MLH1, MSH2, MSH6, PHS2. Over 30% of patients with endometrial cancer have MMR-D. Determining the MMR status holds significance as individuals with MMR-D are potential candidates for immunotherapy. Pathological whole slide image (WSI) of endometrial cancer with immunohistochemistry results of MMR proteins were gathered. Color normalization was applied to the tiles using a CycleGAN-based network. The WSI was divided into tiles at three different magnifications (2.5 × , 5 × , and 10 ×). Three distinct networks of the same architecture were employed to include features from all three magnification levels and were stacked for ensemble learning. Three architectures, InceptionResNetV2, EfficientNetB2, and EfficientNetB3 were employed and subjected to comparison. The per-tile results were gathered to classify MMR status in the WSI, and prediction accuracy was evaluated using the following performance metrics: AUC, accuracy, sensitivity, and specificity. The EfficientNetB2 was able to make predictions with an AUC of 0.821, highest among the three architectures, and an overall AUC range of 0.767 - 0.821 was reported across the three architectures. In summary, our study successfully predicted MMR classification from pathological WSIs in endometrial cancer through a multi-resolution ensemble learning approach, which holds the potential to facilitate swift decisions on tailored treatment, such as immunotherapy, in clinical settings.
Collapse
Affiliation(s)
- Jongwook Whangbo
- Department of Computer Science, Wesleyan University, Middletown, Connecticut, USA
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Young Seop Lee
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Young Jae Kim
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
| | - Jisup Kim
- Department of Pathology, Gil Medical Center, Gachon University College of Medicine, 38-13, Dokjeom-Ro 3Beon-Gil, Namdong-Gu, Incheon, Republic of Korea.
| | - Kwang Gi Kim
- Medical Devices R&D Center, Gachon University Gil Hospital, Incheon, Republic of Korea.
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea.
- Department of Biomedical Engineering, College of Health Science, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
6
|
Gustav M, Reitsam NG, Carrero ZI, Loeffler CML, van Treeck M, Yuan T, West NP, Quirke P, Brinker TJ, Brenner H, Favre L, Märkl B, Stenzinger A, Brobeil A, Hoffmeister M, Calderaro J, Pujals A, Kather JN. Deep learning for dual detection of microsatellite instability and POLE mutations in colorectal cancer histopathology. NPJ Precis Oncol 2024; 8:115. [PMID: 38783059 PMCID: PMC11116442 DOI: 10.1038/s41698-024-00592-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/14/2024] [Indexed: 05/25/2024] Open
Abstract
In the spectrum of colorectal tumors, microsatellite-stable (MSS) tumors with DNA polymerase ε (POLE) mutations exhibit a hypermutated profile, holding the potential to respond to immunotherapy similarly to their microsatellite-instable (MSI) counterparts. Yet, due to their rarity and the associated testing costs, systematic screening for these mutations is not commonly pursued. Notably, the histopathological phenotype resulting from POLE mutations is theorized to resemble that of MSI. This resemblance not only could facilitate their detection by a transformer-based Deep Learning (DL) system trained on MSI pathology slides, but also indicates the possibility for MSS patients with POLE mutations to access enhanced treatment options, which might otherwise be overlooked. To harness this potential, we trained a Deep Learning classifier on a large dataset with the ground truth for microsatellite status and subsequently validated its capabilities for MSI and POLE detection across three external cohorts. Our model accurately identified MSI status in both the internal and external resection cohorts using pathology images alone. Notably, with a classification threshold of 0.5, over 75% of POLE driver mutant patients in the external resection cohorts were flagged as "positive" by a DL system trained on MSI status. In a clinical setting, deploying this DL model as a preliminary screening tool could facilitate the efficient identification of clinically relevant MSI and POLE mutations in colorectal tumors, in one go.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Titus J Brinker
- Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Loëtitia Favre
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Anaïs Pujals
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
7
|
Lo CM, Jiang JK, Lin CC. Detecting microsatellite instability in colorectal cancer using Transformer-based colonoscopy image classification and retrieval. PLoS One 2024; 19:e0292277. [PMID: 38271352 PMCID: PMC10810505 DOI: 10.1371/journal.pone.0292277] [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: 03/25/2023] [Accepted: 09/15/2023] [Indexed: 01/27/2024] Open
Abstract
Colorectal cancer (CRC) is a major global health concern, with microsatellite instability-high (MSI-H) being a defining characteristic of hereditary nonpolyposis colorectal cancer syndrome and affecting 15% of sporadic CRCs. Tumors with MSI-H have unique features and better prognosis compared to MSI-L and microsatellite stable (MSS) tumors. This study proposed establishing a MSI prediction model using more available and low-cost colonoscopy images instead of histopathology. The experiment utilized a database of 427 MSI-H and 1590 MSS colonoscopy images and vision Transformer (ViT) with different feature training approaches to establish the MSI prediction model. The accuracy of combining pre-trained ViT features was 84% with an area under the receiver operating characteristic curve of 0.86, which was better than that of DenseNet201 (80%, 0.80) in the experiment with support vector machine. The content-based image retrieval (CBIR) approach showed that ViT features can obtain a mean average precision of 0.81 compared to 0.79 of DenseNet201. ViT reduced the issues that occur in convolutional neural networks, including limited receptive field and gradient disappearance, and may be better at interpreting diagnostic information around tumors and surrounding tissues. By using CBIR, the presentation of similar images with the same MSI status would provide more convincing deep learning suggestions for clinical use.
Collapse
Affiliation(s)
- Chung-Ming Lo
- Graduate Institute of Library, Information and Archival Studies, National Chengchi University, Taipei, Taiwan
| | - Jeng-Kai Jiang
- Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chi Lin
- Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
8
|
Guitton T, Allaume P, Rabilloud N, Rioux-Leclercq N, Henno S, Turlin B, Galibert-Anne MD, Lièvre A, Lespagnol A, Pécot T, Kammerer-Jacquet SF. Artificial Intelligence in Predicting Microsatellite Instability and KRAS, BRAF Mutations from Whole-Slide Images in Colorectal Cancer: A Systematic Review. Diagnostics (Basel) 2023; 14:99. [PMID: 38201408 PMCID: PMC10795725 DOI: 10.3390/diagnostics14010099] [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: 11/22/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Mismatch repair deficiency (d-MMR)/microsatellite instability (MSI), KRAS, and BRAF mutational status are crucial for treating advanced colorectal cancer patients. Traditional methods like immunohistochemistry or polymerase chain reaction (PCR) can be challenged by artificial intelligence (AI) based on whole slide images (WSI) to predict tumor status. In this systematic review, we evaluated the role of AI in predicting MSI status, KRAS, and BRAF mutations in colorectal cancer. Studies published in PubMed up to June 2023 were included (n = 17), and we reported the risk of bias and the performance for each study. Some studies were impacted by the reduced number of slides included in the data set and the lack of external validation cohorts. Deep learning models for the d-MMR/MSI status showed a good performance in training cohorts (mean AUC = 0.89, [0.74-0.97]) but slightly less than expected in the validation cohort when available (mean AUC = 0.82, [0.63-0.98]). Contrary to the MSI status, the prediction of KRAS and BRAF mutations was less explored with a less robust methodology. The performance was lower, with a maximum of 0.77 in the training cohort, 0.58 in the validation cohort for KRAS, and 0.82 AUC in the training cohort for BRAF.
Collapse
Affiliation(s)
- Theo Guitton
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
| | - Pierre Allaume
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
| | - Noémie Rabilloud
- Impact TEAM, Laboratoire Traitement du Signal et de l’Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, CEDEX 09, 35033 Rennes, France
| | - Nathalie Rioux-Leclercq
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
| | - Sébastien Henno
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
| | - Bruno Turlin
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
| | - Marie-Dominique Galibert-Anne
- Department of Molecular Genetics and Medical Genomics CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (M.-D.G.-A.); (A.L.)
| | - Astrid Lièvre
- Department of Gastro-Entrology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France;
| | - Alexandra Lespagnol
- Department of Molecular Genetics and Medical Genomics CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (M.-D.G.-A.); (A.L.)
| | - Thierry Pécot
- Facility for Artificial Intelligence and Image Analysis (FAIIA), Biosit UAR 3480 CNRS-US18 INSERM, Rennes University, 2 Avenue du Professeur Léon Bernard, 35042 Rennes, France
| | - Solène-Florence Kammerer-Jacquet
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 Rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France; (P.A.); (N.R.-L.); (S.-F.K.-J.)
- Impact TEAM, Laboratoire Traitement du Signal et de l’Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, CEDEX 09, 35033 Rennes, France
| |
Collapse
|
9
|
Jang HJ, Go JH, Kim Y, Lee SH. Deep Learning for the Pathologic Diagnosis of Hepatocellular Carcinoma, Cholangiocarcinoma, and Metastatic Colorectal Cancer. Cancers (Basel) 2023; 15:5389. [PMID: 38001649 PMCID: PMC10670046 DOI: 10.3390/cancers15225389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Diagnosing primary liver cancers, particularly hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC), is a challenging and labor-intensive process, even for experts, and secondary liver cancers further complicate the diagnosis. Artificial intelligence (AI) offers promising solutions to these diagnostic challenges by facilitating the histopathological classification of tumors using digital whole slide images (WSIs). This study aimed to develop a deep learning model for distinguishing HCC, CC, and metastatic colorectal cancer (mCRC) using histopathological images and to discuss its clinical implications. The WSIs from HCC, CC, and mCRC were used to train the classifiers. For normal/tumor classification, the areas under the curve (AUCs) were 0.989, 0.988, and 0.991 for HCC, CC, and mCRC, respectively. Using proper tumor tissues, the HCC/other cancer type classifier was trained to effectively distinguish HCC from CC and mCRC, with a concatenated AUC of 0.998. Subsequently, the CC/mCRC classifier differentiated CC from mCRC with a concatenated AUC of 0.995. However, testing on an external dataset revealed that the HCC/other cancer type classifier underperformed with an AUC of 0.745. After combining the original training datasets with external datasets and retraining, the classification drastically improved, all achieving AUCs of 1.000. Although these results are promising and offer crucial insights into liver cancer, further research is required for model refinement and validation.
Collapse
Affiliation(s)
- Hyun-Jong Jang
- Department of Physiology, CMC Institute for Basic Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jai-Hyang Go
- Department of Pathology, Dankook University College of Medicine, Cheonan 31116, Republic of Korea;
| | - Younghoon Kim
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
10
|
Wagner SJ, Reisenbüchler D, West NP, Niehues JM, Zhu J, Foersch S, Veldhuizen GP, Quirke P, Grabsch HI, van den Brandt PA, Hutchins GGA, Richman SD, Yuan T, Langer R, Jenniskens JCA, Offermans K, Mueller W, Gray R, Gruber SB, Greenson JK, Rennert G, Bonner JD, Schmolze D, Jonnagaddala J, Hawkins NJ, Ward RL, Morton D, Seymour M, Magill L, Nowak M, Hay J, Koelzer VH, Church DN, Matek C, Geppert C, Peng C, Zhi C, Ouyang X, James JA, Loughrey MB, Salto-Tellez M, Brenner H, Hoffmeister M, Truhn D, Schnabel JA, Boxberg M, Peng T, Kather JN. Transformer-based biomarker prediction from colorectal cancer histology: A large-scale multicentric study. Cancer Cell 2023; 41:1650-1661.e4. [PMID: 37652006 PMCID: PMC10507381 DOI: 10.1016/j.ccell.2023.08.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
Deep learning (DL) can accelerate the prediction of prognostic biomarkers from routine pathology slides in colorectal cancer (CRC). However, current approaches rely on convolutional neural networks (CNNs) and have mostly been validated on small patient cohorts. Here, we develop a new transformer-based pipeline for end-to-end biomarker prediction from pathology slides by combining a pre-trained transformer encoder with a transformer network for patch aggregation. Our transformer-based approach substantially improves the performance, generalizability, data efficiency, and interpretability as compared with current state-of-the-art algorithms. After training and evaluating on a large multicenter cohort of over 13,000 patients from 16 colorectal cancer cohorts, we achieve a sensitivity of 0.99 with a negative predictive value of over 0.99 for prediction of microsatellite instability (MSI) on surgical resection specimens. We demonstrate that resection specimen-only training reaches clinical-grade performance on endoscopic biopsy tissue, solving a long-standing diagnostic problem.
Collapse
Affiliation(s)
- Sophia J Wagner
- Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany; School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | - Daniel Reisenbüchler
- Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany
| | - Nicholas P West
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Jan Moritz Niehues
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | - Jiefu Zhu
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | | | - Philip Quirke
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Piet A van den Brandt
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Gordon G A Hutchins
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Susan D Richman
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rupert Langer
- Institute of Pathology und Molecular Pathology, Johannes Kepler University Hospital Linz, Linz, Österreich
| | - Josien C A Jenniskens
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Kelly Offermans
- Department of Epidemiology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | | | - Richard Gray
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Stephen B Gruber
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Joel K Greenson
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Gad Rennert
- Department of Community Medicine & Epidemiology, Lady Davis Carmel Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Steve and Cindy Rasmussen Institute for Genomic Medicine, Lady Davis Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Joseph D Bonner
- Department of Community Medicine & Epidemiology, Lady Davis Carmel Medical Center, Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Daniel Schmolze
- Center for Precision Medicine and Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Jitendra Jonnagaddala
- School of Population Health, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Nicholas J Hawkins
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Robyn L Ward
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Dion Morton
- University Hospital Birmingham, Birmingham, UK
| | | | - Laura Magill
- University of Birmingham Clinical Trials Unit, Birmingham, UK
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jennifer Hay
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Oncology, University of Oxford, Oxford, UK; Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
| | - David N Church
- Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK; Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christian Matek
- Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany; Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nuremberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC), University Hospital Erlangen, FAU Erlangen-Nuremberg, Erlangen, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nuremberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC), University Hospital Erlangen, FAU Erlangen-Nuremberg, Erlangen, Germany
| | - Chaolong Peng
- Medical School, Jianggang Shan University, Jiangxi, China
| | - Cheng Zhi
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoming Ouyang
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jacqueline A James
- Precision Medicine Centre of Excellence, Health Sciences Building, The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Regional Molecular Diagnostic Service, Belfast Health and Social Care Trust, Belfast, UK; The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Maurice B Loughrey
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Department of Cellular Pathology, Belfast Health and Social Care Trust, Belfast, UK; Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Manuel Salto-Tellez
- Precision Medicine Centre of Excellence, Health Sciences Building, The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Regional Molecular Diagnostic Service, Belfast Health and Social Care Trust, Belfast, UK; Integrated Pathology Unit, Institute for Cancer Research and Royal Marsden Hospital, London, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julia A Schnabel
- Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany; School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany; Institute of Pathology Munich-North, Munich, Germany
| | - Tingying Peng
- Helmholtz Munich - German Research Center for Environment and Health, Munich, Germany.
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany; Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg.
| |
Collapse
|
11
|
Zhang Y, Chen S, Wang Y, Li J, Xu K, Chen J, Zhao J. Deep learning-based methods for classification of microsatellite instability in endometrial cancer from HE-stained pathological images. J Cancer Res Clin Oncol 2023; 149:8877-8888. [PMID: 37150803 DOI: 10.1007/s00432-023-04838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/03/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Microsatellite instability (MSI) is one of the essential tumor biomarkers for cancer treatment and prognosis. The presence of more significant PD-L1 expression on the surface of tumor cells in endometrial cancer with MSI suggests that MSI may be a promising biomarker for anti-PD-1/PD-L1 immunotherapy. However, the conventional testing methods are labor-intensive and expensive for patients. METHODS Inspired by classifiers for MSI based on fast and low-cost deep-learning methods in previous investigations, a new architecture for MSI classification based on an attention module is proposed to extract features from pathological images. Especially, slide-level microsatellite status will be obtained by the bag of words method to aggregate probabilities predicted by the proposed model. The H&E-stained whole slide images (WSIs) from The Cancer Genome Atlas endometrial cohort are collected as the dataset. The performances of the proposed model were primarily evaluated by the area under the receiver-operating characteristic curve, accuracy, sensitivity, and F1-Score. RESULTS On the randomly divided test dataset, the proposed model achieved an accuracy of 0.80, a sensitivity of 0.857, a F1-Score of 0.826, and an AUROC of 0.799. We then visualize the results of the microsatellite status classification to capture more specific morphological features, helping pathologists better understand how deep learning performs the classification. CONCLUSIONS This study implements the prediction of microsatellite status in endometrial cancer cases using deep-learning methods directly from H&E-stained WSIs. The proposed architecture can help the model capture more valuable features for classification. In contrast to current laboratory testing methods, the proposed model creates a more convenient screening tool for rapid automated testing for patients. This method can potentially be a clinical method for detecting the microsatellite status of endometrial cancer.
Collapse
Affiliation(s)
- Ying Zhang
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shijie Chen
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuling Wang
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jingjing Li
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kai Xu
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jyhcheng Chen
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jie Zhao
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
12
|
Porta FM, Sajjadi E, Venetis K, Frascarelli C, Cursano G, Guerini-Rocco E, Fusco N, Ivanova M. Immune Biomarkers in Triple-Negative Breast Cancer: Improving the Predictivity of Current Testing Methods. J Pers Med 2023; 13:1176. [PMID: 37511789 PMCID: PMC10381494 DOI: 10.3390/jpm13071176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant challenge in terms of prognosis and disease recurrence. The limited treatment options and the development of resistance to chemotherapy make it particularly difficult to manage these patients. However, recent research has been shifting its focus towards biomarker-based approaches for TNBC, with a particular emphasis on the tumor immune landscape. Immune biomarkers in TNBC are now a subject of great interest due to the presence of tumor-infiltrating lymphocytes (TILs) in these tumors. This characteristic often coincides with the presence of PD-L1 expression on both neoplastic cells and immune cells within the tumor microenvironment. Furthermore, a subset of TNBC harbor mismatch repair deficient (dMMR) TNBC, which is frequently accompanied by microsatellite instability (MSI). All of these immune biomarkers hold actionable potential for guiding patient selection in immunotherapy. To fully capitalize on these opportunities, the identification of additional or complementary biomarkers and the implementation of highly customized testing strategies are of paramount importance in TNBC. In this regard, this article aims to provide an overview of the current state of the art in immune-related biomarkers for TNBC. Specifically, it focuses on the various testing methodologies available and sheds light on the immediate future perspectives for patient selection. By delving into the advancements made in understanding the immune landscape of TNBC, this study aims to contribute to the growing body of knowledge in the field. The ultimate goal is to pave the way for the development of more personalized testing strategies, ultimately improving outcomes for TNBC patients.
Collapse
Affiliation(s)
- Francesca Maria Porta
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| |
Collapse
|
13
|
Lo CM, Yang YW, Lin JK, Lin TC, Chen WS, Yang SH, Chang SC, Wang HS, Lan YT, Lin HH, Huang SC, Cheng HH, Jiang JK, Lin CC. Modeling the survival of colorectal cancer patients based on colonoscopic features in a feature ensemble vision transformer. Comput Med Imaging Graph 2023; 107:102242. [PMID: 37172354 DOI: 10.1016/j.compmedimag.2023.102242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 05/14/2023]
Abstract
The prognosis of patients with colorectal cancer (CRC) mostly relies on the classic tumor node metastasis (TNM) staging classification. A more accurate and convenient prediction model would provide a better prognosis and assist in treatment. From May 2014 to December 2017, patients who underwent an operation for CRC were enrolled. The proposed feature ensemble vision transformer (FEViT) used ensemble classifiers to benefit the combinations of relevant colonoscopy features from the pretrained vision transformer and clinical features, including sex, age, family history of CRC, and tumor location, to establish the prognostic model. A total of 1729 colonoscopy images were enrolled in the current retrospective study. For the prediction of patient survival, FEViT achieved an accuracy of 94 % with an area under the receiver operating characteristic curve of 0.93, which was better than the TNM staging classification (90 %, 0.83) in the experiment. FEViT reduced the limited receptive field and gradient disappearance in the conventional convolutional neural network and was a relatively effective and efficient procedure. The promising accuracy of FEViT in modeling survival makes the prognosis of CRC patients more predictable and practical.
Collapse
Affiliation(s)
- Chung-Ming Lo
- Graduate Institute of Library, Information and Archival Studies, National Chengchi University, Taipei, Taiwan
| | - Yi-Wen Yang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jen-Kou Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Chen Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Shone Chen
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shung-Haur Yang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Surgery, National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan
| | - Shih-Ching Chang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Huann-Sheng Wang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuan-Tzu Lan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hung-Hsin Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sheng-Chieh Huang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hou-Hsuan Cheng
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jeng-Kai Jiang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chi Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
14
|
Kim S, Lee JH, Park EJ, Lee HS, Baik SH, Jeon TJ, Lee KY, Ryu YH, Kang J. Prediction of Microsatellite Instability in Colorectal Cancer Using a Machine Learning Model Based on PET/CT Radiomics. Yonsei Med J 2023; 64:320-326. [PMID: 37114635 PMCID: PMC10151228 DOI: 10.3349/ymj.2022.0548] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
PURPOSE We investigated the feasibility of preoperative 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) radiomics with machine learning to predict microsatellite instability (MSI) status in colorectal cancer (CRC) patients. MATERIALS AND METHODS Altogether, 233 patients with CRC who underwent preoperative FDG PET/CT were enrolled and divided into training (n=139) and test (n=94) sets. A PET-based radiomics signature (rad_score) was established to predict the MSI status in patients with CRC. The predictive ability of the rad_score was evaluated using the area under the receiver operating characteristic curve (AUROC) in the test set. A logistic regression model was used to determine whether the rad_score was an independent predictor of MSI status in CRC. The predictive performance of rad_score was compared with conventional PET parameters. RESULTS The incidence of MSI-high was 15 (10.8%) and 10 (10.6%) in the training and test sets, respectively. The rad_score was constructed based on the two radiomic features and showed similar AUROC values for predicting MSI status in the training and test sets (0.815 and 0.867, respectively; p=0.490). Logistic regression analysis revealed that the rad_score was an independent predictor of MSI status in the training set. The rad_score performed better than metabolic tumor volume when assessed using the AUROC (0.867 vs. 0.794, p=0.015). CONCLUSION Our predictive model incorporating PET radiomic features successfully identified the MSI status of CRC, and it also showed better performance than the conventional PET image parameters.
Collapse
Affiliation(s)
- Soyoung Kim
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Eun Jung Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hyuk Baik
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Joo Jeon
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kang Young Lee
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeonghyun Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Niehues JM, Quirke P, West NP, Grabsch HI, van Treeck M, Schirris Y, Veldhuizen GP, Hutchins GGA, Richman SD, Foersch S, Brinker TJ, Fukuoka J, Bychkov A, Uegami W, Truhn D, Brenner H, Brobeil A, Hoffmeister M, Kather JN. Generalizable biomarker prediction from cancer pathology slides with self-supervised deep learning: A retrospective multi-centric study. Cell Rep Med 2023; 4:100980. [PMID: 36958327 PMCID: PMC10140458 DOI: 10.1016/j.xcrm.2023.100980] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/28/2022] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
Deep learning (DL) can predict microsatellite instability (MSI) from routine histopathology slides of colorectal cancer (CRC). However, it is unclear whether DL can also predict other biomarkers with high performance and whether DL predictions generalize to external patient populations. Here, we acquire CRC tissue samples from two large multi-centric studies. We systematically compare six different state-of-the-art DL architectures to predict biomarkers from pathology slides, including MSI and mutations in BRAF, KRAS, NRAS, and PIK3CA. Using a large external validation cohort to provide a realistic evaluation setting, we show that models using self-supervised, attention-based multiple-instance learning consistently outperform previous approaches while offering explainable visualizations of the indicative regions and morphologies. While the prediction of MSI and BRAF mutations reaches a clinical-grade performance, mutation prediction of PIK3CA, KRAS, and NRAS was clinically insufficient.
Collapse
Affiliation(s)
- Jan Moritz Niehues
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK; Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, 6229 HX Maastricht, the Netherlands
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Yoni Schirris
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; University of Amsterdam, 1012 WP Amsterdam, the Netherlands
| | - Gregory P Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Gordon G A Hutchins
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Susan D Richman
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan; Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Wataru Uegami
- Department of Pathology, Kameda Medical Center, Kamogawa 296-8602, Chiba, Japan
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 01307 Dresden, Germany; Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds LS9 7TF, UK; Department of Medicine I, University Hospital Dresden, 01307 Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
16
|
Wen Z, Wang S, Yang DM, Xie Y, Chen M, Bishop J, Xiao G. Deep learning in digital pathology for personalized treatment plans of cancer patients. Semin Diagn Pathol 2023; 40:109-119. [PMID: 36890029 DOI: 10.1053/j.semdp.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Over the past decade, many new cancer treatments have been developed and made available to patients. However, in most cases, these treatments only benefit a specific subgroup of patients, making the selection of treatment for a specific patient an essential but challenging task for oncologists. Although some biomarkers were found to associate with treatment response, manual assessment is time-consuming and subjective. With the rapid developments and expanded implementation of artificial intelligence (AI) in digital pathology, many biomarkers can be quantified automatically from histopathology images. This approach allows for a more efficient and objective assessment of biomarkers, aiding oncologists in formulating personalized treatment plans for cancer patients. This review presents an overview and summary of the recent studies on biomarker quantification and treatment response prediction using hematoxylin-eosin (H&E) stained pathology images. These studies have shown that an AI-based digital pathology approach can be practical and will become increasingly important in improving the selection of cancer treatments for patients.
Collapse
Affiliation(s)
- Zhuoyu Wen
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shidan Wang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donghan M Yang
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mingyi Chen
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Justin Bishop
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Brummel K, Eerkens AL, de Bruyn M, Nijman HW. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer 2023; 128:451-458. [PMID: 36564565 PMCID: PMC9938191 DOI: 10.1038/s41416-022-02119-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour-infiltrating lymphocytes (TILs) are considered crucial in anti-tumour immunity. Accordingly, the presence of TILs contains prognostic and predictive value. In 2011, we performed a systematic review and meta-analysis on the prognostic value of TILs across cancer types. Since then, the advent of immune checkpoint blockade (ICB) has renewed interest in the analysis of TILs. In this review, we first describe how our understanding of the prognostic value of TIL has changed over the last decade. New insights on novel TIL subsets are discussed and give a broader view on the prognostic effect of TILs in cancer. Apart from prognostic value, evidence on the predictive significance of TILs in the immune therapy era are discussed, as well as new techniques, such as machine learning that strive to incorporate these predictive capacities within clinical trials.
Collapse
Affiliation(s)
- Koen Brummel
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Anneke L Eerkens
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Marco de Bruyn
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Hans W Nijman
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands.
| |
Collapse
|
18
|
Dey A, Mitra A, Pathak S, Prasad S, Zhang AS, Zhang H, Sun XF, Banerjee A. Recent Advancements, Limitations, and Future Perspectives of the use of Personalized Medicine in Treatment of Colon Cancer. Technol Cancer Res Treat 2023; 22:15330338231178403. [PMID: 37248615 PMCID: PMC10240881 DOI: 10.1177/15330338231178403] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/18/2023] [Accepted: 03/13/2023] [Indexed: 08/29/2024] Open
Abstract
Due to the heterogeneity of colon cancer, surgery, chemotherapy, and radiation are ineffective in all cases. The genomic profile and biomarkers associated with the process are considered in personalized medicine, along with the patient's personal history. It is based on the response of the targeted therapies to specific genetic variations. The patient's genetic transcriptomic and epigenetic features are evaluated, and the best therapeutic approach and diagnostic testing are identified through personalized medicine. This review aims to summarize all the necessary, updated information on colon cancer related to personalized medicine. Personalized medicine is gaining prominence as generalized treatments are finding it challenging to contain colon cancer cases which currently rank fourth among global cancer incidence while being the fifth largest in total death cases worldwide. In personalized therapy, patients are grouped into specific categories, and the best therapeutic approach is chosen based on evaluating their molecular features. Various personalized strategies are currently being explored in the treatment of colon cancer involving immunotherapy, phytochemicals, and other biomarker-specific targeted therapies. However, significant challenges must be overcome to integrate personalized medicine into healthcare systems completely. We look at the various signaling pathways and genetic and epigenetic alterations associated with colon cancer to understand and identify biomarkers useful in targeted therapy. The current personalized therapies available in colon cancer treatment and the strategies being explored to improve the existing methods are discussed. This review highlights the advantages and limitations of personalized medicine in colon cancer therapy. The current scenario of personalized medicine in developed countries and the challenges faced in middle- and low-income countries are also summarized. Finally, we discuss the future perspectives of personalized medicine in colon cancer and how it could be integrated into the healthcare systems.
Collapse
Affiliation(s)
- Amit Dey
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Abhijit Mitra
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Suhanya Prasad
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Białystok, Poland
| | | | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Orebro University, Örebro, Sweden
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| |
Collapse
|
19
|
Pham TD, Ravi V, Fan C, Luo B, Sun XF. Classification of IHC Images of NATs With ResNet-FRP-LSTM for Predicting Survival Rates of Rectal Cancer Patients. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2022; 11:87-95. [PMID: 36704244 PMCID: PMC9870269 DOI: 10.1109/jtehm.2022.3229561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Over a decade, tissues dissected adjacent to primary tumors have been considered "normal" or healthy samples (NATs). However, NATs have recently been discovered to be distinct from both tumorous and normal tissues. The ability to predict the survival rate of cancer patients using NATs can open a new door to selecting optimal treatments for cancer and discovering biomarkers. METHODS This paper introduces an artificial intelligence (AI) approach that uses NATs for predicting the 5-year survival of pre-operative radiotherapy patients with rectal cancer. The new approach combines pre-trained deep learning, nonlinear dynamics, and long short-term memory to classify immunohistochemical images of RhoB protein expression on NATs. RESULTS Ten-fold cross-validation results show 88% accuracy of prediction obtained from the new approach, which is also higher than those provided from baseline methods. CONCLUSION Preliminary results not only add objective evidence to recent findings of NATs' molecular characteristics using state-of-the-art AI methods, but also contribute to the discovery of RhoB expression on NATs in rectal-cancer patients. CLINICAL IMPACT The ability to predict the survival rate of cancer patients is extremely important for clinical decision-making. The proposed AI tool is promising for assisting oncologists in their treatments of rectal cancer patients.
Collapse
Affiliation(s)
- Tuan D Pham
- Center for Artificial IntelligencePrince Mohammad Bin Fahd University Khobar 31952 Saudi Arabia
| | - Vinayakumar Ravi
- Center for Artificial IntelligencePrince Mohammad Bin Fahd University Khobar 31952 Saudi Arabia
| | - Chuanwen Fan
- Department of OncologyLinkoping University 58185 Linkoping Sweden
- Department of Biomedical and Clinical SciencesLinkoping University 58185 Linkoping Sweden
| | - Bin Luo
- Department of OncologyLinkoping University 58185 Linkoping Sweden
- Department of Biomedical and Clinical SciencesLinkoping University 58185 Linkoping Sweden
- Department of Gastrointestinal SurgerySichuan Provincial People's Hospital Chengdu 610032 China
| | - Xiao-Feng Sun
- Department of OncologyLinkoping University 58185 Linkoping Sweden
- Department of Biomedical and Clinical SciencesLinkoping University 58185 Linkoping Sweden
| |
Collapse
|
20
|
Nakanishi R, Morooka K, Omori K, Toyota S, Tanaka Y, Hasuda H, Koga N, Nonaka K, Hu Q, Nakaji Y, Nakanoko T, Ando K, Ota M, Kimura Y, Oki E, Oda Y, Yoshizumi T. Artificial Intelligence-Based Prediction of Recurrence after Curative Resection for Colorectal Cancer from Digital Pathological Images. Ann Surg Oncol 2022; 30:3506-3514. [PMID: 36512260 DOI: 10.1245/s10434-022-12926-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/19/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND To develop an artificial intelligence-based model to predict recurrence after curative resection for stage I-III colorectal cancer from digitized pathological slides. PATIENTS AND METHODS In this retrospective study, 471 consecutive patients who underwent curative resection for stage I-III colorectal cancer at our institution from 2004 to 2015 were enrolled, and 512 randomly selected tiles from digitally scanned images of hematoxylin and eosin-stained tumor tissue sections were used to train a convolutional neural network. Five-fold cross-validation was used to validate the model. The association between recurrence and the model's output scores were analyzed in the test cohorts. RESULTS The area under the receiver operating characteristic curve of the cross-validation was 0.7245 [95% confidence interval (CI) 0.6707-0.7783; P < 0.0001]. The score successfully classified patients into those with better and worse recurrence free survival (P < 0.0001). Multivariate analysis revealed that a high score was significantly associated with worse recurrence free survival [odds ratio (OR) 1.857; 95% CI 1.248-2.805; P = 0.0021], which was independent from other predictive factors: male sex (P = 0.0238), rectal cancer (P = 0.0396), preoperative abnormal carcinoembryonic antigen (CEA) level (P = 0.0216), pathological T3/T4 stage (P = 0.0162), and pathological positive lymph node metastasis (P < 0.0001). CONCLUSIONS The artificial intelligence-based prediction model discriminated patients with a high risk of recurrence. This approach could help decision-makers consider the benefits of adjuvant chemotherapy.
Collapse
|
21
|
Shen Y, Shen D, Ke J. Identify Representative Samples by Conditional Random Field of Cancer Histology Images. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:3835-3848. [PMID: 35951579 DOI: 10.1109/tmi.2022.3198526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pathology analysis is crucial to precise cancer diagnoses and the succeeding treatment plan as well. To detect abnormality in histopathology images with prevailing patch-based convolutional neural networks (CNNs), contextual information often serves as a powerful cue. However, as whole-slide images (WSIs) are characterized by intense morphological heterogeneity and extensive tissue scale, a straightforward visual span to a larger context may not well capture the information closely associated with the focal patch. In this paper, we propose a novel pixel-offset based patch-location method to identify high-representative tissues, with a CNN backbone. Pathology Deformable Conditional Random Field (PDCRF) is proposed to learn the offsets and weights of neighboring contexts in a spatial-adaptive manner, to search for high-representative patches. A CNN structure with the localized patches as training input is then capable of consistently reaching superior classification outcomes for histology images. Overall, the proposed method has achieved state-of-the-art performance, in terms of the test classification accuracy improvement to the baseline by 1.15-2.60%, 0.78-1.78%, and 1.47-2.18% on TCGA public datasets of TCGA-STAD, TCGA-COAD, and TCGA-READ respectively. It also achieves 88.95% test accuracy and 0.920 test AUC on Camelyon 16. To show the effectiveness of the proposed framework on downstream tasks, we take a further step by incorporating an active learning model, which noticeably reduces the number of manual annotations by PDCRF to reach a parallel patch-based histology classifier.
Collapse
|
22
|
Lee SH, Jang HJ. Deep learning-based prediction of molecular cancer biomarkers from tissue slides: A new tool for precision oncology. Clin Mol Hepatol 2022; 28:754-772. [PMID: 35443570 PMCID: PMC9597228 DOI: 10.3350/cmh.2021.0394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/17/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular tests are necessary to stratify cancer patients for targeted therapy. However, high cost and technical barriers limit the application of these tests, hindering optimal treatment. Recently, deep learning (DL) has been applied to predict molecular test results from digitized images of tissue slides. Furthermore, treatment response and prognosis can be predicted from tissue slides using DL. In this review, we summarized DL-based studies regarding the prediction of genetic mutation, microsatellite instability, tumor mutational burden, molecular subtypes, gene expression, treatment response, and prognosis directly from hematoxylin- and eosin-stained tissue slides. Although performance needs to be improved, these studies clearly demonstrated the feasibility of DL-based prediction of key molecular features in cancer tissues. With the accumulation of data and technical advances, the performance of the DL system could be improved in the near future. Therefore, we expect that DL could provide cost- and time-effective alternative tools for patient stratification in the era of precision oncology.
Collapse
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, Korea,Corresponding author : Hyun-Jong Jang Department of Physiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-7274, Fax: +82-2-532-9575, E-mail:
| |
Collapse
|
23
|
Lou J, Xu J, Zhang Y, Sun Y, Fang A, Liu J, Mur LAJ, Ji B. PPsNet: An improved deep learning model for microsatellite instability high prediction in colorectal cancer from whole slide images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 225:107095. [PMID: 36057226 DOI: 10.1016/j.cmpb.2022.107095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Recent studies have shown that colorectal cancer (CRC) patients with microsatellite instability high (MSI-H) are more likely to benefit from immunotherapy. However, current MSI testing methods are not available for all patients due to the lack of available equipment and trained personnel, as well as the high cost of the assay. Here, we developed an improved deep learning model to predict MSI-H in CRC from whole slide images (WSIs). METHODS We established the MSI-H prediction model based on two stages: tumor detection and MSI classification. Previous works applied fine-tuning strategy directly for tumor detection, but ignoring the challenge of vanishing gradient due to the large number of convolutional layers. We added auxiliary classifiers to intermediate layers of pre-trained models to help propagate gradients back through in an effective manner. To predict MSI status, we constructed a pair-wise learning model with a synergic network, named parameter partial sharing network (PPsNet), where partial parameters are shared among two deep convolutional neural networks (DCNNs). The proposed PPsNet contained fewer parameters and reduced the problem of intra-class variation and inter-class similarity. We validated the proposed model on a holdout test set and two external test sets. RESULTS 144 H&E-stained WSIs from 144 CRC patients (81 cases with MSI-H and 63 cases with MSI-L/MSS) were collected retrospectively from three hospitals. The experimental results indicate that deep supervision based fine-tuning almost outperforms training from scratch and utilizing fine-tuning directly. The proposed PPsNet always achieves better accuracy and area under the receiver operating characteristic curve (AUC) than other solutions with four different neural network architectures on validation. The proposed method finally achieves obvious improvements than other state-of-the-art methods on the validation dataset with an accuracy of 87.28% and AUC of 94.29%. CONCLUSIONS The proposed method can obviously increase model performance and our model yields better performance than other methods. Additionally, this work also demonstrates the feasibility of MSI-H prediction using digital pathology images based on deep learning in the Asian population. It is hoped that this model could serve as an auxiliary tool to identify CRC patients with MSI-H more time-saving and efficiently.
Collapse
Affiliation(s)
- Jingjiao Lou
- School of Control Science and Engineering, Shandong University, 17923 Jingshi Road, Jinan, Shandong 250061, PR China
| | - Jiawen Xu
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Yuyan Zhang
- School of Control Science and Engineering, Shandong University, 17923 Jingshi Road, Jinan, Shandong 250061, PR China
| | - Yuhong Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Aiju Fang
- Department of Pathology, Shandong Provincial Third Hospital, Shandong University, Jinan, Shandong 250132, PR China
| | - Jixuan Liu
- Department of Pathology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Luis A J Mur
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, Wales SY23 3DZ, UK
| | - Bing Ji
- School of Control Science and Engineering, Shandong University, 17923 Jingshi Road, Jinan, Shandong 250061, PR China.
| |
Collapse
|
24
|
Wang Y, Hu C, Kwok T, Bain CA, Xue X, Gasser RB, Webb GI, Boussioutas A, Shen X, Daly RJ, Song J. DEMoS: a deep learning-based ensemble approach for predicting the molecular subtypes of gastric adenocarcinomas from histopathological images. Bioinformatics 2022; 38:4206-4213. [PMID: 35801909 DOI: 10.1093/bioinformatics/btac456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/16/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION The molecular subtyping of gastric cancer (adenocarcinoma) into four main subtypes based on integrated multiomics profiles, as proposed by The Cancer Genome Atlas (TCGA) initiative, represents an effective strategy for patient stratification. However, this approach requires the use of multiple technological platforms, and is quite expensive and time-consuming to perform. A computational approach that uses histopathological image data to infer molecular subtypes could be a practical, cost- and time-efficient complementary tool for prognostic and clinical management purposes. RESULTS Here, we propose a deep learning ensemble approach (called DEMoS) capable of predicting the four recognized molecular subtypes of gastric cancer directly from histopathological images. DEMoS achieved tile-level area under the receiver-operating characteristic curve (AUROC) values of 0.785, 0.668, 0.762 and 0.811 for the prediction of these four subtypes of gastric cancer [i.e. (i) Epstein-Barr (EBV)-infected, (ii) microsatellite instability (MSI), (iii) genomically stable (GS) and (iv) chromosomally unstable tumors (CIN)] using an independent test dataset, respectively. At the patient-level, it achieved AUROC values of 0.897, 0.764, 0.890 and 0.898, respectively. Thus, these four subtypes are well-predicted by DEMoS. Benchmarking experiments further suggest that DEMoS is able to achieve an improved classification performance for image-based subtyping and prevent model overfitting. This study highlights the feasibility of using a deep learning ensemble-based method to rapidly and reliably subtype gastric cancer (adenocarcinoma) solely using features from histopathological images. AVAILABILITY AND IMPLEMENTATION All whole slide images used in this study was collected from the TCGA database. This study builds upon our previously published HEAL framework, with related documentation and tutorials available at http://heal.erc.monash.edu.au. The source code and related models are freely accessible at https://github.com/Docurdt/DEMoS.git. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yanan Wang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Changyuan Hu
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Terry Kwok
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Christopher A Bain
- Faculty of Information Technology, Monash University, Melbourne 3800, Australia
| | - Xiangyang Xue
- Department of General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Geoffrey I Webb
- Faculty of Information Technology, Monash Centre for Data Science, Monash University, Melbourne 3800, Australia.,Department of Data Science and Artificial Intelligence, Monash University, Melbourne, VIC 3800, Australia
| | - Alex Boussioutas
- The Alfred Hospital, Melbourne, VIC 3004, Australia.,Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3010, Australia
| | - Xian Shen
- Department of General Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Roger J Daly
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Jiangning Song
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia.,Department of Data Science and Artificial Intelligence, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
25
|
Zhang T, Chen J, Lu Y, Yang X, Ouyang Z. Identification of technology frontiers of artificial intelligence-assisted pathology based on patent citation network. PLoS One 2022; 17:e0273355. [PMID: 35994484 PMCID: PMC9394838 DOI: 10.1371/journal.pone.0273355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/05/2022] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES This paper aimed to identify the technology frontiers of artificial intelligence-assisted pathology based on patent citation network. METHODS Patents related to artificial intelligence-assisted pathology were searched and collected from the Derwent Innovation Index (DII), which were imported into Derwent Data Analyzer (DDA, Clarivate Derwent, New York, NY, USA) for authority control, and imported into the freely available computer program Ucinet 6 for drawing the patent citation network. The patent citation network according to the citation relationship could describe the technology development context in the field of artificial intelligence-assisted pathology. The patent citations were extracted from the collected patent data, selected highly cited patents to form a co-occurrence matrix, and built a patent citation network based on the co-occurrence matrix in each period. Text clustering is an unsupervised learning method, an important method in text mining, where similar documents are grouped into clusters. The similarity between documents are determined by calculating the distance between them, and the two documents with the closest distance are combined. The method of text clustering was used to identify the technology frontiers based on the patent citation network, which was according to co-word analysis of the title and abstract of the patents in this field. RESULTS 1704 patents were obtained in the field of artificial intelligence-assisted pathology, which had been currently undergoing three stages, namely the budding period (1992-2000), the development period (2001-2015), and the rapid growth period (2016-2021). There were two technology frontiers in the budding period (1992-2000), namely systems and methods for image data processing in computerized tomography (CT), and immunohistochemistry (IHC), five technology frontiers in the development period (2001-2015), namely spectral analysis methods of biomacromolecules, pathological information system, diagnostic biomarkers, molecular pathology diagnosis, and pathological diagnosis antibody, and six technology frontiers in the rapid growth period (2016-2021), namely digital pathology (DP), deep learning (DL) algorithms-convolutional neural networks (CNN), disease prediction models, computational pathology, pathological image analysis method, and intelligent pathological system. CONCLUSIONS Artificial intelligence-assisted pathology was currently in a rapid development period, and computational pathology, DL and other technologies in this period all involved the study of algorithms. Future research hotspots in this field would focus on algorithm improvement and intelligent diagnosis in order to realize the precise diagnosis. The results of this study presented an overview of the characteristics of research status and development trends in the field of artificial intelligence-assisted pathology, which could help readers broaden innovative ideas and discover new technological opportunities, and also served as important indicators for government policymaking.
Collapse
Affiliation(s)
- Ting Zhang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Juan Chen
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yan Lu
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiaoyi Yang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Zhaolian Ouyang
- Institute of Medical Information & Library, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Lee SH, Lee Y, Jang H. Deep learning captures selective features for discrimination of microsatellite instability from pathologic tissue slides of gastric cancer. Int J Cancer 2022; 152:298-307. [DOI: 10.1002/ijc.34251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology Seoul St. Mary's Hospital
| | - Yujin Lee
- Department of Hospital Pathology St. Vincent's Hospital
| | - Hyun‐Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine The Catholic University of Korea Seoul South Korea
| |
Collapse
|
27
|
Fremond S, Koelzer VH, Horeweg N, Bosse T. The evolving role of morphology in endometrial cancer diagnostics: From histopathology and molecular testing towards integrative data analysis by deep learning. Front Oncol 2022; 12:928977. [PMID: 36059702 PMCID: PMC9433878 DOI: 10.3389/fonc.2022.928977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Endometrial cancer (EC) diagnostics is evolving into a system in which molecular aspects are increasingly important. The traditional histological subtype-driven classification has shifted to a molecular-based classification that stratifies EC into DNA polymerase epsilon mutated (POLEmut), mismatch repair deficient (MMRd), and p53 abnormal (p53abn), and the remaining EC as no specific molecular profile (NSMP). The molecular EC classification has been implemented in the World Health Organization 2020 classification and the 2021 European treatment guidelines, as it serves as a better basis for patient management. As a result, the integration of the molecular class with histopathological variables has become a critical focus of recent EC research. Pathologists have observed and described several morphological characteristics in association with specific genomic alterations, but these appear insufficient to accurately classify patients according to molecular subgroups. This requires pathologists to rely on molecular ancillary tests in routine workup. In this new era, it has become increasingly challenging to assign clinically relevant weights to histological and molecular features on an individual patient basis. Deep learning (DL) technology opens new options for the integrative analysis of multi-modal image and molecular datasets with clinical outcomes. Proof-of-concept studies in other cancers showed promising accuracy in predicting molecular alterations from H&E-stained tumor slide images. This suggests that some morphological characteristics that are associated with molecular alterations could be identified in EC, too, expanding the current understanding of the molecular-driven EC classification. Here in this review, we report the morphological characteristics of the molecular EC classification currently identified in the literature. Given the new challenges in EC diagnostics, this review discusses, therefore, the potential supportive role that DL could have, by providing an outlook on all relevant studies using DL on histopathology images in various cancer types with a focus on EC. Finally, we touch upon how DL might shape the management of future EC patients.
Collapse
Affiliation(s)
- Sarah Fremond
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Viktor Hendrik Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Nanda Horeweg
- Department of Radiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center (LUMC), Leiden, Netherlands
- *Correspondence: Tjalling Bosse,
| |
Collapse
|
28
|
Jiang W, Mei WJ, Xu SY, Ling YH, Li WR, Kuang JB, Li HS, Hui H, Li JB, Cai MY, Pan ZZ, Zhang HZ, Li L, Ding PR. Clinical actionability of triaging DNA mismatch repair deficient colorectal cancer from biopsy samples using deep learning. EBioMedicine 2022; 81:104120. [PMID: 35753152 PMCID: PMC9240789 DOI: 10.1016/j.ebiom.2022.104120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022] Open
Abstract
Background We aimed to develop a deep learning (DL) model to predict DNA mismatch repair (MMR) status in colorectal cancers (CRC) based on hematoxylin and eosin-stained whole-slide images (WSIs) and assess its clinical applicability. Methods The DL model was developed and validated through three-fold cross validation using 441 WSIs from the Cancer Genome Atlas (TCGA) and externally validated using 78 WSIs from the Pathology AI Platform (PAIP), and 355 WSIs from surgical specimens and 341 WSIs from biopsy specimens of the Sun Yet-sun University Cancer Center (SYSUCC). Domain adaption and multiple instance learning (MIL) techniques were adopted for model development. The performance of the models was evaluated using the area under the receiver operating characteristic curve (AUROC). A dual-threshold strategy was also built from the surgical cohorts and validated in the biopsy cohort. Sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), F1-score, and the percentage of patients avoiding IHC testing were evaluated. Findings The MIL model achieved an AUROC of 0·8888±0·0357 in the TCGA-validation cohort, 0·8806±0·0232 in the PAIP cohort, 0·8457±0·0233 in the SYSUCC-surgical cohort, and 0·7679±0·0342 in the SYSUCC-biopsy cohort. A dual-threshold triage strategy was used to rule-in and rule-out dMMR patients with remaining uncertain patients recommended for further IHC testing, which kept sensitivity higher than 90% and specificity higher than 95% on deficient MMR patient triage from both the surgical and biopsy specimens, result in more than half of patients avoiding IHC based MMR testing. Interpretation A DL-based method that could directly predict CRC MMR status from WSIs was successfully developed, and a dual-threshold triage strategy was established to minimize the number of patients for further IHC testing. Funding The study was funded by the National Natural Science Foundation of China (82073159, 81871971 and 81700576), the Natural Science Foundation of Guangdong Province (No. 2021A1515011792 and No.2022A1515012403) and Medical Scientific Research Foundation of Guangdong Province of China (No. A2020392).
Collapse
Affiliation(s)
- Wu Jiang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wei-Jian Mei
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Shuo-Yu Xu
- Bio-totem Pte Ltd, Foshan, PR China; Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Yi-Hong Ling
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wei-Rong Li
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, PR China
| | | | | | - Hui Hui
- Bio-totem Pte Ltd, Foshan, PR China
| | - Ji-Bin Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Mu-Yan Cai
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Zhi-Zhong Pan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Hui-Zhong Zhang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Li Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Pei-Rong Ding
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, PR China; Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| |
Collapse
|
29
|
Alam MR, Abdul-Ghafar J, Yim K, Thakur N, Lee SH, Jang HJ, Jung CK, Chong Y. Recent Applications of Artificial Intelligence from Histopathologic Image-Based Prediction of Microsatellite Instability in Solid Cancers: A Systematic Review. Cancers (Basel) 2022; 14:2590. [PMID: 35681570 PMCID: PMC9179592 DOI: 10.3390/cancers14112590] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/07/2022] [Accepted: 05/22/2022] [Indexed: 12/11/2022] Open
Abstract
Cancers with high microsatellite instability (MSI-H) have a better prognosis and respond well to immunotherapy. However, MSI is not tested in all cancers because of the additional costs and time of diagnosis. Therefore, artificial intelligence (AI)-based models have been recently developed to evaluate MSI from whole slide images (WSIs). Here, we aimed to assess the current state of AI application to predict MSI based on WSIs analysis in MSI-related cancers and suggest a better study design for future studies. Studies were searched in online databases and screened by reference type, and only the full texts of eligible studies were reviewed. The included 14 studies were published between 2018 and 2021, and most of the publications were from developed countries. The commonly used dataset is The Cancer Genome Atlas dataset. Colorectal cancer (CRC) was the most common type of cancer studied, followed by endometrial, gastric, and ovarian cancers. The AI models have shown the potential to predict MSI with the highest AUC of 0.93 in the case of CRC. The relatively limited scale of datasets and lack of external validation were the limitations of most studies. Future studies with larger datasets are required to implicate AI models in routine diagnostic practice for MSI prediction.
Collapse
Affiliation(s)
- Mohammad Rizwan Alam
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Kwangil Yim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Nishant Thakur
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| | - Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.R.A.); (J.A.-G.); (K.Y.); (N.T.); (S.H.L.); (C.K.J.)
| |
Collapse
|
30
|
Area under the curve may hide poor generalisation to external datasets. ESMO Open 2022; 7:100429. [PMID: 35397433 PMCID: PMC9006654 DOI: 10.1016/j.esmoop.2022.100429] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022] Open
|
31
|
Deep Learning on Histopathological Images for Colorectal Cancer Diagnosis: A Systematic Review. Diagnostics (Basel) 2022; 12:diagnostics12040837. [PMID: 35453885 PMCID: PMC9028395 DOI: 10.3390/diagnostics12040837] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is the second most common cancer in women and the third most common in men, with an increasing incidence. Pathology diagnosis complemented with prognostic and predictive biomarker information is the first step for personalized treatment. The increased diagnostic load in the pathology laboratory, combined with the reported intra- and inter-variability in the assessment of biomarkers, has prompted the quest for reliable machine-based methods to be incorporated into the routine practice. Recently, Artificial Intelligence (AI) has made significant progress in the medical field, showing potential for clinical applications. Herein, we aim to systematically review the current research on AI in CRC image analysis. In histopathology, algorithms based on Deep Learning (DL) have the potential to assist in diagnosis, predict clinically relevant molecular phenotypes and microsatellite instability, identify histological features related to prognosis and correlated to metastasis, and assess the specific components of the tumor microenvironment.
Collapse
|
32
|
Cifci D, Foersch S, Kather JN. Artificial intelligence to identify genetic alterations in conventional histopathology. J Pathol 2022; 257:430-444. [PMID: 35342954 DOI: 10.1002/path.5898] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/10/2022]
Abstract
Precision oncology relies on the identification of targetable molecular alterations in tumor tissues. In many tumor types, a limited set of molecular tests is currently part of standard diagnostic workflows. However, universal testing for all targetable alterations, especially rare ones, is limited by the cost and availability of molecular assays. From 2017 to 2021, multiple studies have shown that artificial intelligence (AI) methods can predict the probability of specific genetic alterations directly from conventional hematoxylin and eosin (H&E) tissue slides. Although these methods are currently less accurate than gold-standard testing (e.g. immunohistochemistry, polymerase chain reaction or next-generation sequencing), they could be used as pre-screening tools to reduce the workload of genetic analyses. In this systematic literature review, we summarize the state of the art in predicting molecular alterations from H&E using AI. We found that AI methods perform reasonably well across multiple tumor types, although few algorithms have been broadly validated. In addition, we found that genetic alterations in FGFR, IDH, PIK3CA, BRAF, TP53 and DNA repair pathways are predictable from H&E in multiple tumor types, while many other genetic alterations have rarely been investigated or were only poorly predictable. Finally, we discuss the next steps for the implementation of AI-based surrogate tests in diagnostic workflows. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.,Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
33
|
Artificial Intelligence for Predicting Microsatellite Instability Based on Tumor Histomorphology: A Systematic Review. Int J Mol Sci 2022; 23:ijms23052462. [PMID: 35269607 PMCID: PMC8910565 DOI: 10.3390/ijms23052462] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 02/04/2023] Open
Abstract
Microsatellite instability (MSI)/defective DNA mismatch repair (dMMR) is receiving more attention as a biomarker for eligibility for immune checkpoint inhibitors in advanced diseases. However, due to high costs and resource limitations, MSI/dMMR testing is not widely performed. Some attempts are in progress to predict MSI/dMMR status through histomorphological features on H&E slides using artificial intelligence (AI) technology. In this study, the potential predictive role of this new methodology was reviewed through a systematic review. Studies up to September 2021 were searched through PubMed and Embase database searches. The design and results of each study were summarized, and the risk of bias for each study was evaluated. For colorectal cancer, AI-based systems showed excellent performance with the highest standard of 0.972; for gastric and endometrial cancers they showed a relatively low but satisfactory performance, with the highest standard of 0.81 and 0.82, respectively. However, analyzing the risk of bias, most studies were evaluated at high-risk. AI-based systems showed a high potential in predicting the MSI/dMMR status of different cancer types, and particularly of colorectal cancers. Therefore, a confirmation test should be required only for the results that are positive in the AI test.
Collapse
|
34
|
Ranasinghe R, Mathai M, Zulli A. A synopsis of modern - day colorectal cancer: Where we stand. Biochim Biophys Acta Rev Cancer 2022; 1877:188699. [DOI: 10.1016/j.bbcan.2022.188699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
|
35
|
Bustos A, Payá A, Torrubia A, Jover R, Llor X, Bessa X, Castells A, Carracedo Á, Alenda C. xDEEP-MSI: Explainable Bias-Rejecting Microsatellite Instability Deep Learning System in Colorectal Cancer. Biomolecules 2021; 11:1786. [PMID: 34944430 PMCID: PMC8699085 DOI: 10.3390/biom11121786] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023] Open
Abstract
The prediction of microsatellite instability (MSI) using deep learning (DL) techniques could have significant benefits, including reducing cost and increasing MSI testing of colorectal cancer (CRC) patients. Nonetheless, batch effects or systematic biases are not well characterized in digital histology models and lead to overoptimistic estimates of model performance. Methods to not only palliate but to directly abrogate biases are needed. We present a multiple bias rejecting DL system based on adversarial networks for the prediction of MSI in CRC from tissue microarrays (TMAs), trained and validated in 1788 patients from EPICOLON and HGUA. The system consists of an end-to-end image preprocessing module that tile samples at multiple magnifications and a tissue classification module linked to the bias-rejecting MSI predictor. We detected three biases associated with the learned representations of a baseline model: the project of origin of samples, the patient's spot and the TMA glass where each spot was placed. The system was trained to directly avoid learning the batch effects of those variables. The learned features from the bias-ablated model achieved maximum discriminative power with respect to the task and minimal statistical mean dependence with the biases. The impact of different magnifications, types of tissues and the model performance at tile vs patient level is analyzed. The AUC at tile level, and including all three selected tissues (tumor epithelium, mucin and lymphocytic regions) and 4 magnifications, was 0.87 ± 0.03 and increased to 0.9 ± 0.03 at patient level. To the best of our knowledge, this is the first work that incorporates a multiple bias ablation technique at the DL architecture in digital pathology, and the first using TMAs for the MSI prediction task.
Collapse
Affiliation(s)
- Aurelia Bustos
- AI Cancer Research Unit, MedBravo, 03560 Alicante, Spain;
| | - Artemio Payá
- Pathology Department, Alicante University General Hospital (HGUA), 03010 Alicante, Spain; (A.P.); (R.J.)
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | | | - Rodrigo Jover
- Pathology Department, Alicante University General Hospital (HGUA), 03010 Alicante, Spain; (A.P.); (R.J.)
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | - Xavier Llor
- Department of Medicine, Yale Cancer Center, Yale University, New Haven, CT 06511, USA;
| | - Xavier Bessa
- Gastroenterology Department, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain;
| | - Antoni Castells
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain;
| | - Ángel Carracedo
- Fundación Pública Gallega de Medicina Genómica (GMX), 15706 Santiago de Compostela, Spain;
| | - Cristina Alenda
- Pathology Department, Alicante University General Hospital (HGUA), 03010 Alicante, Spain; (A.P.); (R.J.)
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
36
|
Murchan P, Ó’Brien C, O’Connell S, McNevin CS, Baird AM, Sheils O, Ó Broin P, Finn SP. Deep Learning of Histopathological Features for the Prediction of Tumour Molecular Genetics. Diagnostics (Basel) 2021; 11:1406. [PMID: 34441338 PMCID: PMC8393642 DOI: 10.3390/diagnostics11081406] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Advanced diagnostics are enabling cancer treatments to become increasingly tailored to the individual through developments in immunotherapies and targeted therapies. However, long turnaround times and high costs of molecular testing hinder the widespread implementation of targeted cancer treatments. Meanwhile, gold-standard histopathological assessment carried out by a trained pathologist is widely regarded as routine and mandatory in most cancers. Recently, methods have been developed to mine hidden information from histopathological slides using deep learning applied to scanned and digitized slides; deep learning comprises a collection of computational methods which learn patterns in data in order to make predictions. Such methods have been reported to be successful in a variety of cancers for predicting the presence of biomarkers such as driver mutations, tumour mutational burden, and microsatellite instability. This information could prove valuable to pathologists and oncologists in clinical decision making for cancer treatment and triage for in-depth sequencing. In addition to identifying molecular features, deep learning has been applied to predict prognosis and treatment response in certain cancers. Despite reported successes, many challenges remain before the clinical implementation of such diagnostic strategies in the clinical setting is possible. This review aims to outline recent developments in the field of deep learning for predicting molecular genetics from histopathological slides, as well as to highlight limitations and pitfalls of working with histopathology slides in deep learning.
Collapse
Affiliation(s)
- Pierre Murchan
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland; (P.M.); (C.Ó.); (C.S.M.)
| | - Cathal Ó’Brien
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland; (P.M.); (C.Ó.); (C.S.M.)
- Department of Histopathology, St James’s Hospital, P.O. Box 580, James’s Street, D08 X4RX Dublin, Ireland
| | - Shane O’Connell
- School of Mathematics, Statistics, and Applied Mathematics, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.O.); (P.Ó.B.)
| | - Ciara S. McNevin
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland; (P.M.); (C.Ó.); (C.S.M.)
- Department of Medical Oncology, St James’s Hospital, D08 NHY1 Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D02 A440 Dublin, Ireland; (A.-M.B.); (O.S.)
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D02 A440 Dublin, Ireland; (A.-M.B.); (O.S.)
| | - Pilib Ó Broin
- School of Mathematics, Statistics, and Applied Mathematics, National University of Ireland Galway, H91 TK33 Galway, Ireland; (S.O.); (P.Ó.B.)
| | - Stephen P. Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland; (P.M.); (C.Ó.); (C.S.M.)
- Department of Histopathology, St James’s Hospital, P.O. Box 580, James’s Street, D08 X4RX Dublin, Ireland
| |
Collapse
|
37
|
Jang HJ, Song IH, Lee SH. Deep Learning for Automatic Subclassification of Gastric Carcinoma Using Whole-Slide Histopathology Images. Cancers (Basel) 2021; 13:3811. [PMID: 34359712 PMCID: PMC8345042 DOI: 10.3390/cancers13153811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Histomorphologic types of gastric cancer (GC) have significant prognostic values that should be considered during treatment planning. Because the thorough quantitative review of a tissue slide is a laborious task for pathologists, deep learning (DL) can be a useful tool to support pathologic workflow. In the present study, a fully automated approach was applied to distinguish differentiated/undifferentiated and non-mucinous/mucinous tumor types in GC tissue whole-slide images from The Cancer Genome Atlas (TCGA) stomach adenocarcinoma dataset (TCGA-STAD). By classifying small patches of tissue images into differentiated/undifferentiated and non-mucinous/mucinous tumor tissues, the relative proportion of GC tissue subtypes can be easily quantified. Furthermore, the distribution of different tissue subtypes can be clearly visualized. The patch-level areas under the curves for the receiver operating characteristic curves for the differentiated/undifferentiated and non-mucinous/mucinous classifiers were 0.932 and 0.979, respectively. We also validated the classifiers on our own GC datasets and confirmed that the generalizability of the classifiers is excellent. The results indicate that the DL-based tissue classifier could be a useful tool for the quantitative analysis of cancer tissue slides. By combining DL-based classifiers for various molecular and morphologic variations in tissue slides, the heterogeneity of tumor tissues can be unveiled more efficiently.
Collapse
Affiliation(s)
- Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - In-Hye Song
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Sung-Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| |
Collapse
|
38
|
Evaluating mismatch repair deficiency for solid tumor immunotherapy eligibility: immunohistochemistry versus microsatellite molecular testing. Hum Pathol 2021; 115:10-18. [PMID: 34052294 DOI: 10.1016/j.humpath.2021.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022]
Abstract
While many landmark solid tumor immunotherapy studies show clinical benefits for solid tumors with high microsatellite instability (MSI-H) and mismatch repair deficiency (dMMR), the methodologies focus only on confirmatory polymerase chain reaction (PCR) testing for MSI-H. Because some tumors are either dMMR or MSI-H but not the other, clinicians must choose between two testing methods for a broad patient population. We investigated the level of correlation between MMR protein immunohistochemistry (IHC) and microsatellite PCR testing results in 62 cancer patients. Thirty-five of the 62 cases (56.5%) were MSI-H by PCR, whereas 35 (56.5%) were dMMR by IHC. MMR IHC results correlated well with MSI PCR in 32 co-positive cases (91.4%) and 24 co-negative cases (88.9%). Six discrepant cases (9.7%) were identified, among which three were MSI-H and MMR intact, and three were dMMR and microsatellite stable. The results of this study highlight the implications of dMMR/MSI testing strategies on precision oncology. Co-testing with both MMR IHC and MSI PCR may be an effective screening strategy for evaluating immunotherapy eligibility status for solid tumors.
Collapse
|