1
|
Zhao Z, Wang H, Wu D, Zhu Q, Tan X, Hu S, Ge Y. PEDRA-EFB0: colorectal cancer prognostication using deep learning with patch embeddings and dual residual attention. Med Biol Eng Comput 2025; 63:1627-1647. [PMID: 39833600 DOI: 10.1007/s11517-025-03292-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
In computer-aided diagnosis systems, precise feature extraction from CT scans of colorectal cancer using deep learning is essential for effective prognosis. However, existing convolutional neural networks struggle to capture long-range dependencies and contextual information, resulting in incomplete CT feature extraction. To address this, the PEDRA-EFB0 architecture integrates patch embeddings and a dual residual attention mechanism for enhanced feature extraction and survival prediction in colorectal cancer CT scans. A patch embedding method processes CT scans into patches, creating positional features for global representation and guiding spatial attention computation. Additionally, a dual residual attention mechanism during the upsampling stage selectively combines local and global features, enhancing CT data utilization. Furthermore, this paper proposes a feature selection algorithm that combines autoencoders and entropy technology, encoding and compressing high-dimensional data to reduce redundant information and using entropy to assess the importance of features, thereby achieving precise feature selection. Experimental results indicate the PEDRA-EFB0 model outperforms traditional methods on colorectal cancer CT metrics, notably in C-index, BS, MCC, and AUC, enhancing survival prediction accuracy. Our code is freely available at https://github.com/smile0208z/PEDRA .
Collapse
Affiliation(s)
- Zihao Zhao
- School of Internet of Things Engineering, Jiangnan University, Wuxi, 214122, China
| | - Hao Wang
- School of Internet of Things Engineering, Jiangnan University, Wuxi, 214122, China
| | - Dinghui Wu
- Key Laboratory of Light Industry, Jiangnan University, Wuxi, 214122, China.
| | - Qibing Zhu
- Key Laboratory of Light Industry, Jiangnan University, Wuxi, 214122, China
| | - Xueping Tan
- School of Internet of Things Engineering, Jiangnan University, Wuxi, 214122, China
| | - Shudong Hu
- Radiol Dept, Jiangnan Univ, Affiliated Hosp, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Yuxi Ge
- Radiol Dept, Jiangnan Univ, Affiliated Hosp, Wuxi, 214122, Jiangsu, People's Republic of China
| |
Collapse
|
2
|
Beltzung F, Le VL, Molnar I, Boutault E, Darcha C, Le Loarer F, Kossai M, Saut O, Biau J, Penault-Llorca F, Chautard E. Leveraging Deep Learning for Immune Cell Quantification and Prognostic Evaluation in Radiotherapy-Treated Oropharyngeal Squamous Cell Carcinomas. J Transl Med 2025; 105:104094. [PMID: 39826685 DOI: 10.1016/j.labinv.2025.104094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The tumor microenvironment plays a critical role in cancer progression and therapeutic responsiveness, with the tumor immune microenvironment (TIME) being a key modulator. In head and neck squamous cell carcinomas (HNSCCs), immune cell infiltration significantly influences the response to radiotherapy (RT). A better understanding of the TIME in HNSCCs could help identify patients most likely to benefit from combining RT with immunotherapy. Standardized, cost-effective methods for studying TIME in HNSCCs are currently lacking. This study aims to leverage deep learning (DL) to quantify immune cell densities using immunohistochemistry in untreated oropharyngeal squamous cell carcinoma (OPSCC) biopsies of patients scheduled for curative RT and assess their prognostic value. We analyzed 84 pretreatment formalin-fixed paraffin-embedded tumor biopsies from OPSCC patients. Immunohistochemistry was performed for CD3, CD8, CD20, CD163, and FOXP3, and whole slide images were digitized for analysis using a U-Net-based DL model. Two quantification approaches were applied: a cell-counting method and an area-based method. These methods were applied to stained regions. The DL model achieved high accuracy in detecting stained cells across all biomarkers. Strong correlations were found between our DL pipeline, the HALO Image Analysis Platform, and the open-source QuPath software for estimating immune cell densities. Our DL pipeline provided an accurate and reproducible approach for quantifying immune cells in OPSCC. The area-based method demonstrated superior prognostic value for recurrence-free survival, when compared with the cell-counting method. Elevated densities of CD3, CD8, CD20, and FOXP3 were associated with improved recurrence-free survival, whereas CD163 showed no significant prognostic association. These results highlight the potential of DL in digital pathology for assessing TIME and predicting patient outcomes.
Collapse
Affiliation(s)
- Fanny Beltzung
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France.
| | - Van-Linh Le
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France; Department of Data and Digital Health, Bergonié Institute, Bordeaux, France
| | - Ioana Molnar
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Clinical Research Division, Clinical Research & Innovation Division, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Erwan Boutault
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France
| | - Claude Darcha
- Department of Pathology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - François Le Loarer
- Department of Pathology, Bergonié Institute, Bordeaux, France; Bordeaux Institute of Oncology (BRIC U1312), INSERM, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Myriam Kossai
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Olivier Saut
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France
| | - Julian Biau
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Radiation Therapy, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| |
Collapse
|
3
|
Huang Z, Yang E, Shen J, Gratzinger D, Eyerer F, Liang B, Nirschl J, Bingham D, Dussaq AM, Kunder C, Rojansky R, Gilbert A, Chang-Graham AL, Howitt BE, Liu Y, Ryan EE, Tenney TB, Zhang X, Folkins A, Fox EJ, Montine KS, Montine TJ, Zou J. A pathologist-AI collaboration framework for enhancing diagnostic accuracies and efficiencies. Nat Biomed Eng 2025; 9:455-470. [PMID: 38898173 DOI: 10.1038/s41551-024-01223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 05/03/2024] [Indexed: 06/21/2024]
Abstract
In pathology, the deployment of artificial intelligence (AI) in clinical settings is constrained by limitations in data collection and in model transparency and interpretability. Here we describe a digital pathology framework, nuclei.io, that incorporates active learning and human-in-the-loop real-time feedback for the rapid creation of diverse datasets and models. We validate the effectiveness of the framework via two crossover user studies that leveraged collaboration between the AI and the pathologist, including the identification of plasma cells in endometrial biopsies and the detection of colorectal cancer metastasis in lymph nodes. In both studies, nuclei.io yielded considerable diagnostic performance improvements. Collaboration between clinicians and AI will aid digital pathology by enhancing accuracies and efficiencies.
Collapse
Affiliation(s)
- Zhi Huang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Yang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeanne Shen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Frederick Eyerer
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke Liang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey Nirschl
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Bingham
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex M Dussaq
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian Kunder
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rebecca Rojansky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aubre Gilbert
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Brooke E Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Emily E Ryan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Troy B Tenney
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoming Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ann Folkins
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Edward J Fox
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen S Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - James Zou
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Zhou S, Xie Y, Feng X, Li Y, Shen L, Chen Y. Artificial intelligence in gastrointestinal cancer research: Image learning advances and applications. Cancer Lett 2025; 614:217555. [PMID: 39952597 DOI: 10.1016/j.canlet.2025.217555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
With the rapid advancement of artificial intelligence (AI) technologies, including deep learning, large language models, and neural networks, these methodologies are increasingly being developed and integrated into cancer research. Gastrointestinal tumors are characterized by complexity and heterogeneity, posing significant challenges for early detection, diagnostic accuracy, and the development of personalized treatment strategies. The application of AI in digestive oncology has demonstrated its transformative potential. AI not only alleviates the diagnostic burden on clinicians, but it improves tumor screening sensitivity, specificity, and accuracy. Additionally, AI aids the detection of biomarkers such as microsatellite instability and mismatch repair, supports intraoperative assessments of tumor invasion depth, predicts treatment responses, and facilitates the design of personalized treatment plans to potentially significantly enhance patient outcomes. Moreover, the integration of AI with multiomics analyses and imaging technologies has led to substantial advancements in foundational research on the tumor microenvironment. This review highlights the progress of AI in gastrointestinal oncology over the past 5 years with focus on early tumor screening, diagnosis, molecular marker identification, treatment planning, and prognosis predictions. We also explored the potential of AI to enhance medical imaging analyses to aid tumor detection and characterization as well as its role in automating and refining histopathological assessments.
Collapse
Affiliation(s)
- Shengyuan Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing, 102200, China.
| |
Collapse
|
5
|
Zhu X, Sun H, Wang Y, Hu G, Shao L, Zhang S, Liu F, Chi C, He K, Tang J, An Y, Tian J, Liu Z. Prediction of Lymph Node Metastasis in Colorectal Cancer Using Intraoperative Fluorescence Multi-Modal Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:1568-1580. [PMID: 40030456 DOI: 10.1109/tmi.2024.3510836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The diagnosis of lymph node metastasis (LNM) is essential for colorectal cancer (CRC) treatment. The primary method of identifying LNM is to perform frozen sections and pathologic analysis, but this method is labor-intensive and time-consuming. Therefore, combining intraoperative fluorescence imaging with deep learning (DL) methods can improve efficiency. The majority of recent studies only analyze uni-modal fluorescence imaging, which provides less semantic information. In this work, we mainly established a multi-modal fluorescence imaging feature fusion prediction (MFI-FFP) model combining white light, fluorescence, and pseudo-color imaging of lymph nodes for LNM prediction. Firstly, based on the properties of various modal imaging, distinct feature extraction networks are chosen for feature extraction, which could significantly enhance the complementarity of various modal information. Secondly, the multi-modal feature fusion (MFF) module, which combines global and local information, is designed to fuse the extracted features. Furthermore, a novel loss function is formulated to tackle the issue of imbalanced samples, challenges in differentiating samples, and enhancing sample variety. Lastly, the experiments show that the model has a higher area under the receiver operating characteristic (ROC) curve (AUC), accuracy (ACC), and F1 score than the uni-modal and bi-modal models and has a better performance compared to other efficient image classification networks. Our study demonstrates that the MFI-FFP model has the potential to help doctors predict LNM and shows its promise in medical image analysis.
Collapse
|
6
|
Zhu M, Zhai Z, Wang Y, Chen F, Liu R, Yang X, Zhao G. Advancements in the application of artificial intelligence in the field of colorectal cancer. Front Oncol 2025; 15:1499223. [PMID: 40071094 PMCID: PMC11893421 DOI: 10.3389/fonc.2025.1499223] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignant tumor in the digestive system. As reported in the 2020 global cancer statistics, CRC accounted for more than 1.9 million new cases and 935,000 deaths, making it the third most common cancer worldwide in terms of incidence and the second leading cause of cancer-related deaths globally. This poses a significant threat to global public health. Early screening methods, such as fecal occult blood tests, colonoscopies, and imaging techniques, are crucial for detecting early lesions and enabling timely intervention before cancer becomes invasive. Early detection greatly enhances treatment possibilities, such as surgery, radiation therapy, and chemotherapy, with surgery being the main approach for treating early-stage CRC. In this context, artificial intelligence (AI) has shown immense potential in revolutionizing CRC management, serving as one of the most effective screening tools. AI, utilizing machine learning (ML) and deep learning (DL) algorithms, improves early detection, diagnosis, and treatment by processing large volumes of medical data, uncovering hidden patterns, and forecasting disease development. DL, a more advanced form of ML, simulates the brain's processing power, enhancing the accuracy of tumor detection, differentiation, and prognosis predictions. These innovations offer the potential to revolutionize cancer care by boosting diagnostic accuracy, refining treatment approaches, and ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Mengying Zhu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Zhenzhu Zhai
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yue Wang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Fang Chen
- Department of Gynecology, People’s Hospital of Liaoning Province, Shenyang, China
| | - Ruibin Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Xiaoquan Yang
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guohua Zhao
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
7
|
Marmé F, Krieghoff-Henning EI, Kiehl L, Wies C, Hauke J, Hahnen E, Harter P, Schouten PC, Brodkorb T, Kayali M, Heitz F, Zamagni C, González-Martin A, Treilleux I, Kommoss S, Prieske K, Gaiser T, Fröhling S, Ray-Coquard I, Pujade-Lauraine E, Brinker TJ. Predicting benefit from PARP inhibitors using deep learning on H&E-stained ovarian cancer slides. Eur J Cancer 2025; 216:115199. [PMID: 39742559 DOI: 10.1016/j.ejca.2024.115199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/03/2025]
Abstract
PURPOSE Ovarian cancer patients with a Homologous Recombination Deficiency (HRD) often benefit from polyadenosine diphosphate-ribose polymerase (PARP) inhibitor maintenance therapy after response to platinum-based chemotherapy. HR status is currently analyzed via complex molecular tests. Predicting benefit from PARP inhibitors directly on histological whole slide images (WSIs) could be a fast and cheap alternative. PATIENTS AND METHODS We trained a Deep Learning (DL) model on H&E stained WSIs with "shrunken centroid" (SC) based HRD ground truth using the AGO-TR1 cohort (n = 208: 108 training, 100 test) and tested its ability to predict HRD as evaluated by the Myriad classifier and the benefit from olaparib in the PAOLA-1 cohort (n = 447) in a blinded manner. RESULTS In contrast to the HRD prediction AUROC of 72 % on hold-out, our model only yielded an AUROC of 57 % external. Kaplan-Meier analysis showed that progression free survival (PFS) in the PARP inhibitor treated PAOLA-1 patients was significantly improved in the HRD positive group as defined by our model, but not in the HRD negative group. PFS improvement in PARP inhibitor-treated patients was substantially longer in our HRD positive group, hinting at a biologically meaningful prediction of benefit from PARP inhibitors. CONCLUSION Together, our results indicate that it might be possible to generate a predictor of benefit from PARP inhibitors based on the DL-mediated analysis of WSIs. However, further studies with larger cohorts and further methodological improvements will be necessary to generate a predictor with clinically useful accuracy across independent patient cohorts.
Collapse
Affiliation(s)
- Frederik Marmé
- University Hospital Mannheim, Department of Obstetrics and Gynaecology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany and DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Eva I Krieghoff-Henning
- Division of Digital Prevention, Diagnostics and Therapy Guidance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lennard Kiehl
- Division of Digital Prevention, Diagnostics and Therapy Guidance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Wies
- Division of Digital Prevention, Diagnostics and Therapy Guidance, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, University Heidelberg, Heidelberg, Germany
| | - Jan Hauke
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University Hospital Cologne, Cologne, Nordrhein-Westfalen, Germany
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University Hospital Cologne, Cologne, Nordrhein-Westfalen, Germany
| | - Philipp Harter
- Ev. Kliniken Essen Mitte, Essen, and Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) Studiengruppe, Germany
| | - Philip C Schouten
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom; Department of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tobias Brodkorb
- University Hospital Mannheim, Department of Obstetrics and Gynaecology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany and DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Mohamad Kayali
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University Hospital Cologne, Cologne, Nordrhein-Westfalen, Germany
| | - Florian Heitz
- Ev. Kliniken Essen Mitte, Essen, and Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) Studiengruppe, Germany
| | - Claudio Zamagni
- IRCCS Azienda Ospedaliero-universitaria di Bologna, and MITO, Italy
| | | | | | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Katharina Prieske
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Gaiser
- Department of Pathology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany
| | - Stefan Fröhling
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg Germany and German Cancer Consortium (DKTK), Heidelberg, Germany; Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelbergg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | | - Eric Pujade-Lauraine
- Association de Recherche sur les CAncers dont GYnécologiques (ARCAGY)-GINECO, Paris, France
| | - Titus J Brinker
- Division of Digital Prevention, Diagnostics and Therapy Guidance, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
8
|
Meneghetti AR, Hernández ML, Kuehn JP, Löck S, Carrero ZI, Perez-Lopez R, Bressem K, Brinker TK, Pearson AT, Truhn D, Nebelung S, Kather JN. End-to-end prediction of clinical outcomes in head and neck squamous cell carcinoma with foundation model-based multiple instance learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.22.25320517. [PMID: 39974018 PMCID: PMC11839013 DOI: 10.1101/2025.01.22.25320517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Foundation models (FMs) show promise in medical AI by learning flexible features from large datasets, potentially surpassing handcrafted radiomics. Outcome prediction of head and neck squamous cell carcinomas (HNSCC) with FMs using routine imaging remains unexplored. Purpose To evaluate end-to-end FM-based multiple instance learning (MIL) for 2-year overall survival (OS), locoregional control (LRC), and freedom from distant metastasis (FFDM) prediction and risk group stratification using pretreatment CT scans in HNSCC. Materials and Methods We analyzed data of 2485 patients from three retrospective HNSCC cohorts (RADCURE, HN1, HN-PET-CT), treated between 2004 and 2017 with available pre-treatment CTs and primary gross tumor volume (GTVp) segmentations. The RADCURE cohort was split into training (n=1464) and test (N=606), with HN1 (n=131) and HN-PET-CT (n=284) as additional test cohorts. FM-based MIL models (2D, multiview and 3D) for 2-year endpoint prediction and risk stratification wre evaluated based on area under the receiver operator curve (AUROC) and Kaplan-Meier (KM) with hazard ratios (HR), compared with radiomics and assessed for multimodal enhancement with clinical baselines. Results 2D MIL models achieved 2-year test AUROCs of 0.75-0.84 (OS), 0.66-0.75 (LRC) and 0.71-0.78 (FFDM), outperforming multiview and 3D MIL (AUROCs: 0.50-0.77, p≥0.15) and comparable or superior to radiomics (AUROCs: 0.64-0.74, p≥0.012). Significant stratification was observed (HRs: 2.14-4.77, p≤0.039). Multimodal enhancement of 2-year OS/FFDM (AUROCs: 0.82-0.87, p≤0.018) was observed for patients without human papilloma virus positive (HPV+) tumors. Conclusion FM-based MIL demonstrates promise in HNSCC risk prediction, showing similar or superior performance to radiomics and enhancing clinical baselines in non-HPV+ patients.
Collapse
Affiliation(s)
- Asier Rabasco Meneghetti
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marta Ligero Hernández
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Jens-Peter Kuehn
- Institute and Policlinic for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Steffen Löck
- German Cancer Consortium (DKTK), Partner site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universitat Dresden; Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Zunamys Itzel Carrero
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Keno Bressem
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, School of Medicine and Health, Klinikum rechts der Isar, TUM University Hospital, Ismaninger Str. 22, 81675 Munich
- Department of Cardiovascular Radiology and Nuclear Medicine, Technical University of Munich, School of Medicine and Health, German Heart Center, TUM University Hospital, Lazarethstr. 36, 80636, Munich
| | - Titus K Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), INF 223, 69120 Heidelberg, Germany
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Sven Nebelung
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Liu H, Zou Q, Zhang H, Ma X. Development of a prediction model based on Hemoglobin, Albumin, Lymphocyte count, and Platelet-score for lymph node metastasis in rectal cancer. Eur J Cancer Prev 2025:00008469-990000000-00204. [PMID: 39835527 DOI: 10.1097/cej.0000000000000954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
This study aimed to evaluate the ability of the preoperative Hemoglobin, Albumin, Lymphocyte count, and Platelet (HALP) score to predict lymph node metastasis (LNM) in patients with rectal cancer (RC) and improve prediction accuracy by incorporating clinical parameters. Data from 263 patients with RC were analyzed. The receiver operating characteristic (ROC) curve was used to determine the optimal cutoff value (OCV) for the HALP score in predicting LNM. Based on this cutoff value, patients were divided into two groups. A baseline analysis was conducted to identify independent factors linked to LNM. A support vector machine (SVM) prediction model was developed, and its performance was evaluated using ROC, calibration curves, decision curve analysis, and Kolmogorov-Smirnov curve. The OCV for HALP score was 45.979. Patients were then classified into a low HALP group (n = 182) and a high HALP group (n = 81). The analysis found 21 clinical factors significantly associated with LNM. Among them, the key risk factors included high inflammatory status, poor nutritional condition, and a low HALP score. The SVM model incorporated these factors and showed robust predictive performance, with area under the curve values of 0.897, 0.813, and 0.750 for the training, validation, and testing datasets, respectively. The HALP score was significantly associated with LNM in RC patients. A machine learning model integrating the HALP score and inflammatory markers may be an effective tool for predicting LNM in RC.
Collapse
Affiliation(s)
- Huanhui Liu
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | | | | | | |
Collapse
|
10
|
Zheng Q, Jiao P, Yang R, Fan J, Liu Y, Yang X, Yuan J, Chen Z, Liu X. Deep learning-based lymph node metastasis status predicts prognosis from muscle-invasive bladder cancer histopathology. World J Urol 2025; 43:65. [PMID: 39792275 DOI: 10.1007/s00345-025-05440-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE To develop a deep learning (DL) model based on primary tumor tissue to predict the lymph node metastasis (LNM) status of muscle invasive bladder cancer (MIBC), while validating the prognostic value of the predicted aiN score in MIBC patients. METHODS A total of 323 patients from The Cancer Genome Atlas (TCGA) were used as the training and internal validation set, with image features extracted using a visual encoder called UNI. We investigated the ability to predict LNM status while assessing the prognostic value of aiN score. External validation was conducted on 139 patients from Renmin Hospital of Wuhan University (RHWU; Wuhan, China). RESULTS The DL model achieved area under the receiver operating characteristic curves of 0.79 (95% confidence interval [CI], 0.69-0.88) in the internal validation set for predicting LNM status, and 0.72 (95% CI, 0.68-0.75) in the external validation set. In multivariable Cox analysis, the model-predicted aiN score emerged as an independent predictor of survival for MIBC patients, with a hazard ratio of 1.608 (95% CI, 1.128-2.291; p = 0.008) in the TCGA cohort and 2.746 (95% CI, 1.486-5.076; p < 0.001) in the RHWU cohort. Additionally, the aiN score maintained prognostic value across different subgroups. CONCLUSION In this study, DL-based image analysis showed promising results by directly extracting relevant prognostic information from H&E-stained histology to predict the LNM status of MIBC patients. It might be used for personalized management of MIBC patients following prospective validation in the future.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Junjie Fan
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Trusted Computing and Information Assurance Laboratory, Institute of Software, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yunxun Liu
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Xiangxiang Yang
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhang Zhi-dong Road, Wuhan, Hubei, 430060, P.R. China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
11
|
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
|
12
|
Wang Y, Zhao H, Fu P, Tian L, Su Y, Lyu Z, Gu W, Wang Y, Liu S, Wang X, Zheng H, Du J, Zhang R. Preoperative prediction of lymph node metastasis in colorectal cancer using 18F-FDG PET/CT peritumoral radiomics analysis. Med Phys 2024; 51:5214-5225. [PMID: 38801340 DOI: 10.1002/mp.17193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Radiomics has been used in the diagnosis of tumor lymph node metastasis (LNM). However, to date, most studies have been based on intratumoral radiomics. Few studies have focused on the use of 18F-fluorodeoxyglucose positron emission computed tomography (18F-FDG PET/CT) peritumoral radiomics for the diagnosis of LNM in colorectal cancer (CRC). PURPOSE Determining the value of radiomics features extracted from 18F-FDG PET/CT images of the peritumoral region in predicting LNM in patients with CRC. METHODS The clinical data and preoperative 18F-FDG PET/CT images of 244 CRC patients were retrospectively analyzed. Intratumoral and peritumoral radiomics features were screened using the mutual information method, and least absolute shrinkage and selection operator regression. Based on the selected radiomics features, a radiomics score (Rad-score) was calculated, and independent risk factors obtained from univariate and multivariate logistic regression analyses were used to construct clinical and combined (Radiomics + Clinical) models. The performance of these models was evaluated using the DeLong test, while their clinical utility was assessed by decision curve analysis. Finally, a nomogram was constructed to visualize the predictive model. RESULTS The most optimal set of features retained by the feature filtering process were all peritumoral radiomic features. Carcinoembryonic antigen levels, PET/CT-reported lymph node status and Rad-score were found to be independent risk factors for LNM. All three LNM risk assessment models exhibited good predictive performance, with the combined model showing the best classification results, with areas under the curve of 0.85 and 0.76 in the training and validation groups, respectively. The DeLong test revealed that the performance of the combined model was superior to that of the clinical and radiomics models in both the training and validation groups, although this difference was only statistically significant in the training group. DCA indicated that the combined model displayed better clinical utility. CONCLUSIONS 18F-FDG PET/CT peritumoral radiomics is uniquely suited to predict the presence of LNM in patients with CRC. In particular, the predictive efficacy of LNM for precision therapy and individualized patient management can be improved by using a combination of clinical risk factors.
Collapse
Affiliation(s)
- Yan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Fu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Tian
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yexin Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhehao Lyu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Han Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Du
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui Zhang
- Department of Magnetic Resonance, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, China
| |
Collapse
|
13
|
Cheng M, Zhang H, Huang W, Li F, Gao J. Deep Learning Radiomics Analysis of CT Imaging for Differentiating Between Crohn's Disease and Intestinal Tuberculosis. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1516-1528. [PMID: 38424279 PMCID: PMC11300798 DOI: 10.1007/s10278-024-01059-0] [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: 11/23/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
This study aimed to develop and evaluate a CT-based deep learning radiomics model for differentiating between Crohn's disease (CD) and intestinal tuberculosis (ITB). A total of 330 patients with pathologically confirmed as CD or ITB from the First Affiliated Hospital of Zhengzhou University were divided into the validation dataset one (CD: 167; ITB: 57) and validation dataset two (CD: 78; ITB: 28). Based on the validation dataset one, the synthetic minority oversampling technique (SMOTE) was adopted to create balanced dataset as training data for feature selection and model construction. The handcrafted and deep learning (DL) radiomics features were extracted from the arterial and venous phases images, respectively. The interobserver consistency analysis, Spearman's correlation, univariate analysis, and the least absolute shrinkage and selection operator (LASSO) regression were used to select features. Based on extracted multi-phase radiomics features, six logistic regression models were finally constructed. The diagnostic performances of different models were compared using ROC analysis and Delong test. The arterial-venous combined deep learning radiomics model for differentiating between CD and ITB showed a high prediction quality with AUCs of 0.885, 0.877, and 0.800 in SMOTE dataset, validation dataset one, and validation dataset two, respectively. Moreover, the deep learning radiomics model outperformed the handcrafted radiomics model in same phase images. In validation dataset one, the Delong test results indicated that there was a significant difference in the AUC of the arterial models (p = 0.037), while not in venous and arterial-venous combined models (p = 0.398 and p = 0.265) as comparing deep learning radiomics models and handcrafted radiomics models. In our study, the arterial-venous combined model based on deep learning radiomics analysis exhibited good performance in differentiating between CD and ITB.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Medical Information, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Hanyue Zhang
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Fei Li
- School of Cyber Science and Engineering, Wuhan University, Wuhan, 430072, China
| | - Jianbo Gao
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
14
|
Sung YN, Lee H, Kim E, Jung WY, Sohn JH, Lee YJ, Keum B, Ahn S, Lee SH. Interpretable deep learning model to predict lymph node metastasis in early gastric cancer using whole slide images. Am J Cancer Res 2024; 14:3513-3522. [PMID: 39113867 PMCID: PMC11301296 DOI: 10.62347/rjbh6076] [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: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
In early gastric cancer (EGC), the presence of lymph node metastasis (LNM) is a crucial factor for determining the treatment options. Endoscopic resection is used for treatment of EGC with minimal risk of LNM. However, owing to the lack of definitive criteria for identifying patients who require additional surgery, some patients undergo unnecessary additional surgery. Considering that histopathologic patterns are significant factor for predicting lymph node metastasis in gastric cancer, we aimed to develop a machine learning algorithm which can predict LNM status using hematoxylin and eosin (H&E)-stained images. The images were obtained from several institutions. Our pipeline comprised two sequential approaches including a feature extractor and a risk classifier. For the feature extractor, a segmentation network (DeepLabV3+) was trained on 243 WSIs across three datasets to differentiate each histological subtype. The risk classifier was trained with XGBoost using 70 morphological features inferred from the trained feature extractor. The trained segmentation network, the feature extractor, achieved high performance, with pixel accuracies of 0.9348 and 0.8939 for the internal and external datasets in patch level, respectively. The risk classifier achieved an overall AUC of 0.75 in predicting LNM status. Remarkably, one of the datasets also showed a promising result with an AUC of 0.92. This is the first multi-institution study to develop machine learning algorithm for predicting LNM status in patients with EGC using H&E-stained histopathology images. Our findings have the potential to improve the selection of patients who require surgery among those with EGC showing high-risk histological features.
Collapse
Affiliation(s)
- You-Na Sung
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Hyeseong Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| | - Eunsu Kim
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| | - Woon Yong Jung
- Department of Pathology, Hanyang University Guri Hospital, College of Medicine, Hanyang UniversityGuri, South Korea
| | - Jin-Hee Sohn
- Department of Pathology, Samkwang Medical LaboratoriesSeoul, South Korea
| | - Yoo Jin Lee
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
| | - Sangjeong Ahn
- Department of Pathology, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
- Artificial Intelligence Center, Korea University Anam Hospital, College of Medicine, Korea UniversitySeoul, South Korea
- Department of Medical Informatics, College of Medicine, Korea UniversitySeoul, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic UniversitySeoul, South Korea
| |
Collapse
|
15
|
Wang D, He X, Huang C, Li W, Li H, Huang C, Hu C. Magnetic resonance imaging-based radiomics and deep learning models for predicting lymph node metastasis of squamous cell carcinoma of the tongue. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:214-224. [PMID: 38378316 DOI: 10.1016/j.oooo.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/14/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024]
Abstract
OBJECTIVE This study aimed to establish a combined method of radiomics and deep learning (DL) in magnetic resonance imaging (MRI) to predict lymph node metastasis (LNM) preoperatively in patients with squamous cell carcinoma of the tongue. STUDY DESIGN In total, MR images of 196 patients with lingual squamous cell carcinoma were divided into training (n = 156) and test (n = 40) cohorts. Radiomics and DL features were extracted from MR images and selected to construct machine learning models. A DL radiomics nomogram was established via multivariate logistic regression by incorporating the radiomics signature, the DL signature, and MRI-reported LN status. RESULTS Nine radiomics and 3 DL features were selected. In the radiomics test cohort, the multilayer perceptron model performed best with an area under the receiver operating characteristic curve (AUC) of 0.747, but in the DL cohort, the best model (logistic regression) performed less well (AUC = 0.655). The DL radiomics nomogram showed good calibration and performance with an AUC of 0.934 (outstanding discrimination ability) in the training cohort and 0.757 (acceptable discrimination ability) in the test cohort. The decision curve analysis demonstrated that the nomogram could offer more net benefit than a single radiomics or DL signature. CONCLUSION The DL radiomics nomogram exhibited promising performance in predicting LNM, which facilitates personalized treatment of tongue cancer.
Collapse
Affiliation(s)
- Dawei Wang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao He
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunming Huang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqiang Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haosen Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cicheng Huang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
16
|
Kurz A, Müller H, Kather JN, Schneider L, Bucher TC, Brinker TJ. 3-Dimensional Reconstruction From Histopathological Sections: A Systematic Review. J Transl Med 2024; 104:102049. [PMID: 38513977 DOI: 10.1016/j.labinv.2024.102049] [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: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Although pathological tissue analysis is typically performed on single 2-dimensional (2D) histologic reference slides, 3-dimensional (3D) reconstruction from a sequence of histologic sections could provide novel opportunities for spatial analysis of the extracted tissue. In this review, we analyze recent works published after 2018 and report information on the extracted tissue types, the section thickness, and the number of sections used for reconstruction. By analyzing the technological requirements for 3D reconstruction, we observe that software tools exist, both free and commercial, which include the functionality to perform 3D reconstruction from a sequence of histologic images. Through the analysis of the most recent works, we provide an overview of the workflows and tools that are currently used for 3D reconstruction from histologic sections and address points for future work, such as a missing common file format or computer-aided analysis of the reconstructed model.
Collapse
Affiliation(s)
- Alexander Kurz
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heimo Müller
- Diagnostics and Research Institute for Pathology, Medical University of Graz, Graz, Austria
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea C Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
Hetz MJ, Bucher TC, Brinker TJ. Multi-domain stain normalization for digital pathology: A cycle-consistent adversarial network for whole slide images. Med Image Anal 2024; 94:103149. [PMID: 38574542 DOI: 10.1016/j.media.2024.103149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/11/2023] [Accepted: 03/20/2024] [Indexed: 04/06/2024]
Abstract
The variation in histologic staining between different medical centers is one of the most profound challenges in the field of computer-aided diagnosis. The appearance disparity of pathological whole slide images causes algorithms to become less reliable, which in turn impedes the wide-spread applicability of downstream tasks like cancer diagnosis. Furthermore, different stainings lead to biases in the training which in case of domain shifts negatively affect the test performance. Therefore, in this paper we propose MultiStain-CycleGAN, a multi-domain approach to stain normalization based on CycleGAN. Our modifications to CycleGAN allow us to normalize images of different origins without retraining or using different models. We perform an extensive evaluation of our method using various metrics and compare it to commonly used methods that are multi-domain capable. First, we evaluate how well our method fools a domain classifier that tries to assign a medical center to an image. Then, we test our normalization on the tumor classification performance of a downstream classifier. Furthermore, we evaluate the image quality of the normalized images using the Structural similarity index and the ability to reduce the domain shift using the Fréchet inception distance. We show that our method proves to be multi-domain capable, provides a very high image quality among the compared methods, and can most reliably fool the domain classifier while keeping the tumor classifier performance high. By reducing the domain influence, biases in the data can be removed on the one hand and the origin of the whole slide image can be disguised on the other, thus enhancing patient data privacy.
Collapse
Affiliation(s)
- Martin J Hetz
- Division of Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea-Clara Bucher
- Division of Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Titus J Brinker
- Division of Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
18
|
Xu Y, Guo J, Yang N, Zhu C, Zheng T, Zhao W, Liu J, Song J. Predicting rectal cancer prognosis from histopathological images and clinical information using multi-modal deep learning. Front Oncol 2024; 14:1353446. [PMID: 38690169 PMCID: PMC11060749 DOI: 10.3389/fonc.2024.1353446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Objective The objective of this study was to provide a multi-modal deep learning framework for forecasting the survival of rectal cancer patients by utilizing both digital pathological images data and non-imaging clinical data. Materials and methods The research included patients diagnosed with rectal cancer by pathological confirmation from January 2015 to December 2016. Patients were allocated to training and testing sets in a randomized manner, with a ratio of 4:1. The tissue microarrays (TMAs) and clinical indicators were obtained. Subsequently, we selected distinct deep learning models to individually forecast patient survival. We conducted a scanning procedure on the TMAs in order to transform them into digital pathology pictures. Additionally, we performed pre-processing on the clinical data of the patients. Subsequently, we selected distinct deep learning algorithms to conduct survival prediction analysis using patients' pathological images and clinical data, respectively. Results A total of 292 patients with rectal cancer were randomly allocated into two groups: a training set consisting of 234 cases, and a testing set consisting of 58 instances. Initially, we make direct predictions about the survival status by using pre-processed Hematoxylin and Eosin (H&E) pathological images of rectal cancer. We utilized the ResNest model to extract data from histopathological images of patients, resulting in a survival status prediction with an AUC (Area Under the Curve) of 0.797. Furthermore, we employ a multi-head attention fusion (MHAF) model to combine image features and clinical features in order to accurately forecast the survival rate of rectal cancer patients. The findings of our experiment show that the multi-modal structure works better than directly predicting from histopathological images. It achieves an AUC of 0.837 in predicting overall survival (OS). Conclusions Our study highlights the potential of multi-modal deep learning models in predicting survival status from histopathological images and clinical information, thus offering valuable insights for clinical applications.
Collapse
Affiliation(s)
- Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiedong Guo
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Na Yang
- Artificial Intelligence Unit, Department of Medical Equipment Management, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Can Zhu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianlei Zheng
- Artificial Intelligence Unit, Department of Medical Equipment Management, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Weiguo Zhao
- Artificial Intelligence Unit, Department of Medical Equipment Management, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jia Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
19
|
Wang X, Li H, Chen H, Fang K, Chang X. Overexpression of circulating CD38+ NK cells in colorectal cancer was associated with lymph node metastasis and poor prognosis. Front Oncol 2024; 14:1309785. [PMID: 38463232 PMCID: PMC10921414 DOI: 10.3389/fonc.2024.1309785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Lymph node metastasis (LNM) is a critical prognostic factor for colorectal cancer (CRC). Due to the potential influence of immune system on CRC progression, investigation into lymphocyte subsets as clinical markers has gained attention. The objective of this study was to assess the capability of lymphocyte subsets in evaluating the lymph node status and prognosis of CRC. Methods Lymphocyte subsets, including T cells (CD3+), natural killer cells (NK, CD3- CD56+), natural killer-like T cells (NK-like T, CD3+ CD56+), CD38+ NK cells (CD3- CD56+ CD38+) and CD38+ NK-like T cells (CD3+ CD56+ CD38+), were detected by flow cytometry. Univariate and multivariate analyses were used to assess the risk factors of LNM. The prognostic role of parameters was evaluated by survival analysis. Results The proportion of CD38+ NK cells within the NK cell population was significantly higher in LNM-positive patients (p <0.0001). However, no significant differences were observed in the proportions of other lymphocyte subsets. Poorer histologic grade (odds ratio [OR] =4.76, p =0.03), lymphovascular invasion (LVI) (OR =22.38, p <0.01), and CD38+ NK cells (high) (OR =4.54, p <0.01) were identified as independent risk factors for LNM. Furthermore, high proportion of CD38+ NK cells was associated with poor prognosis of CRC patients (HR=2.37, p =0.03). Conclusions It was demonstrated that the proportion of CD38+ NK cells was a marker overexpressed in LNM-positive patients compared with LNM-negative patients. Moreover, an elevated proportion of CD38+ NK cells is a risk factor for LNM and poor prognosis in CRC.
Collapse
Affiliation(s)
- Xueling Wang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huixian Chen
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kehua Fang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Chang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Jiang W, Wang H, Dong X, Zhao Y, Long C, Chen D, Yan B, Cheng J, Lin Z, Zhuo S, Wang H, Yan J. Association of the pathomics-collagen signature with lymph node metastasis in colorectal cancer: a retrospective multicenter study. J Transl Med 2024; 22:103. [PMID: 38273371 PMCID: PMC10811897 DOI: 10.1186/s12967-024-04851-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) is a prognostic biomarker and affects therapeutic selection in colorectal cancer (CRC). Current evaluation methods are not adequate for estimating LNM in CRC. H&E images contain much pathological information, and collagen also affects the biological behavior of tumor cells. Hence, the objective of the study is to investigate whether a fully quantitative pathomics-collagen signature (PCS) in the tumor microenvironment can be used to predict LNM. METHODS Patients with histologically confirmed stage I-III CRC who underwent radical surgery were included in the training cohort (n = 329), the internal validation cohort (n = 329), and the external validation cohort (n = 315). Fully quantitative pathomics features and collagen features were extracted from digital H&E images and multiphoton images of specimens, respectively. LASSO regression was utilized to develop the PCS. Then, a PCS-nomogram was constructed incorporating the PCS and clinicopathological predictors for estimating LNM in the training cohort. The performance of the PCS-nomogram was evaluated via calibration, discrimination, and clinical usefulness. Furthermore, the PCS-nomogram was tested in internal and external validation cohorts. RESULTS By LASSO regression, the PCS was developed based on 11 pathomics and 9 collagen features. A significant association was found between the PCS and LNM in the three cohorts (P < 0.001). Then, the PCS-nomogram based on PCS, preoperative CEA level, lymphadenectasis on CT, venous emboli and/or lymphatic invasion and/or perineural invasion (VELIPI), and pT stage achieved AUROCs of 0.939, 0.895, and 0.893 in the three cohorts. The calibration curves identified good agreement between the nomogram-predicted and actual outcomes. Decision curve analysis indicated that the PCS-nomogram was clinically useful. Moreover, the PCS was still an independent predictor of LNM at station Nos. 1, 2, and 3. The PCS nomogram displayed AUROCs of 0.849-0.939 for the training cohort, 0.837-0.902 for the internal validation cohort, and 0.851-0.895 for the external validation cohorts in the three nodal stations. CONCLUSIONS This study proposed that PCS integrating pathomics and collagen features was significantly associated with LNM, and the PCS-nomogram has the potential to be a useful tool for predicting individual LNM in CRC patients.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- School of Science, Jimei University, Xiamen, Fujian, 361021, People's Republic of China
| | - Huaiming Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Xiaoyu Dong
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, People's Republic of China
| | - Chenyan Long
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530000, People's Republic of China
| | - Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Botao Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Zexi Lin
- School of Science, Jimei University, Xiamen, Fujian, 361021, People's Republic of China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, Fujian, 361021, People's Republic of China.
| | - Hui Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510655, People's Republic of China.
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China.
- Department of Gastrointestinal Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People's Republic of China.
| |
Collapse
|
21
|
Solovev IA. [Artificial intelligence in pathological anatomy]. Arkh Patol 2024; 86:65-71. [PMID: 38591909 DOI: 10.17116/patol20248602165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
The review presents key concepts and global developments in the field of artificial intelligence used in pathological anatomy. The work examines two types of artificial intelligence (AI): weak and strong ones. A review of experimental algorithms using both deep machine learning and computer vision technologies to work with WSI images of preparations, diagnose and make a prognosis for various malignant neoplasms is carried out. It has been established that weak artificial intelligence at this stage of development of computer (digital) pathological anatomy shows significantly better results in speeding up and refining diagnostic procedures than strong artificial intelligence having signs of general intelligence. The article also discusses three options for the further development of AI assistants for pathologists based on the technologies of large language models (strong AI) ChatGPT (PathAsst), Flan-PaLM2 and LIMA. As a result of the analysis of the literature, key problems in the field were identified: the equipment of pathology institutions, the lack of experts in training neural networks, the lack of strict criteria for the clinical viability of AI diagnostic technologies.
Collapse
Affiliation(s)
- I A Solovev
- Pitirim Sorokin Syktyvkar State University, Syktyvkar, Russia
| |
Collapse
|
22
|
Marmé F, Krieghoff-Henning E, Gerber B, Schmitt M, Zahm DM, Bauerschlag D, Forstbauer H, Hildebrandt G, Ataseven B, Brodkorb T, Denkert C, Stachs A, Krug D, Heil J, Golatta M, Kühn T, Nekljudova V, Gaiser T, Schönmehl R, Brochhausen C, Loibl S, Reimer T, Brinker TJ. Deep learning to predict breast cancer sentinel lymph node status on INSEMA histological images. Eur J Cancer 2023; 195:113390. [PMID: 37890350 DOI: 10.1016/j.ejca.2023.113390] [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: 09/11/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Sentinel lymph node (SLN) status is a clinically important prognostic biomarker in breast cancer and is used to guide therapy, especially for hormone receptor-positive, HER2-negative cases. However, invasive lymph node staging is increasingly omitted before therapy, and studies such as the randomised Intergroup Sentinel Mamma (INSEMA) trial address the potential for further de-escalation of axillary surgery. Therefore, it would be helpful to accurately predict the pretherapeutic sentinel status using medical images. METHODS Using a ResNet 50 architecture pretrained on ImageNet and a previously successful strategy, we trained deep learning (DL)-based image analysis algorithms to predict sentinel status on hematoxylin/eosin-stained images of predominantly luminal, primary breast tumours from the INSEMA trial and three additional, independent cohorts (The Cancer Genome Atlas (TCGA) and cohorts from the University hospitals of Mannheim and Regensburg), and compared their performance with that of a logistic regression using clinical data only. Performance on an INSEMA hold-out set was investigated in a blinded manner. RESULTS None of the generated image analysis algorithms yielded significantly better than random areas under the receiver operating characteristic curves on the test sets, including the hold-out test set from INSEMA. In contrast, the logistic regression fitted on the Mannheim cohort retained a better than random performance on INSEMA and Regensburg. Including the image analysis model output in the logistic regression did not improve performance further on INSEMA. CONCLUSIONS Employing DL-based image analysis on histological slides, we could not predict SLN status for unseen cases in the INSEMA trial and other predominantly luminal cohorts.
Collapse
Affiliation(s)
- Frederik Marmé
- Department of Obstetrics and Gynaecology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany
| | - Eva Krieghoff-Henning
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernd Gerber
- Department of Obstetrics and Gynecology, University Hospital of Rostock, Rostock, Germany
| | - Max Schmitt
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Dirk Bauerschlag
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | | | - Guido Hildebrandt
- Department of Radiotherapy, University Medicine Rostock, Rostock, Germany
| | - Beyhan Ataseven
- Department of Gynecology, Gynecologic Oncology and Obstetrics, Klinikum Lippe, Bielefeld University, Medical School and University Medical Center East Westphalia-Lippe, Bielefeld, Germany
| | - Tobias Brodkorb
- Department of Obstetrics and Gynaecology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany
| | - Carsten Denkert
- Institute of Pathology, University Clinic Marburg, Marburg, Germany
| | - Angrit Stachs
- Department of Obstetrics and Gynecology, University Hospital of Rostock, Rostock, Germany
| | - David Krug
- Klinik für Strahlentherapie, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Jörg Heil
- Brustzentrum Heidelberg - Klinik St. Elisabeth, Heidelberg, Germany; Department of Obstetrics and Gynecology, Uniklinikum Heidelberg, Heidelberg, Germany
| | - Michael Golatta
- Brustzentrum Heidelberg - Klinik St. Elisabeth, Heidelberg, Germany; Department of Obstetrics and Gynecology, Uniklinikum Heidelberg, Heidelberg, Germany
| | - Thorsten Kühn
- Department of Gynaecology and Obstetrics, Klinikum Esslingen, Neckar, Germany
| | | | - Timo Gaiser
- Institute of Pathology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany
| | - Rebecca Schönmehl
- Institute of Pathology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany; Institute of Pathology, University Regensburg, Regensburg, Germany
| | - Sibylle Loibl
- German Breast Group, GBG Forschungs GmbH, Neu-Isenburg, Germany
| | - Toralf Reimer
- Department of Obstetrics and Gynecology, University Hospital of Rostock, Rostock, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
23
|
Li G, Li C, Liu J, Peng H, Lu S, Wei D, Guo J, Wang M, Yang N. Prediction of lymph node metastasis of lung squamous cell carcinoma by machine learning algorithm classifiers. J Cancer Res Ther 2023; 19:1533-1543. [PMID: 38156919 DOI: 10.4103/jcrt.jcrt_2352_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 07/31/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Lymph node metastasis (LNM) is an essential factor affecting the prognosis of patients with lung squamous cell carcinoma (LUSC), as well as a critical consideration for the choice of treatment strategy. Exploring effective methods for predicting LNM in LUSC may benefit clinical decision making. MATERIALS AND METHODS We used data collected from the Surveillance, Epidemiology, and End Results (SEER) database to develop machine learning algorithm classifiers, including boosted trees (BTs), based on the primary clinical parameters of patients to predict LNM in LUSC. Training on a large-sample training cohort (n = 8,063) allowed for the construction of several concise classifiers for LNM prediction in LUSC, which were then validated using test and in-house cohorts (n = 2,017 and 57, respectively). RESULTS The six classifiers established in this research enabled distinction between patients with and without LNM. Among these classifiers, the BT classifier was the top performer, with accuracy, F1 scores, precision, recall, sensitivity, and specificity values of 0.654, 0.621, 0.654, 0.592, 0.592, and 0.711, respectively. The precision recall (PR) and receiver operating characteristic (ROC) (with area under the curve = 0.714) curves also supported this result, which was validated by the in-house cohort. Notably, the tumor stage was a critical factor in determining LNM in patients with LUSC. CONCLUSIONS The use of classifiers, especially the BT classifier, may serve as a useful tool for improving clinical precision and individualized treatment of patients with LUSC.
Collapse
Affiliation(s)
- Guosheng Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Changqian Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huajian Peng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shuyu Lu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Donglin Wei
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jianji Guo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Meijing Wang
- Department of Cardiothoracic Surgery, Guilin People's Hospital, Guilin, China
| | - Nuo Yang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
Kurz A, Krahl D, Kutzner H, Barnhill R, Perasole A, Figueras MTF, Ferrara G, Braun SA, Starz H, Llamas-Velasco M, Utikal JS, Fröhling S, von Kalle C, Kather JN, Schneider L, Brinker TJ. A 3-dimensional histology computer model of malignant melanoma and its implications for digital pathology. Eur J Cancer 2023; 193:113294. [PMID: 37690178 DOI: 10.1016/j.ejca.2023.113294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Historically, cancer diagnoses have been made by pathologists using two-dimensional histological slides. However, with the advent of digital pathology and artificial intelligence, slides are being digitised, providing new opportunities to integrate their information. Since nature is 3-dimensional (3D), it seems intuitive to digitally reassemble the 3D structure for diagnosis. OBJECTIVE To develop the first human-3D-melanoma-histology-model with full data and code availability. Further, to evaluate the 3D-simulation together with experienced pathologists in the field and discuss the implications of digital 3D-models for the future of digital pathology. METHODS A malignant melanoma of the skin was digitised via 3 µm cuts by a slide scanner; an open-source software was then leveraged to construct the 3D model. A total of nine pathologists from four different countries with at least 10 years of experience in the histologic diagnosis of melanoma tested the model and discussed their experiences as well as implications for future pathology. RESULTS We successfully constructed and tested the first 3D-model of human melanoma. Based on testing, 88.9% of pathologists believe that the technology is likely to enter routine pathology within the next 10 years; advantages include a better reflectance of anatomy, 3D assessment of symmetry and the opportunity to simultaneously evaluate different tissue levels at the same time; limitations include the high consumption of tissue and a yet inferior resolution due to computational limitations. CONCLUSIONS 3D-histology-models are promising for digital pathology of cancer and melanoma specifically, however, there are yet limitations which need to be carefully addressed.
Collapse
Affiliation(s)
- Alexander Kurz
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Krahl
- Dres. Krahl Dermatopathology, Heidelberg, Germany
| | - Heinz Kutzner
- Dermatopathology Friedrichshafen, Friedrichshafen, Germany
| | - Raymond Barnhill
- Departments of Pathology and Translational Research, Institut Curie, Paris, France
| | - Antonio Perasole
- Division of Histopathology, Cerba Healthcare S.r.l. Rete Diagnostica Italiana, Limena, Italy
| | - Maria Teresa Fernandez Figueras
- University General Hospital of Catalonia, Grupo Quironsalud, International University of Catalonia, Sant Cugat del Vallés, Barcelona, Spain
| | - Gerardo Ferrara
- Anatomic Pathology and Cytopathology Unit Istituto Nazionale Tumori IRCCS Fondazione 'G. Pascale, Naples, Italy
| | - Stephan A Braun
- Department of Dermatology, University of Münster, Münster, Germany; Department of Dermatology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Mar Llamas-Velasco
- Department of Dermatology, University Hospital La Princesa, Madrid, Spain
| | - Jochen Sven Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Christof von Kalle
- Department of Clinical-Translational Sciences, Berlin Institute of Health (BIH), Charité University Medicine, Berlin, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Lucas Schneider
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
25
|
Muti HS, Röcken C, Behrens HM, Löffler CML, Reitsam NG, Grosser B, Märkl B, Stange DE, Jiang X, Velduizen GP, Truhn D, Ebert MP, Grabsch HI, Kather JN. Deep learning trained on lymph node status predicts outcome from gastric cancer histopathology: a retrospective multicentric study. Eur J Cancer 2023; 194:113335. [PMID: 37862795 DOI: 10.1016/j.ejca.2023.113335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/15/2023] [Accepted: 09/03/2023] [Indexed: 10/22/2023]
Abstract
AIM Gastric cancer (GC) is a tumour entity with highly variant outcomes. Lymph node metastasis is a prognostically adverse biomarker. We hypothesised that GC primary tissue contains information that is predictive of lymph node status and patient prognosis and that this information can be extracted using deep learning (DL). METHODS Using three patient cohorts comprising 1146 patients, we trained and validated a DL system to predict lymph node status directly from haematoxylin and eosin-stained GC tissue sections. We investigated the concordance between the DL-based prediction from the primary tumour slides (aiN score) and the histopathological lymph node status (pN). Furthermore, we assessed the prognostic value of the aiN score alone and when combined with the pN status. RESULTS The aiN score predicted the pN status reaching area under the receiver operating characteristic curves of 0.71 in the training cohort and 0.69 and 0.65 in the two test cohorts. In a multivariate Cox analysis, the aiN score was an independent predictor of patient survival with hazard ratios of 1.5 in the training cohort and of 1.3 and 2.2 in the two test cohorts. A combination of the aiN score and the pN status prognostically stratified patients by survival with p-values <0.05 in logrank tests. CONCLUSION GC primary tumour tissue contains additional prognostic information that is accessible using the aiN score. In combination with the pN status, this can be used for personalised management of GC patients after prospective validation.
Collapse
Affiliation(s)
- Hannah S Muti
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Chiara M L Löffler
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany
| | - Nic G Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Bianca Grosser
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Daniel E Stange
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Xiaofeng Jiang
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Gregory P Velduizen
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Germany
| | - Matthias P Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DKFZ-Hector Cancer Institute at the University Medical Center, Mannheim, Germany; Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
26
|
Wu L, Wu H, Li C, Zhang B, Li X, Zhen Y, Li H. Radiomics in colorectal cancer. IRADIOLOGY 2023; 1:236-244. [DOI: 10.1002/ird3.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2024]
Abstract
AbstractColorectal cancer (CRC) is a global health challenge with high morbidity and mortality. Radiomics, an emerging field, utilizes quantitative imaging features extracted from medical images for CRC diagnosis, staging, treatment response assessment, and prognostication. This review highlights the potential of radiomics for personalized CRC management. Radiomics enables noninvasive tumor characterization, aiding in early detection and accurate diagnosis, and it can be used to predict tumor stage, lymph node involvement, and prognosis. Furthermore, radiomics guides personalized therapies by assessing the treatment response and identifying patients who could benefit. Challenges include standardizing imaging protocols and analysis techniques. Robust validation frameworks and user‐friendly software are needed for the integration of radiomics into clinical practice. Despite challenges, radiomics offers valuable insights into tumor biology, treatment response, and prognosis in CRC. Overcoming technical and clinical hurdles will unlock its full potential in CRC management.
Collapse
Affiliation(s)
- Long Wu
- Department of Anus and Intestinal Surgery The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Huan Wu
- Department of Infectious Diseases The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy Free University of Berlin Berlin Germany
| | - Baofang Zhang
- Department of Infectious Diseases The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Xiaoyun Li
- Department of Anus and Intestinal Surgery The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Yunhuan Zhen
- Department of Anus and Intestinal Surgery The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| | - Haiyang Li
- Department of Hepatobiliary Surgery The Affiliated Hospital of Guizhou Medical University Guiyang Guizhou China
| |
Collapse
|
27
|
Wang X, Li H, Chang X, Tian Z. High serum mannose in colorectal cancer: a novel biomarker of lymph node metastasis and poor prognosis. Front Oncol 2023; 13:1213952. [PMID: 37675224 PMCID: PMC10479890 DOI: 10.3389/fonc.2023.1213952] [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: 05/22/2023] [Accepted: 07/27/2023] [Indexed: 09/08/2023] Open
Abstract
Background Lymph node status is an important prognostic indicator and it significantly influences treatment decisions for colorectal cancer (CRC). The objective of this study was to evaluate the ability of serum monosaccharides in predicting lymph node metastasis (LNM) and prognosis. Methods High performance anion exchange chromatography coupled with pulsed amperometric detector (HPAEC-PAD) was used to quantify serum monosaccharides from 252 CRC patients. Receiver operating characteristic (ROC) curves were used to evaluate predictive performance of parameters. Predictors of LNM were evaluated by univariate and multivariate analyses. The prognostic role of the factors was evaluated by survival analysis. Results The levels of serum mannose (Man) and galactose (Gal) were significantly increased in patients with LNM (p <0.0001, p =0.0017, respectively). The area under the curves (AUCs) of Man was 0.8140, which was higher than carcinoembryonic antigen (CEA) (AUC =0.6523). Univariate and multivariate analyses demonstrated histologic grade (G3) (odds ratio [OR] =2.60, p =0.043), histologic grade (mucin-producing subtype) (odds ratio [OR] =3.38, p =0.032), lymphovascular invasion (LVI) (OR =2.42, p <0.01), CEA (>5ng/ml) (OR =1.85, p =0.042) and high Man (OR =2.65, p =0.006) to be independent risk factors of LNM. The survival analysis showed that the high serum Man was independent risk factor for poor prognosis in CRC patients (HR=1.75, p =0.004). Conclusions The Man is superior to CEA in prediction of LNM for CRC patients. Man is expected to be a predictor for LNM in CRC. High serum Man is associated with poor prognosis of CRC patients.
Collapse
Affiliation(s)
- Xueling Wang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotian Chang
- Center for Clinical Research, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
28
|
Zheng Y, Carrillo-Perez F, Pizurica M, Heiland DH, Gevaert O. Spatial cellular architecture predicts prognosis in glioblastoma. Nat Commun 2023; 14:4122. [PMID: 37433817 DOI: 10.1038/s41467-023-39933-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Intra-tumoral heterogeneity and cell-state plasticity are key drivers for the therapeutic resistance of glioblastoma. Here, we investigate the association between spatial cellular organization and glioblastoma prognosis. Leveraging single-cell RNA-seq and spatial transcriptomics data, we develop a deep learning model to predict transcriptional subtypes of glioblastoma cells from histology images. Employing this model, we phenotypically analyze 40 million tissue spots from 410 patients and identify consistent associations between tumor architecture and prognosis across two independent cohorts. Patients with poor prognosis exhibit higher proportions of tumor cells expressing a hypoxia-induced transcriptional program. Furthermore, a clustering pattern of astrocyte-like tumor cells is associated with worse prognosis, while dispersion and connection of the astrocytes with other transcriptional subtypes correlate with decreased risk. To validate these results, we develop a separate deep learning model that utilizes histology images to predict prognosis. Applying this model to spatial transcriptomics data reveal survival-associated regional gene expression programs. Overall, our study presents a scalable approach to unravel the transcriptional heterogeneity of glioblastoma and establishes a critical connection between spatial cellular architecture and clinical outcomes.
Collapse
Affiliation(s)
- Yuanning Zheng
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
| | - Francisco Carrillo-Perez
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
- Department of Architecture and Computer Technology (ATC), University of Granada, Granada, 18014, Spain
| | - Marija Pizurica
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Technologiepark-Zwijnaarde 126, Ghent, 9052, Gent, Belgium
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, Freiburg, 79106, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, 79106, Germany
| | - Olivier Gevaert
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
29
|
Zheng Q, Jian J, Wang J, Wang K, Fan J, Xu H, Ni X, Yang S, Yuan J, Wu J, Jiao P, Yang R, Chen Z, Liu X, Wang L. Predicting Lymph Node Metastasis Status from Primary Muscle-Invasive Bladder Cancer Histology Slides Using Deep Learning: A Retrospective Multicenter Study. Cancers (Basel) 2023; 15:cancers15113000. [PMID: 37296961 DOI: 10.3390/cancers15113000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Accurate prediction of lymph node metastasis (LNM) status in patients with muscle-invasive bladder cancer (MIBC) before radical cystectomy can guide the use of neoadjuvant chemotherapy and the extent of pelvic lymph node dissection. We aimed to develop and validate a weakly-supervised deep learning model to predict LNM status from digitized histopathological slides in MIBC. METHODS We trained a multiple instance learning model with an attention mechanism (namely SBLNP) from a cohort of 323 patients in the TCGA cohort. In parallel, we collected corresponding clinical information to construct a logistic regression model. Subsequently, the score predicted by the SBLNP was incorporated into the logistic regression model. In total, 417 WSIs from 139 patients in the RHWU cohort and 230 WSIs from 78 patients in the PHHC cohort were used as independent external validation sets. RESULTS In the TCGA cohort, the SBLNP achieved an AUROC of 0.811 (95% confidence interval [CI], 0.771-0.855), the clinical classifier achieved an AUROC of 0.697 (95% CI, 0.661-0.728) and the combined classifier yielded an improvement to 0.864 (95% CI, 0.827-0.906). Encouragingly, the SBLNP still maintained high performance in the RHWU cohort and PHHC cohort, with an AUROC of 0.762 (95% CI, 0.725-0.801) and 0.746 (95% CI, 0.687-0.799), respectively. Moreover, the interpretability of SBLNP identified stroma with lymphocytic inflammation as a key feature of predicting LNM presence. CONCLUSIONS Our proposed weakly-supervised deep learning model can predict the LNM status of MIBC patients from routine WSIs, demonstrating decent generalization performance and holding promise for clinical implementation.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kai Wang
- Department of Urology, People's Hospital of Hanchuan City, Xiaogan 432300, China
| | - Junjie Fan
- University of Chinese Academy of Sciences, Beijing 100049, China
- Trusted Computing and Information Assurance Laboratory, Institute of Software, Chinese Academy of Sciences, Beijing 100190, China
| | - Huazhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Song Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiejun Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
30
|
Olatunji I, Cui F. Multimodal AI for prediction of distant metastasis in carcinoma patients. FRONTIERS IN BIOINFORMATICS 2023; 3:1131021. [PMID: 37228671 PMCID: PMC10203594 DOI: 10.3389/fbinf.2023.1131021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Metastasis of cancer is directly related to death in almost all cases, however a lot is yet to be understood about this process. Despite advancements in the available radiological investigation techniques, not all cases of Distant Metastasis (DM) are diagnosed at initial clinical presentation. Also, there are currently no standard biomarkers of metastasis. Early, accurate diagnosis of DM is however crucial for clinical decision making, and planning of appropriate management strategies. Previous works have achieved little success in attempts to predict DM from either clinical, genomic, radiology, or histopathology data. In this work we attempt a multimodal approach to predict the presence of DM in cancer patients by combining gene expression data, clinical data and histopathology images. We tested a novel combination of Random Forest (RF) algorithm with an optimization technique for gene selection, and investigated if gene expression pattern in the primary tissues of three cancer types (Bladder Carcinoma, Pancreatic Adenocarcinoma, and Head and Neck Squamous Carcinoma) with DM are similar or different. Gene expression biomarkers of DM identified by our proposed method outperformed Differentially Expressed Genes (DEGs) identified by the DESeq2 software package in the task of predicting presence or absence of DM. Genes involved in DM tend to be more cancer type specific rather than general across all cancers. Our results also indicate that multimodal data is more predictive of metastasis than either of the three unimodal data tested, and genomic data provides the highest contribution by a wide margin. The results re-emphasize the importance for availability of sufficient image data when a weakly supervised training technique is used. Code is made available at: https://github.com/rit-cui-lab/Multimodal-AI-for-Prediction-of-Distant-Metastasis-in-Carcinoma-Patients.
Collapse
Affiliation(s)
| | - Feng Cui
- Thomas H. Gosnell School of Life Science, Rochester Institute of Technology, Rochester, NY, United States
| |
Collapse
|
31
|
Xiao Y, Wang S, Ling R, Song Y. Application of artificial neural network algorithm in pathological diagnosis and prognosis prediction of digestive tract malignant tumors. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:243-248. [PMID: 37283110 DOI: 10.3724/zdxbyxb-2022-0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The application of artificial neural network algorithm in pathological diagnosis of gastrointestinal malignant tumors has become a research hotspot. In the previous studies, the algorithm research mainly focused on the model development based on convolutional neural networks, while only a few studies used the combination of convolutional neural networks and recurrent neural networks. The research contents included classical histopathological diagnosis and molecular typing of malignant tumors, and the prediction of patient prognosis by utilizing artificial neural networks. This article reviews the research progress on artificial neural network algorithm in the pathological diagnosis and prognosis prediction of digestive tract malignant tumors.
Collapse
Affiliation(s)
- Ya Xiao
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China.
| | - Shuyang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai 200032, China
| | - Ren Ling
- Shanghai Laizi Software Technology Co. Ltd., Shanghai 201499, China
| | - Yufei Song
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo 315046, Zhejiang Province, China.
| |
Collapse
|
32
|
Krogue JD, Azizi S, Tan F, Flament-Auvigne I, Brown T, Plass M, Reihs R, Müller H, Zatloukal K, Richeson P, Corrado GS, Peng LH, Mermel CH, Liu Y, Chen PHC, Gombar S, Montine T, Shen J, Steiner DF, Wulczyn E. Predicting lymph node metastasis from primary tumor histology and clinicopathologic factors in colorectal cancer using deep learning. COMMUNICATIONS MEDICINE 2023; 3:59. [PMID: 37095223 PMCID: PMC10125969 DOI: 10.1038/s43856-023-00282-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Presence of lymph node metastasis (LNM) influences prognosis and clinical decision-making in colorectal cancer. However, detection of LNM is variable and depends on a number of external factors. Deep learning has shown success in computational pathology, but has struggled to boost performance when combined with known predictors. METHODS Machine-learned features are created by clustering deep learning embeddings of small patches of tumor in colorectal cancer via k-means, and then selecting the top clusters that add predictive value to a logistic regression model when combined with known baseline clinicopathological variables. We then analyze performance of logistic regression models trained with and without these machine-learned features in combination with the baseline variables. RESULTS The machine-learned extracted features provide independent signal for the presence of LNM (AUROC: 0.638, 95% CI: [0.590, 0.683]). Furthermore, the machine-learned features add predictive value to the set of 6 clinicopathologic variables in an external validation set (likelihood ratio test, p < 0.00032; AUROC: 0.740, 95% CI: [0.701, 0.780]). A model incorporating these features can also further risk-stratify patients with and without identified metastasis (p < 0.001 for both stage II and stage III). CONCLUSION This work demonstrates an effective approach to combine deep learning with established clinicopathologic factors in order to identify independently informative features associated with LNM. Further work building on these specific results may have important impact in prognostication and therapeutic decision making for LNM. Additionally, this general computational approach may prove useful in other contexts.
Collapse
Affiliation(s)
| | | | - Fraser Tan
- Google Health, Palo Alto, California, USA
| | | | | | | | | | | | | | - Pema Richeson
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | - Yun Liu
- Google Health, Palo Alto, California, USA
| | | | - Saurabh Gombar
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Jeanne Shen
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | | | | |
Collapse
|
33
|
Tan L, Li H, Yu J, Zhou H, Wang Z, Niu Z, Li J, Li Z. Colorectal cancer lymph node metastasis prediction with weakly supervised transformer-based multi-instance learning. Med Biol Eng Comput 2023; 61:1565-1580. [PMID: 36809427 PMCID: PMC10182132 DOI: 10.1007/s11517-023-02799-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/31/2023] [Indexed: 02/23/2023]
Abstract
Lymph node metastasis examined by the resected lymph nodes is considered one of the most important prognostic factors for colorectal cancer (CRC). However, it requires careful and comprehensive inspection by expert pathologists. To relieve the pathologists' burden and speed up the diagnostic process, in this paper, we develop a deep learning system with the binary positive/negative labels of the lymph nodes to solve the CRC lymph node classification task. The multi-instance learning (MIL) framework is adopted in our method to handle the whole slide images (WSIs) of gigapixels in size at once and get rid of the labor-intensive and time-consuming detailed annotations. First, a transformer-based MIL model, DT-DSMIL, is proposed in this paper based on the deformable transformer backbone and the dual-stream MIL (DSMIL) framework. The local-level image features are extracted and aggregated with the deformable transformer, and the global-level image features are obtained with the DSMIL aggregator. The final classification decision is made based on both the local and the global-level features. After the effectiveness of our proposed DT-DSMIL model is demonstrated by comparing its performance with its predecessors, a diagnostic system is developed to detect, crop, and finally identify the single lymph nodes within the slides based on the DT-DSMIL and the Faster R-CNN model. The developed diagnostic model is trained and tested on a clinically collected CRC lymph node metastasis dataset composed of 843 slides (864 metastasis lymph nodes and 1415 non-metastatic lymph nodes), achieving the accuracy of 95.3% and the area under the receiver operating characteristic curve (AUC) of 0.9762 (95% confidence interval [CI]: 0.9607-0.9891) for the single lymph node classification. As for the lymph nodes with micro-metastasis and macro-metastasis, our diagnostic system achieves the AUC of 0.9816 (95% CI: 0.9659-0.9935) and 0.9902 (95% CI: 0.9787-0.9983), respectively. Moreover, the system shows reliable diagnostic region localizing performance: the model can always identify the most likely metastases, no matter the model's predictions or manual labels, showing great potential in avoiding false negatives and discovering incorrectly labeled slides in actual clinical use.
Collapse
Affiliation(s)
- Luxin Tan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jinze Yu
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing, 100191, China.,School of Computer Science and Engineering, Beihang University, Beijing, 100191, China.,Shenyuan Honors College, Beihang University, Beijing, 100191, China
| | - Haoyi Zhou
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing, 100191, China.,College of Software, Beihang University, Beijing, 100191, China
| | - Zhi Wang
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Zhiyong Niu
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China.
| | - Jianxin Li
- Beijing Advanced Innovation Center for Big Data and Brain Computing, Beihang University, Beijing, 100191, China. .,School of Computer Science and Engineering, Beihang University, Beijing, 100191, China.
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
34
|
Kindler C, Elfwing S, Öhrvik J, Nikberg M. A Deep Neural Network-Based Decision Support Tool for the Detection of Lymph Node Metastases in Colorectal Cancer Specimens. Mod Pathol 2023; 36:100015. [PMID: 36853787 DOI: 10.1016/j.modpat.2022.100015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 01/11/2023]
Abstract
The identification of lymph node metastases in colorectal cancer (CRC) specimens is crucial for the planning of postoperative treatment and can be a time-consuming task for pathologists. In this study, we developed a deep neural network (DNN) algorithm for the detection of metastatic CRC in digitized histologic sections of lymph nodes and evaluated its performance as a diagnostic support tool. First, the DNN algorithm was trained using pixel-level annotations of cancerous areas on 758 whole slide images (360 with cancerous areas). The algorithm's performance was evaluated on 74 whole slide images (43 with cancerous areas). Second, the algorithm was evaluated as a decision support tool on 288 whole slide images covering 1517 lymph node sections, randomized in 16 batches. Two senior pathologists (C.K. and C.O.) assessed each batch with and without the help of the algorithm in a 2 × 2 crossover design, with a washout period of 1 month in between. The time needed for the evaluation of each node section was recorded. The DNN algorithm achieved a median pixel-level accuracy of 0.952 on slides with cancerous areas and 0.996 on slides with benign samples. N+ disease (metastases, micrometastases, or tumor deposits) was present in 103 of the 1517 sections. The algorithm highlighted cancerous areas in 102 of these sections, with a sensitivity of 0.990. Assisted by the algorithm, the median time needed for evaluation was significantly shortened for both pathologists (median time for pathologist 1, 26 vs 14 seconds; P < .001; 95% CI, 11.0-12.0; median time for pathologist 2, 25 vs 23 seconds; P < .001; 95% CI, 2.0-4.0). Our DNN showed high accuracy for detecting metastatic CRC in digitized histologic sections of lymph nodes. This decision support tool has the potential to improve the diagnostic workflow by shortening the time needed for the evaluation of lymph nodes in CRC specimens without impairing diagnostic accuracy.
Collapse
Affiliation(s)
- Csaba Kindler
- Department of Pathology, Laboratory Medicine, Västmanlands Hospital, Västerås, Sweden; Centre for Clinical Research, Uppsala University, Västerås, Sweden.
| | | | - John Öhrvik
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Maziar Nikberg
- Centre for Clinical Research, Uppsala University, Västerås, Sweden; Department of Surgery, Västmanlands Hospital, Västerås, Sweden
| |
Collapse
|
35
|
Hu X, Jiang L, Wu J, Mao W. Prognostic value of log odds of positive lymph nodes, lymph node ratio, and N stage in patients with colorectal signet ring cell carcinoma: A retrospective cohort study. Front Surg 2023; 9:1019454. [PMID: 36684239 PMCID: PMC9849566 DOI: 10.3389/fsurg.2022.1019454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/28/2022] [Indexed: 01/07/2023] Open
Abstract
Aim Little attention has been paid in the prognosis of colorectal signet ring cell carcinoma (SRCC). This study aims to explore the predictive capacity of log odds of positive lymph nodes (LODDS), lymph node ratio (LNR), and pN stage in the prognosis of patients with colorectal SRCC. Methods A retrospective cohort study was designed, and data were extracted from the Surveillance, Epidemiology and End Results (SEER) database. Data on demographic characteristics, clinicopathological features, and treatment were extracted. Outcomes were overall survival (OS) and cancer-specific survival (CSS). Association of LODDS, LNR, and pN stage with OS and CSS were explored using Cox proportional hazard model and Cox competing risk model, respectively, with results showing as hazard ratio and 95% confidence interval (CI). Predictive performance of LODDS, LNR, and pN stage in OS and CSS was assessed by calculating C-index. Results A total of 2,198 patients were included in this study. LODDS, LNR, and pN stage were associated with the OS and CSS of colorectal SRCC patients (all P < 0.05). LODDS showed a good performance in the OS (C-index: 0.704, 95% CI: 0.690-0.718), which was superior to LNR (C-index: 0.657, 95% CI: 0.643-0.671) and pN stage (C-index: 0.643, 95% CI: 0.629-0.657). The C-index of LODDS, LNR, and pN stage for CSS was 0.733 (95% CI: 0.719-0.747), 0.713 (95% CI: 0.697-0.729), and 0.667 (95% CI: 0.651-0.683), respectively. Conclusions LODDS displayed a better predictive capacity in the OS and CSS than LNR and pN stage, indicating that LODDS may be effective to predict the prognosis of colorectal SRCC in the clinic.
Collapse
|
36
|
Yavuz A, Alpsoy A, Gedik EO, Celik MY, Bassorgun CI, Unal B, Elpek GO. Artificial intelligence applications in predicting the behavior of gastrointestinal cancers in pathology. Artif Intell Gastroenterol 2022; 3:142-162. [DOI: 10.35712/aig.v3.i5.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Recent research has provided a wealth of data supporting the application of artificial intelligence (AI)-based applications in routine pathology practice. Indeed, it is clear that these methods can significantly support an accurate and rapid diagnosis by eliminating errors, increasing reliability, and improving workflow. In addition, the effectiveness of AI in the pathological evaluation of prognostic parameters associated with behavior, course, and treatment in many types of tumors has also been noted. Regarding gastrointestinal system (GIS) cancers, the contribution of AI methods to pathological diagnosis has been investigated in many studies. On the other hand, studies focusing on AI applications in evaluating parameters to determine tumor behavior are relatively few. For this purpose, the potential of AI models has been studied over a broad spectrum, from tumor subtyping to the identification of new digital biomarkers. The capacity of AI to infer genetic alterations of cancer tissues from digital slides has been demonstrated. Although current data suggest the merit of AI-based approaches in assessing tumor behavior in GIS cancers, a wide range of challenges still need to be solved, from laboratory infrastructure to improving the robustness of algorithms, before incorporating AI applications into real-life GIS pathology practice. This review aims to present data from AI applications in evaluating pathological parameters related to the behavior of GIS cancer with an overview of the opportunities and challenges encountered in implementing AI in pathology.
Collapse
Affiliation(s)
- Aysen Yavuz
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Anil Alpsoy
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Elif Ocak Gedik
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | | | | | - Betul Unal
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Gulsum Ozlem Elpek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| |
Collapse
|
37
|
Nero C, Boldrini L, Lenkowicz J, Giudice MT, Piermattei A, Inzani F, Pasciuto T, Minucci A, Fagotti A, Zannoni G, Valentini V, Scambia G. Deep-Learning to Predict BRCA Mutation and Survival from Digital H&E Slides of Epithelial Ovarian Cancer. Int J Mol Sci 2022; 23:ijms231911326. [PMID: 36232628 PMCID: PMC9570450 DOI: 10.3390/ijms231911326] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
BRCA 1/2 genes mutation status can already determine the therapeutic algorithm of high grade serous ovarian cancer patients. Nevertheless, its assessment is not sufficient to identify all patients with genomic instability, since BRCA 1/2 mutations are only the most well-known mechanisms of homologous recombination deficiency (HR-d) pathway, and patients displaying HR-d behave similarly to BRCA mutated patients. HRd assessment can be challenging and is progressively overcoming BRCA testing not only for prognostic information but more importantly for drugs prescriptions. However, HR testing is not already integrated in clinical practice, it is quite expensive and it is not refundable in many countries. Selecting patients who are more likely to benefit from this assessment (BRCA 1/2 WT patients) at an early stage of the diagnostic process, would allow an optimization of genomic profiling resources. In this study, we sought to explore whether somatic BRCA1/2 genes status can be predicted using computational pathology from standard hematoxylin and eosin histology. In detail, we adopted a publicly available, deep-learning-based weakly supervised method that uses attention-based learning to automatically identify sub regions of high diagnostic value to accurately classify the whole slide (CLAM). The same model was also tested for progression free survival (PFS) prediction. The model was tested on a cohort of 664 (training set: n = 464, testing set: n = 132) ovarian cancer patients, of whom 233 (35.1%) had a somatic BRCA 1/2 mutation. An area under the curve of 0.7 and 0.55 was achieved in the training and testing set respectively. The model was then further refined by manually identifying areas of interest in half of the cases. 198 images were used for training (126/72) and 87 images for validation (55/32). The model reached a zero classification error on the training set, but the performance was 0.59 in terms of validation ROC AUC, with a 0.57 validation accuracy. Finally, when applied to predict PFS, the model achieved an AUC of 0.71, with a negative predictive value of 0.69, and a positive predictive value of 0.75. Based on these analyses, we have planned further steps of development such as proving a reference classification performance, exploring the hyperparameters space for training optimization, eventually tweaking the learning algorithms and the neural networks architecture for better suiting this specific task. These actions may allow the model to improve performances for all the considered outcomes.
Collapse
Affiliation(s)
- Camilla Nero
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-30154979
| | - Luca Boldrini
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiomics Core Facility, 00168 Rome, Italy
| | - Jacopo Lenkowicz
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiomics Core Facility, 00168 Rome, Italy
| | - Maria Teresa Giudice
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| | - Alessia Piermattei
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Frediano Inzani
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Tina Pasciuto
- Fondazione Policlinico Agostino Gemelli, IRCCS, Data Collection Core Facility, 00168 Rome, Italy
| | - Angelo Minucci
- Fondazione Policlinico Agostino Gemelli, IRCCS, Genomics Core Facility, 00168 Rome, Italy
| | - Anna Fagotti
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| | - Gianfranco Zannoni
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Vincenzo Valentini
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiation Oncology, 00168 Rome, Italy
| | - Giovanni Scambia
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| |
Collapse
|
38
|
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: 35] [Impact Index Per Article: 11.7] [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
|
39
|
Nam D, Chapiro J, Paradis V, Seraphin TP, Kather JN. Artificial intelligence in liver diseases: improving diagnostics, prognostics and response prediction. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100443. [PMID: 35243281 PMCID: PMC8867112 DOI: 10.1016/j.jhepr.2022.100443] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022]
Abstract
Clinical routine in hepatology involves the diagnosis and treatment of a wide spectrum of metabolic, infectious, autoimmune and neoplastic diseases. Clinicians integrate qualitative and quantitative information from multiple data sources to make a diagnosis, prognosticate the disease course, and recommend a treatment. In the last 5 years, advances in artificial intelligence (AI), particularly in deep learning, have made it possible to extract clinically relevant information from complex and diverse clinical datasets. In particular, histopathology and radiology image data contain diagnostic, prognostic and predictive information which AI can extract. Ultimately, such AI systems could be implemented in clinical routine as decision support tools. However, in the context of hepatology, this requires further large-scale clinical validation and regulatory approval. Herein, we summarise the state of the art in AI in hepatology with a particular focus on histopathology and radiology data. We present a roadmap for the further development of novel biomarkers in hepatology and outline critical obstacles which need to be overcome.
Collapse
|
40
|
Schneider L, Laiouar-Pedari S, Kuntz S, Krieghoff-Henning E, Hekler A, Kather JN, Gaiser T, Fröhling S, Brinker TJ. Integration of deep learning-based image analysis and genomic data in cancer pathology: A systematic review. Eur J Cancer 2021; 160:80-91. [PMID: 34810047 DOI: 10.1016/j.ejca.2021.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Over the past decade, the development of molecular high-throughput methods (omics) increased rapidly and provided new insights for cancer research. In parallel, deep learning approaches revealed the enormous potential for medical image analysis, especially in digital pathology. Combining image and omics data with deep learning tools may enable the discovery of new cancer biomarkers and a more precise prediction of patient prognosis. This systematic review addresses different multimodal fusion methods of convolutional neural network-based image analyses with omics data, focussing on the impact of data combination on the classification performance. METHODS PubMed was screened for peer-reviewed articles published in English between January 2015 and June 2021 by two independent researchers. Search terms related to deep learning, digital pathology, omics, and multimodal fusion were combined. RESULTS We identified a total of 11 studies meeting the inclusion criteria, namely studies that used convolutional neural networks for haematoxylin and eosin image analysis of patients with cancer in combination with integrated omics data. Publications were categorised according to their endpoints: 7 studies focused on survival analysis and 4 studies on prediction of cancer subtypes, malignancy or microsatellite instability with spatial analysis. CONCLUSIONS Image-based classifiers already show high performances in prognostic and predictive cancer diagnostics. The integration of omics data led to improved performance in all studies described here. However, these are very early studies that still require external validation to demonstrate their generalisability and robustness. Further and more comprehensive studies with larger sample sizes are needed to evaluate performance and determine clinical benefits.
Collapse
Affiliation(s)
- Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Laiouar-Pedari
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Kuntz
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Krieghoff-Henning
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Achim Hekler
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob N Kather
- Department of Medicine III, RWTH Aachen University Hospital, Aachen, Germany; Medical Oncology, National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefan Fröhling
- Translational Medical Oncology, National Center for Tumour Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|