1
|
Ai Y, Liu J, Li Y, Wang F, Du X, Jain RK, Lin L, Chen YW. SAMA: A Self-and-Mutual Attention Network for Accurate Recurrence Prediction of Non-Small Cell Lung Cancer Using Genetic and CT Data. IEEE J Biomed Health Inform 2025; 29:3220-3233. [PMID: 39348246 DOI: 10.1109/jbhi.2024.3471194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Accurate preoperative recurrence prediction for non-small cell lung cancer (NSCLC) is a challenging issue in the medical field. Existing studies primarily conduct image and molecular analyses independently or directly fuse multimodal information through radiomics and genomics, which fail to fully exploit and effectively utilize the highly heterogeneous cross-modal information at different levels and model the complex relationships between modalities, resulting in poor fusion performance and becoming the bottleneck of precise recurrence prediction. To address these limitations, we propose a novel unified framework, the Self-and-Mutual Attention (SAMA) Network, designed to efficiently fuse and utilize macroscopic CT images and microscopic gene data for precise NSCLC recurrence prediction, integrating handcrafted features, deep features, and gene features. Specifically, we design a Self-and-Mutual Attention Module that performs three-stage fusion: the self-enhancement stage enhances modality-specific features; the gene-guided and CT-guided cross-modality fusion stages perform bidirectional cross-guidance on the self-enhanced features, complementing and refining each modality, enhancing heterogeneous feature expression; and the optimized feature aggregation stage ensures the refined interactive features for precise prediction. Extensive experiments on both publicly available datasets from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA) demonstrate that our method achieves state-of-the-art performance and exhibits broad applicability to various cancers.
Collapse
|
2
|
Wang X, Yue X, Hang T, Liu S. Analysis of Hyperspectral Imaging Using CNN-GRU for Gastric Adenomatous Polyp and Adenocarcinoma Classification. JOURNAL OF BIOPHOTONICS 2025:e70047. [PMID: 40288998 DOI: 10.1002/jbio.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Early identification of gastric adenomatous polyps and adenocarcinoma is vital for improving patient outcomes. This study proposes a hybrid CNN-GRU model to classify one-dimensional hyperspectral data from ex vivo gastric tissues, addressing limitations of traditional diagnostics. Our model innovatively combines convolutional neural networks (CNNs) and gated recurrent units (GRUs) to capture both spatial and sequential dependencies in spectral data. Experimental results demonstrate that our model achieves an accuracy of 86%, sensitivity of 88%, and specificity of 85%. Additionally, receiver operating characteristic analysis further underscores its robust performance with an area under the curve of 0.86, surpassing traditional methods and other baseline models. These findings highlight the potential of leveraging advanced machine learning techniques to enhance early diagnostic accuracy and treatment strategies. The proposed approach offers a promising tool for rapid, accurate differentiation of gastric lesions, underscoring the importance of integrating innovative technologies in clinical diagnostics.
Collapse
Affiliation(s)
- Xuzhe Wang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoqing Yue
- Nanjing University of Chinese Medicine, Nanjing, China
- Yucheng People's Hospital, Dezhou, China
| | - Tianyi Hang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuai Liu
- Department of Pathology, The Fourth People's Hospital of Jinan, Jinan, China
| |
Collapse
|
3
|
Chen J, Liu P, Chen C, Su Y, Zuo E, Li M, Wang J, Yan Z, Chen X, Chen C, Lv X. TDMFS: Tucker decomposition multimodal fusion model for pan-cancer survival prediction. Artif Intell Med 2025; 162:103099. [PMID: 40037056 DOI: 10.1016/j.artmed.2025.103099] [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: 01/22/2024] [Revised: 11/05/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Integrated analysis of multimodal data offers a more comprehensive view for cancer survival prediction, yet it faces challenges like computational intensity, overfitting, and challenges in achieving a unified representation due to data heterogeneity. To address the above issues, the first Tucker decomposition multimodal fusion model was hereby proposed for pan-cancer survival prediction (TDMFS). The model employed Tucker decomposition to limit complex tensor parameters during fusion, achieving deep modality integration with reduced computational cost and lower overfitting risk. The individual modality-specific representations were then fully exploited by signal modulation mechanisms in a bilinear pooling decomposition to serve as complementary information for the deep fusion representation. Furthermore, the performance of TDMFS was evaluated using a 5-fold cross-validation method with two modal data, gene expression (GeneExpr), and copy number variation (CNV), for 33 cancers from The Cancer Genome Atlas (TCGA) database. The experiments demonstrated that the proposed TDMFS model achieved an average C-index of 0.757 across 33 cancer datasets, with a C-index exceeding 0.80 on 10 of these datasets. Survival curves for both high and low risk patients plotted on 27 cancer datasets were statistically significant. The TDMFS model demonstrated superior performance in survival prediction, outperforming models like LinearSum and Multimodal Factorisation Higher Order Pooling, making it a valuable asset for advancing clinical cancer research.
Collapse
Affiliation(s)
- Jinchao Chen
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Pei Liu
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Chen Chen
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Ying Su
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Enguang Zuo
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Min Li
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jiajia Wang
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Ziwei Yan
- College of Software, Xinjiang University, Urumqi 830046, China
| | - Xinya Chen
- College of Computer Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Cheng Chen
- College of Software, Xinjiang University, Urumqi 830046, China.
| | - Xiaoyi Lv
- College of Software, Xinjiang University, Urumqi 830046, China; The Key Laboratory of Signal Detection and Processing, Xinjiang Uygur Autonomous Region, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
4
|
Miao L, Li Z, Gao J. A multi-model machine learning framework for breast cancer risk stratification using clinical and imaging data. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2025; 33:360-375. [PMID: 39973793 DOI: 10.1177/08953996241308175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
PurposeThis study presents a comprehensive machine learning framework for assessing breast cancer malignancy by integrating clinical features with imaging features derived from deep learning.MethodsThe dataset included 1668 patients with documented breast lesions, incorporating clinical data (e.g., age, BI-RADS category, lesion size, margins, and calcifications) alongside mammographic images processed using four CNN architectures: EfficientNet, ResNet, DenseNet, and InceptionNet. Three predictive configurations were developed: an imaging-only model, a hybrid model combining imaging and clinical data, and a stacking-based ensemble model that aggregates both data types to enhance predictive accuracy. Twelve feature selection techniques, including ReliefF and Fisher Score, were applied to identify key predictive features. Model performance was evaluated using accuracy and AUC, with 5-fold cross-valida tion and hyperparameter tuning to ensure robustness.ResultsThe imaging-only models demonstrated strong predictive performance, with EfficientNet achieving an AUC of 0.76. The hybrid model combining imaging and clinical data reached the highest accuracy of 83% and an AUC of 0.87, underscoring the benefits of data integration. The stacking-based ensemble model further optimized accuracy, reaching a peak AUC of 0.94, demonstrating its potential as a reliable tool for malignancy risk assessment.ConclusionThis study highlights the importance of integrating clinical and deep imaging features for breast cancer risk stratification, with the stacking-based model.
Collapse
Affiliation(s)
- Lu Miao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Zidong Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
5
|
Zhou C, Mooyaart AL, Kerkour T, Louwman MWJ, Wakkee M, Li Y, Voorham QJM, Bruggink A, Nijsten TEC, Hollestein LM. The Dutch Early-Stage Melanoma (D-ESMEL) study: a discovery set and validation cohort to predict the absolute risk of distant metastases in stage I/II cutaneous melanoma. Eur J Epidemiol 2025; 40:27-42. [PMID: 39786688 PMCID: PMC11799080 DOI: 10.1007/s10654-024-01188-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025]
Abstract
Early-stage cutaneous melanoma patients generally have a favorable prognosis, yet a significant proportion of metastatic melanoma cases arise from this group, highlighting the need for improved risk stratification using novel prognostic biomarkers. The Dutch Early-Stage Melanoma (D-ESMEL) study introduces a robust, population-based methodology to develop an absolute risk prediction model for stage I/II melanoma, incorporating clinical, imaging, and multi-omics data to identify patients at increased risk for distant metastases. Utilizing the Netherlands Cancer Registry and Dutch Nationwide Pathology Databank, we collected primary tumor samples from early-stage melanoma patients, with and without distant metastases during follow-up. Our study design includes a discovery set of metastatic cases and matched controls to identify novel prognostic factors, followed by a validation cohort using a nested case-control design to validate these factors and to build a risk prediction model. Tissue sections underwent Hematoxylin & Eosin (H&E) staining, RNA sequencing (RNAseq), DNA sequencing (DNAseq), immunohistochemistry (IHC), and multiplex immunofluorescence (MxIF).The discovery set included 442 primary melanoma samples (221 case-control sets), with 46% stage I and 54% stage II melanomas. The median time to distant metastasis was 3.4 years, while controls had a median follow-up time of 9.8 years. The validation cohort included 154 cases and 154 controls from a random population-based selection of 5,815 patients. Our approach enabled the collection of a large number of early-stage melanoma samples from population-based databases with extensive follow-up and a sufficient number of metastatic events. This methodology in prognostic cancer research holds the potential to impact clinical decision-making through absolute risk prediction.
Collapse
Affiliation(s)
- Catherine Zhou
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Antien L Mooyaart
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Thamila Kerkour
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marieke W J Louwman
- Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
| | - Marlies Wakkee
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Annette Bruggink
- Dutch Nationwide Pathology Databank (Palga), Houten, The Netherlands
| | - Tamar E C Nijsten
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Loes M Hollestein
- Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands.
| |
Collapse
|
6
|
Vázquez-Lema D, Mosqueira-Rey E, Hernández-Pereira E, Fernandez-Lozano C, Seara-Romera F, Pombo-Otero J. Segmentation, classification and interpretation of breast cancer medical images using human-in-the-loop machine learning. Neural Comput Appl 2024. [DOI: 10.1007/s00521-024-10799-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
|
7
|
Ghanegolmohammadi F, Eslami M, Ohya Y. Systematic data analysis pipeline for quantitative morphological cell phenotyping. Comput Struct Biotechnol J 2024; 23:2949-2962. [PMID: 39104709 PMCID: PMC11298594 DOI: 10.1016/j.csbj.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Quantitative morphological phenotyping (QMP) is an image-based method used to capture morphological features at both the cellular and population level. Its interdisciplinary nature, spanning from data collection to result analysis and interpretation, can lead to uncertainties, particularly among those new to this actively growing field. High analytical specificity for a typical QMP is achieved through sophisticated approaches that can leverage subtle cellular morphological changes. Here, we outline a systematic workflow to refine the QMP methodology. For a practical review, we describe the main steps of a typical QMP; in each step, we discuss the available methods, their applications, advantages, and disadvantages, along with the R functions and packages for easy implementation. This review does not cover theoretical backgrounds, but provides several references for interested researchers. It aims to broaden the horizons for future phenome studies and demonstrate how to exploit years of endeavors to achieve more with less.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mohammad Eslami
- Harvard Ophthalmology AI Lab, Schepen’s Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
8
|
Tafavvoghi M, Bongo LA, Shvetsov N, Busund LTR, Møllersen K. Publicly available datasets of breast histopathology H&E whole-slide images: A scoping review. J Pathol Inform 2024; 15:100363. [PMID: 38405160 PMCID: PMC10884505 DOI: 10.1016/j.jpi.2024.100363] [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: 09/14/2023] [Revised: 11/24/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Advancements in digital pathology and computing resources have made a significant impact in the field of computational pathology for breast cancer diagnosis and treatment. However, access to high-quality labeled histopathological images of breast cancer is a big challenge that limits the development of accurate and robust deep learning models. In this scoping review, we identified the publicly available datasets of breast H&E-stained whole-slide images (WSIs) that can be used to develop deep learning algorithms. We systematically searched 9 scientific literature databases and 9 research data repositories and found 17 publicly available datasets containing 10 385 H&E WSIs of breast cancer. Moreover, we reported image metadata and characteristics for each dataset to assist researchers in selecting proper datasets for specific tasks in breast cancer computational pathology. In addition, we compiled 2 lists of breast H&E patches and private datasets as supplementary resources for researchers. Notably, only 28% of the included articles utilized multiple datasets, and only 14% used an external validation set, suggesting that the performance of other developed models may be susceptible to overestimation. The TCGA-BRCA was used in 52% of the selected studies. This dataset has a considerable selection bias that can impact the robustness and generalizability of the trained algorithms. There is also a lack of consistent metadata reporting of breast WSI datasets that can be an issue in developing accurate deep learning models, indicating the necessity of establishing explicit guidelines for documenting breast WSI dataset characteristics and metadata.
Collapse
Affiliation(s)
- Masoud Tafavvoghi
- Department of Community Medicine, Uit The Arctic University of Norway, Tromsø, Norway
| | - Lars Ailo Bongo
- Department of Computer Science, Uit The Arctic University of Norway, Tromsø, Norway
| | - Nikita Shvetsov
- Department of Computer Science, Uit The Arctic University of Norway, Tromsø, Norway
| | | | - Kajsa Møllersen
- Department of Community Medicine, Uit The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
9
|
Wang FA, Li Y, Zeng T. Deep Learning of radiology-genomics integration for computational oncology: A mini review. Comput Struct Biotechnol J 2024; 23:2708-2716. [PMID: 39035833 PMCID: PMC11260400 DOI: 10.1016/j.csbj.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
In the field of computational oncology, patient status is often assessed using radiology-genomics, which includes two key technologies and data, such as radiology and genomics. Recent advances in deep learning have facilitated the integration of radiology-genomics data, and even new omics data, significantly improving the robustness and accuracy of clinical predictions. These factors are driving artificial intelligence (AI) closer to practical clinical applications. In particular, deep learning models are crucial in identifying new radiology-genomics biomarkers and therapeutic targets, supported by explainable AI (xAI) methods. This review focuses on recent developments in deep learning for radiology-genomics integration, highlights current challenges, and outlines some research directions for multimodal integration and biomarker discovery of radiology-genomics or radiology-omics that are urgently needed in computational oncology.
Collapse
Affiliation(s)
- Feng-ao Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yixue Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Tao Zeng
- Guangzhou National Laboratory, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Hosseini MS, Bejnordi BE, Trinh VQH, Chan L, Hasan D, Li X, Yang S, Kim T, Zhang H, Wu T, Chinniah K, Maghsoudlou S, Zhang R, Zhu J, Khaki S, Buin A, Chaji F, Salehi A, Nguyen BN, Samaras D, Plataniotis KN. Computational pathology: A survey review and the way forward. J Pathol Inform 2024; 15:100357. [PMID: 38420608 PMCID: PMC10900832 DOI: 10.1016/j.jpi.2023.100357] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/02/2024] Open
Abstract
Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.
Collapse
Affiliation(s)
- Mahdi S. Hosseini
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | | | - Vincent Quoc-Huy Trinh
- Institute for Research in Immunology and Cancer of the University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Lyndon Chan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Danial Hasan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Xingwen Li
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Stephen Yang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Taehyo Kim
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Haochen Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Theodore Wu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Kajanan Chinniah
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Sina Maghsoudlou
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ryan Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jiadai Zhu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Samir Khaki
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Andrei Buin
- Huron Digitial Pathology, St. Jacobs, ON N0B 2N0, Canada
| | - Fatemeh Chaji
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ala Salehi
- Department of Electrical and Computer Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Bich Ngoc Nguyen
- University of Montreal Hospital Center, Montreal, QC H2X 0C2, Canada
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11794, United States
| | - Konstantinos N. Plataniotis
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| |
Collapse
|
11
|
Wagner VM. Is Histopathology Deep Learning Artificial Intelligence the Future of Precision Oncology? J Clin Oncol 2024; 42:3513-3516. [PMID: 39259925 PMCID: PMC11556860 DOI: 10.1200/jco-24-01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/18/2024] [Indexed: 09/13/2024] Open
Abstract
In the article that accompanies this editorial, Bergstrom et al. present DeepHRD, a deep learning algorithm that predicts homologous recombination deficiency (HRD) and clinical outcomes directly from digital histopathology slides, demonstrating its accuracy and generalizability across multiple independent cohorts of breast and ovarian cancer patients. This deep learning approach has the potential to be the next wave of precision medicine in oncology care but there are many challenges to overcome before wide clinical adoption including robust validation, prospective evaluation and regulatory approval.
Collapse
Affiliation(s)
- Vincent M Wagner
- The University of Iowa, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Iowa City, IA
| |
Collapse
|
12
|
Calderaro J, Žigutytė L, Truhn D, Jaffe A, Kather JN. Artificial intelligence in liver cancer - new tools for research and patient management. Nat Rev Gastroenterol Hepatol 2024; 21:585-599. [PMID: 38627537 DOI: 10.1038/s41575-024-00919-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 07/31/2024]
Abstract
Liver cancer has high incidence and mortality globally. Artificial intelligence (AI) has advanced rapidly, influencing cancer care. AI systems are already approved for clinical use in some tumour types (for example, colorectal cancer screening). Crucially, research demonstrates that AI can analyse histopathology, radiology and natural language in liver cancer, and can replace manual tasks and access hidden information in routinely available clinical data. However, for liver cancer, few of these applications have translated into large-scale clinical trials or clinically approved products. Here, we advocate for the incorporation of AI in all stages of liver cancer management. We present a taxonomy of AI approaches in liver cancer, highlighting areas with academic and commercial potential, and outline a policy for AI-based liver cancer management, including interdisciplinary training of researchers, clinicians and patients. The potential of AI in liver cancer is immense, but effort is required to ensure that AI can fulfil expectations.
Collapse
Affiliation(s)
- Julien Calderaro
- Département de Pathologie, Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Henri Mondor, Créteil, France
- Institut Mondor de Recherche Biomédicale, MINT-HEP Mondor Integrative Hepatology, Université Paris Est Créteil, Créteil, France
| | - Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Ariel Jaffe
- Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
13
|
Zhao Y, Li X, Zhou C, Peng H, Zheng Z, Chen J, Ding W. A review of cancer data fusion methods based on deep learning. INFORMATION FUSION 2024; 108:102361. [DOI: 10.1016/j.inffus.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
14
|
Cui C, Tang Y, Zhang W. Deep Survival Analysis With Latent Clustering and Contrastive Learning. IEEE J Biomed Health Inform 2024; 28:3090-3101. [PMID: 38319782 DOI: 10.1109/jbhi.2024.3362850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Survival analysis is employed to analyze the time before the event of interest occurs, which is broadly applied in many fields. The existence of censored data with incomplete supervision information about survival outcomes is one key challenge in survival analysis tasks. Although some progress has been made on this issue recently, the present methods generally treat the instances as separate ones while ignoring their potential correlations, thus rendering unsatisfactory performance. In this study, we propose a novel Deep Survival Analysis model with latent Clustering and Contrastive learning (DSACC). Specifically, we jointly optimize representation learning, latent clustering and survival prediction in a unified framework. In this way, the clusters distribution structure in latent representation space is revealed, and meanwhile the structure of the clusters is well incorporated to improve the ability of survival prediction. Besides, by virtue of the learned clusters, we further propose a contrastive loss function, where the uncensored data in each cluster are set as anchors, and the censored data are treated as positive/negative sample pairs according to whether they belong to the same cluster or not. This design enables the censored data to make full use of the supervision information of the uncensored samples. Through extensive experiments on four popular clinical datasets, we demonstrate that our proposed DSACC achieves advanced performance in terms of both C-index (0.6722, 0.6793, 0.6350, and 0.7943) and Integrated Brier Score (IBS) (0.1616, 0.1826, 0.2028, and 0.1120).
Collapse
|
15
|
Sun K, Zheng Y, Yang X, Jia W. A novel transformer-based aggregation model for predicting gene mutations in lung adenocarcinoma. Med Biol Eng Comput 2024; 62:1427-1440. [PMID: 38233683 DOI: 10.1007/s11517-023-03004-9] [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: 07/02/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
In recent years, predicting gene mutations on whole slide imaging (WSI) has gained prominence. The primary challenge is extracting global information and achieving unbiased semantic aggregation. To address this challenge, we propose a novel Transformer-based aggregation model, employing a self-learning weight aggregation mechanism to mitigate semantic bias caused by the abundance of features in WSI. Additionally, we adopt a random patch training method, which enhances model learning richness by randomly extracting feature vectors from WSI, thus addressing the issue of limited data. To demonstrate the model's effectiveness in predicting gene mutations, we leverage the lung adenocarcinoma dataset from Shandong Provincial Hospital for prior knowledge learning. Subsequently, we assess TP53, CSMD3, LRP1B, and TTN gene mutations using lung adenocarcinoma tissue pathology images and clinical data from The Cancer Genome Atlas (TCGA). The results indicate a notable increase in the AUC (Area Under the ROC Curve) value, averaging 4%, attesting to the model's performance improvement. Our research offers an efficient model to explore the correlation between pathological image features and molecular characteristics in lung adenocarcinoma patients. This model introduces a novel approach to clinical genetic testing, expected to enhance the efficiency of identifying molecular features and genetic testing in lung adenocarcinoma patients, ultimately providing more accurate and reliable results for related studies.
Collapse
Affiliation(s)
- Kai Sun
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, 250014, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, 250014, China.
| | - Xinbo Yang
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, 250014, China
| | - Weikuan Jia
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, 250014, China.
| |
Collapse
|
16
|
Subramanian V, Syeda-Mahmood T, Do MN. Modelling-based joint embedding of histology and genomics using canonical correlation analysis for breast cancer survival prediction. Artif Intell Med 2024; 149:102787. [PMID: 38462287 DOI: 10.1016/j.artmed.2024.102787] [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: 01/24/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
Traditional approaches to predicting breast cancer patients' survival outcomes were based on clinical subgroups, the PAM50 genes, or the histological tissue's evaluation. With the growth of multi-modality datasets capturing diverse information (such as genomics, histology, radiology and clinical data) about the same cancer, information can be integrated using advanced tools and have improved survival prediction. These methods implicitly exploit the key observation that different modalities originate from the same cancer source and jointly provide a complete picture of the cancer. In this work, we investigate the benefits of explicitly modelling multi-modality data as originating from the same cancer under a probabilistic framework. Specifically, we consider histology and genomics as two modalities originating from the same breast cancer under a probabilistic graphical model (PGM). We construct maximum likelihood estimates of the PGM parameters based on canonical correlation analysis (CCA) and then infer the underlying properties of the cancer patient, such as survival. Equivalently, we construct CCA-based joint embeddings of the two modalities and input them to a learnable predictor. Real-world properties of sparsity and graph-structures are captured in the penalized variants of CCA (pCCA) and are better suited for cancer applications. For generating richer multi-dimensional embeddings with pCCA, we introduce two novel embedding schemes that encourage orthogonality to generate more informative embeddings. The efficacy of our proposed prediction pipeline is first demonstrated via low prediction errors of the hidden variable and the generation of informative embeddings on simulated data. When applied to breast cancer histology and RNA-sequencing expression data from The Cancer Genome Atlas (TCGA), our model can provide survival predictions with average concordance-indices of up to 68.32% along with interpretability. We also illustrate how the pCCA embeddings can be used for survival analysis through Kaplan-Meier curves.
Collapse
Affiliation(s)
- Vaishnavi Subramanian
- Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA.
| | | | - Minh N Do
- Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA
| |
Collapse
|
17
|
Wies C, Schneider L, Haggenmüller S, Bucher TC, Hobelsberger S, Heppt MV, Ferrara G, Krieghoff-Henning EI, Brinker TJ. Evaluating deep learning-based melanoma classification using immunohistochemistry and routine histology: A three center study. PLoS One 2024; 19:e0297146. [PMID: 38241314 PMCID: PMC10798511 DOI: 10.1371/journal.pone.0297146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/28/2023] [Indexed: 01/21/2024] Open
Abstract
Pathologists routinely use immunohistochemical (IHC)-stained tissue slides against MelanA in addition to hematoxylin and eosin (H&E)-stained slides to improve their accuracy in diagnosing melanomas. The use of diagnostic Deep Learning (DL)-based support systems for automated examination of tissue morphology and cellular composition has been well studied in standard H&E-stained tissue slides. In contrast, there are few studies that analyze IHC slides using DL. Therefore, we investigated the separate and joint performance of ResNets trained on MelanA and corresponding H&E-stained slides. The MelanA classifier achieved an area under receiver operating characteristics curve (AUROC) of 0.82 and 0.74 on out of distribution (OOD)-datasets, similar to the H&E-based benchmark classification of 0.81 and 0.75, respectively. A combined classifier using MelanA and H&E achieved AUROCs of 0.85 and 0.81 on the OOD datasets. DL MelanA-based assistance systems show the same performance as the benchmark H&E classification and may be improved by multi stain classification to assist pathologists in their clinical routine.
Collapse
Affiliation(s)
- Christoph Wies
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty, University Heidelberg, Heidelberg, Germany
| | - Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Haggenmüller
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea-Clara Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Hobelsberger
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Markus V. Heppt
- Department of Dermatology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gerardo Ferrara
- Anatomic Pathology and Cytopathology Unit—Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Naples, Italy
| | - Eva I. Krieghoff-Henning
- 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
|
18
|
Tak S, Han G, Leem SH, Lee SY, Paek K, Kim JA. Prediction of anticancer drug resistance using a 3D microfluidic bladder cancer model combined with convolutional neural network-based image analysis. Front Bioeng Biotechnol 2024; 11:1302983. [PMID: 38268938 PMCID: PMC10806080 DOI: 10.3389/fbioe.2023.1302983] [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: 09/27/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024] Open
Abstract
Bladder cancer is the most common urological malignancy worldwide, and its high recurrence rate leads to poor survival outcomes. The effect of anticancer drug treatment varies significantly depending on individual patients and the extent of drug resistance. In this study, we developed a validation system based on an organ-on-a-chip integrated with artificial intelligence technologies to predict resistance to anticancer drugs in bladder cancer. As a proof-of-concept, we utilized the gemcitabine-resistant bladder cancer cell line T24 with four distinct levels of drug resistance (parental, early, intermediate, and late). These cells were co-cultured with endothelial cells in a 3D microfluidic chip. A dataset comprising 2,674 cell images from the chips was analyzed using a convolutional neural network (CNN) to distinguish the extent of drug resistance among the four cell groups. The CNN achieved 95.2% accuracy upon employing data augmentation and a step decay learning rate with an initial value of 0.001. The average diagnostic sensitivity and specificity were 90.5% and 96.8%, respectively, and all area under the curve (AUC) values were over 0.988. Our proposed method demonstrated excellent performance in accurately identifying the extent of drug resistance, which can assist in the prediction of drug responses and in determining the appropriate treatment for bladder cancer patients.
Collapse
Affiliation(s)
- Sungho Tak
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| | - Gyeongjin Han
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Sun-Hee Leem
- Department of Biomedical Sciences, Dong-A University, Busan, Republic of Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
| | - Sang-Yeop Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
Xiao X, Kong Y, Li R, Wang Z, Lu H. Transformer with convolution and graph-node co-embedding: An accurate and interpretable vision backbone for predicting gene expressions from local histopathological image. Med Image Anal 2024; 91:103040. [PMID: 38007979 DOI: 10.1016/j.media.2023.103040] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 11/04/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
Inferring gene expressions from histopathological images has long been a fascinating yet challenging task, primarily due to the substantial disparities between the two modality. Existing strategies using local or global features of histological images are suffering model complexity, GPU consumption, low interpretability, insufficient encoding of local features, and over-smooth prediction of gene expressions among neighboring sites. In this paper, we develop TCGN (Transformer with Convolution and Graph-Node co-embedding method) for gene expression estimation from H&E-stained pathological slide images. TCGN comprises a combination of convolutional layers, transformer encoders, and graph neural networks, and is the first to integrate these blocks in a general and interpretable computer vision backbone. Notably, TCGN uniquely operates with just a single spot image as input for histopathological image analysis, simplifying the process while maintaining interpretability. We validate TCGN on three publicly available spatial transcriptomic datasets. TCGN consistently exhibited the best performance (with median PCC 0.232). TCGN offers superior accuracy while keeping parameters to a minimum (just 86.241 million), and it consumes minimal memory, allowing it to run smoothly even on personal computers. Moreover, TCGN can be extended to handle bulk RNA-seq data while providing the interpretability. Enhancing the accuracy of omics information prediction from pathological images not only establishes a connection between genotype and phenotype, enabling the prediction of costly-to-measure biomarkers from affordable histopathological images, but also lays the groundwork for future multi-modal data modeling. Our results confirm that TCGN is a powerful tool for inferring gene expressions from histopathological images in precision health applications.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, National Center for Translational Medicine, MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China; Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Yan Kong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, National Center for Translational Medicine, MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghan Li
- SJTU-Yale Joint Center for Biostatistics and Data Science, National Center for Translational Medicine, MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China; Zhiyuan College, Shanghai Jiao Tong University, Shanghai, China
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Hui Lu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China; SJTU-Yale Joint Center for Biostatistics and Data Science, National Center for Translational Medicine, MoE Key Lab of Artificial Intelligence, AI Institute, Shanghai Jiao Tong University, Shanghai, China; NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology & Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai Engineering Research Center for Big Data in Pediatric Precision Medicine, Shanghai, China.
| |
Collapse
|
20
|
Ng CW, Wong KK. Deep learning-enabled breast cancer endocrine response determination from H&E staining based on ESR1 signaling activity. Sci Rep 2023; 13:21454. [PMID: 38052873 PMCID: PMC10698147 DOI: 10.1038/s41598-023-48830-x] [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: 06/01/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
Estrogen receptor (ER) positivity by immunohistochemistry has long been a main selection criterium for breast cancer patients to be treated with endocrine therapy. However, ER positivity might not directly correlate with activated ER signaling activity, which is a better predictor for endocrine therapy responsiveness. In this study, we investigated if a deep learning method using whole-slide H&E-stained images could predict ER signaling activity. First, ER signaling activity score was determined using RNAseq data available from each of the 1082 breast cancer samples in the TCGA Pan-Cancer dataset based on the Hallmark Estrogen Response Early gene set from the Molecular Signature Database (MSigDB). Then the processed H&E-stained images and ER signaling activity scores from a training cohort were fed into ResNet101 with three additional fully connected layers to generate a predicted ER activity score. The trained models were subsequently applied to an independent testing cohort. The result demonstrated that ER + /HER2- breast cancer patients with a higher predicted ER activity score had longer progression-free survival (p = 0.0368) than those with lower predicted ER activity score. In conclusion, a convolutional deep neural network can predict prognosis and endocrine therapy response in breast cancer patients based on whole-slide H&E-stained images. The trained models were found to robustly predict the prognosis of ER + /HER2- patients. This information is valuable for patient management, as it does not require RNA-seq or microarray data analyses. Thus, these models can reduce the cost of the diagnosis workflow if such information is required.
Collapse
Affiliation(s)
- Chun Wai Ng
- Department of Gynecologic Oncology and Reproductive Medicine, Unit 1362, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, Unit 1362, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Schacherer DP, Herrmann MD, Clunie DA, Höfener H, Clifford W, Longabaugh WJR, Pieper S, Kikinis R, Fedorov A, Homeyer A. The NCI Imaging Data Commons as a platform for reproducible research in computational pathology. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107839. [PMID: 37832430 PMCID: PMC10841477 DOI: 10.1016/j.cmpb.2023.107839] [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: 04/15/2023] [Revised: 09/20/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND AND OBJECTIVES Reproducibility is a major challenge in developing machine learning (ML)-based solutions in computational pathology (CompPath). The NCI Imaging Data Commons (IDC) provides >120 cancer image collections according to the FAIR principles and is designed to be used with cloud ML services. Here, we explore its potential to facilitate reproducibility in CompPath research. METHODS Using the IDC, we implemented two experiments in which a representative ML-based method for classifying lung tumor tissue was trained and/or evaluated on different datasets. To assess reproducibility, the experiments were run multiple times with separate but identically configured instances of common ML services. RESULTS The results of different runs of the same experiment were reproducible to a large extent. However, we observed occasional, small variations in AUC values, indicating a practical limit to reproducibility. CONCLUSIONS We conclude that the IDC facilitates approaching the reproducibility limit of CompPath research (i) by enabling researchers to reuse exactly the same datasets and (ii) by integrating with cloud ML services so that experiments can be run in identically configured computing environments.
Collapse
Affiliation(s)
- Daniela P Schacherer
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany
| | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Henning Höfener
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany
| | | | | | | | - Ron Kikinis
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrey Fedorov
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - André Homeyer
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Straße 2, 28359 Bremen, Germany.
| |
Collapse
|
22
|
Duroux D, Wohlfart C, Van Steen K, Vladimirova A, King M. Graph-based multi-modality integration for prediction of cancer subtype and severity. Sci Rep 2023; 13:19653. [PMID: 37949935 PMCID: PMC10638406 DOI: 10.1038/s41598-023-46392-6] [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: 07/18/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Personalised cancer screening before therapy paves the way toward improving diagnostic accuracy and treatment outcomes. Most approaches are limited to a single data type and do not consider interactions between features, leaving aside the complementary insights that multimodality and systems biology can provide. In this project, we demonstrate the use of graph theory for data integration via individual networks where nodes and edges are individual-specific. We showcase the consequences of early, intermediate, and late graph-based fusion of RNA-Seq data and histopathology whole-slide images for predicting cancer subtypes and severity. The methodology developed is as follows: (1) we create individual networks; (2) we compute the similarity between individuals from these graphs; (3) we train our model on the similarity matrices; (4) we evaluate the performance using the macro F1 score. Pros and cons of elements of the pipeline are evaluated on publicly available real-life datasets. We find that graph-based methods can increase performance over methods that do not study interactions. Additionally, merging multiple data sources often improves classification compared to models based on single data, especially through intermediate fusion. The proposed workflow can easily be adapted to other disease contexts to accelerate and enhance personalized healthcare.
Collapse
Affiliation(s)
- Diane Duroux
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liège, 4000, Liège, Belgium.
- Post-Doctoral Fellow, ETH AI center, Zürich, Switzerland.
| | | | - Kristel Van Steen
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liège, 4000, Liège, Belgium
- Department of Human Genetics, BIO3 - Systems Medicine, 3000, Leuven, Belgium
| | - Antoaneta Vladimirova
- Roche Information Solutions, Roche Diagnostics Corporation, Santa Clara, California, United States of America
| | | |
Collapse
|
23
|
Sehring J, Dohmen H, Selignow C, Schmid K, Grau S, Stein M, Uhl E, Mukhopadhyay A, Németh A, Amsel D, Acker T. Leveraging Attention-Based Convolutional Neural Networks for Meningioma Classification in Computational Histopathology. Cancers (Basel) 2023; 15:5190. [PMID: 37958364 PMCID: PMC10647687 DOI: 10.3390/cancers15215190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Convolutional neural networks (CNNs) are becoming increasingly valuable tools for advanced computational histopathology, promoting precision medicine through exceptional visual decoding abilities. Meningiomas, the most prevalent primary intracranial tumors, necessitate accurate grading and classification for informed clinical decision-making. Recently, DNA methylation-based molecular classification of meningiomas has proven to be more effective in predicting tumor recurrence than traditional histopathological methods. However, DNA methylation profiling is expensive, labor-intensive, and not widely accessible. Consequently, a digital histology-based prediction of DNA methylation classes would be advantageous, complementing molecular classification. In this study, we developed and rigorously assessed an attention-based multiple-instance deep neural network for predicting meningioma methylation classes using tumor methylome data from 142 (+51) patients and corresponding hematoxylin-eosin-stained histological sections. Pairwise analysis of sample cohorts from three meningioma methylation classes demonstrated high accuracy in two combinations. The performance of our approach was validated using an independent set of 51 meningioma patient samples. Importantly, attention map visualization revealed that the algorithm primarily focuses on tumor regions deemed significant by neuropathologists, offering insights into the decision-making process of the CNN. Our findings highlight the capacity of CNNs to effectively harness phenotypic information from histological sections through computerized images for precision medicine. Notably, this study is the first demonstration of predicting clinically relevant DNA methylome information using computer vision applied to standard histopathology. The introduced AI framework holds great potential in supporting, augmenting, and expediting meningioma classification in the future.
Collapse
Affiliation(s)
- Jannik Sehring
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Hildegard Dohmen
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Carmen Selignow
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Kai Schmid
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Stefan Grau
- Department of Neurosurgery, Hospital Fulda, Pacelliallee 4, D-36043 Fulda, Germany
| | - Marco Stein
- Department of Neurosurgery, University Hospital Gießen, Klinikstr. 33, D-35392 Giessen, Germany
| | - Eberhard Uhl
- Department of Neurosurgery, University Hospital Gießen, Klinikstr. 33, D-35392 Giessen, Germany
| | - Anirban Mukhopadhyay
- Department of Computer Science, Technical University of Darmstadt, Fraunhoferstraße 5, D-64283 Darmstadt, Germany
| | - Attila Németh
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Daniel Amsel
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| | - Till Acker
- Institute of Neuropathology, Justus Liebig University Giessen, Arndtstr. 16, D-35392 Giessen, Germany; (J.S.)
| |
Collapse
|
24
|
Tschodu D, Lippoldt J, Gottheil P, Wegscheider AS, Käs JA, Niendorf A. Re-evaluation of publicly available gene-expression databases using machine-learning yields a maximum prognostic power in breast cancer. Sci Rep 2023; 13:16402. [PMID: 37798300 PMCID: PMC10556090 DOI: 10.1038/s41598-023-41090-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/22/2023] [Indexed: 10/07/2023] Open
Abstract
Gene expression signatures refer to patterns of gene activities and are used to classify different types of cancer, determine prognosis, and guide treatment decisions. Advancements in high-throughput technology and machine learning have led to improvements to predict a patient's prognosis for different cancer phenotypes. However, computational methods for analyzing signatures have not been used to evaluate their prognostic power. Contention remains on the utility of gene expression signatures for prognosis. The prevalent approaches include random signatures, expert knowledge, and machine learning to construct an improved signature. We unify these approaches to evaluate their prognostic power. Re-evaluation of publicly available gene-expression data from 8 databases with 9 machine-learning models revealed previously unreported results. Gene-expression signatures are confirmed to be useful in predicting a patient's prognosis. Convergent evidence from [Formula: see text] 10,000 signatures implicates a maximum prognostic power. By calculating the concordance index, which measures how well patients with different prognoses can be discriminated, we show that a signature can correctly discriminate patients' prognoses no more than 80% of the time. Additionally, we show that more than 50% of the potentially available information is still missing at this value. We surmise that an accurate prognosis must incorporate molecular, clinical, histological, and other complementary factors.
Collapse
Affiliation(s)
- Dimitrij Tschodu
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany.
| | - Jürgen Lippoldt
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany
| | - Pablo Gottheil
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany
| | - Anne-Sophie Wegscheider
- Institute for Histology, Cytology and Molecular Diagnostics, MVZ Prof. Dr. med. A. Niendorf Pathologie Hamburg-West GmbH, 22767, Hamburg, Germany
| | - Josef A Käs
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103, Leipzig, Germany.
| | - Axel Niendorf
- Institute for Histology, Cytology and Molecular Diagnostics, MVZ Prof. Dr. med. A. Niendorf Pathologie Hamburg-West GmbH, 22767, Hamburg, Germany.
| |
Collapse
|
25
|
Carrillo-Perez F, Pizurica M, Ozawa MG, Vogel H, West RB, Kong CS, Herrera LJ, Shen J, Gevaert O. Synthetic whole-slide image tile generation with gene expression profile-infused deep generative models. CELL REPORTS METHODS 2023; 3:100534. [PMID: 37671024 PMCID: PMC10475789 DOI: 10.1016/j.crmeth.2023.100534] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/10/2023] [Accepted: 06/22/2023] [Indexed: 09/07/2023]
Abstract
In this work, we propose an approach to generate whole-slide image (WSI) tiles by using deep generative models infused with matched gene expression profiles. First, we train a variational autoencoder (VAE) that learns a latent, lower-dimensional representation of multi-tissue gene expression profiles. Then, we use this representation to infuse generative adversarial networks (GANs) that generate lung and brain cortex tissue tiles, resulting in a new model that we call RNA-GAN. Tiles generated by RNA-GAN were preferred by expert pathologists compared with tiles generated using traditional GANs, and in addition, RNA-GAN needs fewer training epochs to generate high-quality tiles. Finally, RNA-GAN was able to generalize to gene expression profiles outside of the training set, showing imputation capabilities. A web-based quiz is available for users to play a game distinguishing real and synthetic tiles: https://rna-gan.stanford.edu/, and the code for RNA-GAN is available here: https://github.com/gevaertlab/RNA-GAN.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, 1265 Welch Road, Stanford, CA 94305-547, USA
- Computer Engineering, Automatics and Robotics Department, University of Granada, C. Periodista Daniel Saucedo Aranda, s/n, Granada, 18014 Granada, Spain
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, 1265 Welch Road, Stanford, CA 94305-547, USA
- Internet Technology and Data Science Lab (IDLab), Ghent University, Technologiepark-Zwijnaarde 126, Gent, 9052 Gent, Belgium
| | - Michael G. Ozawa
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Palo Alto, CA 94304, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Palo Alto, CA 94304, USA
| | - Robert B. West
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Palo Alto, CA 94304, USA
| | - Christina S. Kong
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Palo Alto, CA 94304, USA
| | - Luis Javier Herrera
- Computer Engineering, Automatics and Robotics Department, University of Granada, C. Periodista Daniel Saucedo Aranda, s/n, Granada, 18014 Granada, Spain
| | - Jeanne Shen
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr, Palo Alto, CA 94304, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, 1265 Welch Road, Stanford, CA 94305-547, USA
- Department of Biomedical Data Science, Stanford University, School of Medicine, Medical School Office Building (MSOB), 1265 Welch Road, Stanford, CA 94305-547, USA
| |
Collapse
|
26
|
Cui C, Yang H, Wang Y, Zhao S, Asad Z, Coburn LA, Wilson KT, Landman BA, Huo Y. Deep multimodal fusion of image and non-image data in disease diagnosis and prognosis: a review. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2023; 5:10.1088/2516-1091/acc2fe. [PMID: 37360402 PMCID: PMC10288577 DOI: 10.1088/2516-1091/acc2fe] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
The rapid development of diagnostic technologies in healthcare is leading to higher requirements for physicians to handle and integrate the heterogeneous, yet complementary data that are produced during routine practice. For instance, the personalized diagnosis and treatment planning for a single cancer patient relies on various images (e.g. radiology, pathology and camera images) and non-image data (e.g. clinical data and genomic data). However, such decision-making procedures can be subjective, qualitative, and have large inter-subject variabilities. With the recent advances in multimodal deep learning technologies, an increasingly large number of efforts have been devoted to a key question: how do we extract and aggregate multimodal information to ultimately provide more objective, quantitative computer-aided clinical decision making? This paper reviews the recent studies on dealing with such a question. Briefly, this review will include the (a) overview of current multimodal learning workflows, (b) summarization of multimodal fusion methods, (c) discussion of the performance, (d) applications in disease diagnosis and prognosis, and (e) challenges and future directions.
Collapse
Affiliation(s)
- Can Cui
- Department of Computer Science, Vanderbilt University, Nashville, TN 37235, United States of America
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37215, United States of America
| | - Yaohong Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37215, United States of America
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37215, United States of America
| | - Zuhayr Asad
- Department of Computer Science, Vanderbilt University, Nashville, TN 37235, United States of America
| | - Lori A Coburn
- Division of Gastroenterology Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, United States of America
| | - Keith T Wilson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37215, United States of America
- Division of Gastroenterology Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, United States of America
| | - Bennett A Landman
- Department of Computer Science, Vanderbilt University, Nashville, TN 37235, United States of America
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, United States of America
| | - Yuankai Huo
- Department of Computer Science, Vanderbilt University, Nashville, TN 37235, United States of America
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN 37235, United States of America
| |
Collapse
|
27
|
Schneider L, Wies C, Krieghoff-Henning EI, Bucher TC, Utikal JS, Schadendorf D, Brinker TJ. Multimodal integration of image, epigenetic and clinical data to predict BRAF mutation status in melanoma. Eur J Cancer 2023; 183:131-138. [PMID: 36854237 DOI: 10.1016/j.ejca.2023.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND In machine learning, multimodal classifiers can provide more generalised performance than unimodal classifiers. In clinical practice, physicians usually also rely on a range of information from different examinations for diagnosis. In this study, we used BRAF mutation status prediction in melanoma as a model system to analyse the contribution of different data types in a combined classifier because BRAF status can be determined accurately by sequencing as the current gold standard, thus nearly eliminating label noise. METHODS We trained a deep learning-based classifier by combining individually trained random forests of image, clinical and methylation data to predict BRAF-V600 mutation status in primary and metastatic melanomas of The Cancer Genome Atlas cohort. RESULTS With our multimodal approach, we achieved an area under the receiver operating characteristic curve of 0.80, whereas the individual classifiers yielded areas under the receiver operating characteristic curve of 0.63 (histopathologic image data), 0.66 (clinical data) and 0.66 (methylation data) on an independent data set. CONCLUSIONS Our combined approach can predict BRAF status to some extent by identifying BRAF-V600 specific patterns at the histologic, clinical and epigenetic levels. The multimodal classifiers have improved generalisability in predicting BRAF mutation status.
Collapse
Affiliation(s)
- Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christoph Wies
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Eva I Krieghoff-Henning
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Tabea-Clara Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Jochen S Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| |
Collapse
|
28
|
Mohammed MA, Abdulkareem KH, Dinar AM, Zapirain BG. Rise of Deep Learning Clinical Applications and Challenges in Omics Data: A Systematic Review. Diagnostics (Basel) 2023; 13:664. [PMID: 36832152 PMCID: PMC9955380 DOI: 10.3390/diagnostics13040664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
This research aims to review and evaluate the most relevant scientific studies about deep learning (DL) models in the omics field. It also aims to realize the potential of DL techniques in omics data analysis fully by demonstrating this potential and identifying the key challenges that must be addressed. Numerous elements are essential for comprehending numerous studies by surveying the existing literature. For example, the clinical applications and datasets from the literature are essential elements. The published literature highlights the difficulties encountered by other researchers. In addition to looking for other studies, such as guidelines, comparative studies, and review papers, a systematic approach is used to search all relevant publications on omics and DL using different keyword variants. From 2018 to 2022, the search procedure was conducted on four Internet search engines: IEEE Xplore, Web of Science, ScienceDirect, and PubMed. These indexes were chosen because they offer enough coverage and linkages to numerous papers in the biological field. A total of 65 articles were added to the final list. The inclusion and exclusion criteria were specified. Of the 65 publications, 42 are clinical applications of DL in omics data. Furthermore, 16 out of 65 articles comprised the review publications based on single- and multi-omics data from the proposed taxonomy. Finally, only a small number of articles (7/65) were included in papers focusing on comparative analysis and guidelines. The use of DL in studying omics data presented several obstacles related to DL itself, preprocessing procedures, datasets, model validation, and testbed applications. Numerous relevant investigations were performed to address these issues. Unlike other review papers, our study distinctly reflects different observations on omics with DL model areas. We believe that the result of this study can be a useful guideline for practitioners who look for a comprehensive view of the role of DL in omics data analysis.
Collapse
Affiliation(s)
- Mazin Abed Mohammed
- College of Computer Science and Information Technology, University of Anbar, Anbar 31001, Iraq
- eVIDA Lab, University of Deusto, 48007 Bilbao, Spain
| | - Karrar Hameed Abdulkareem
- College of Agriculture, Al-Muthanna University, Samawah 66001, Iraq
- College of Engineering, University of Warith Al-Anbiyaa, Karbala 56001, Iraq
| | - Ahmed M. Dinar
- Computer Engineering Department, University of Technology- Iraq, Baghdad 19006, Iraq
| | | |
Collapse
|
29
|
Li L, Liang Y, Shao M, Lu S, Liao S, Ouyang D. Self-supervised learning-based Multi-Scale feature Fusion Network for survival analysis from whole slide images. Comput Biol Med 2023; 153:106482. [PMID: 36586231 DOI: 10.1016/j.compbiomed.2022.106482] [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: 10/23/2022] [Revised: 12/16/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022]
Abstract
Understanding prognosis and mortality is critical for evaluating the treatment plan of patients. Advances in digital pathology and deep learning techniques have made it practical to perform survival analysis in whole slide images (WSIs). Current methods are usually based on a multi-stage framework which includes patch sampling, feature extraction and prediction. However, the random patch sampling strategy is highly unstable and prone to sampling non-ROI. Feature extraction typically relies on hand-crafted features or convolutional neural networks (CNNs) pre-trained on ImageNet, while the artificial error or domain gaps may affect the survival prediction performance. Besides, the limited information representation of local sampling patches will create a bottleneck limitation on the effectiveness of prediction. To address the above challenges, we propose a novel patch sampling strategy based on image information entropy and construct a Multi-Scale feature Fusion Network (MSFN) based on self-supervised feature extractor. Specifically, we adopt image information entropy as a criterion to select representative sampling patches, thereby avoiding the noise interference caused by random to blank regions. Meanwhile, we pretrain the feature extractor utilizing self-supervised learning mechanism to improve the efficiency of feature extraction. Furthermore, a global-local feature fusion prediction network based on the attention mechanism is constructed to improve the survival prediction effect of WSIs with comprehensive multi-scale information representation. The proposed method is validated by adequate experiments and achieves competitive results on both of the most popular WSIs survival analysis datasets, TCGA-GBM and TCGA-LUSC. Code and trained models are made available at: https://github.com/Mercuriiio/MSFN.
Collapse
Affiliation(s)
- Le Li
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China.
| | - Yong Liang
- Peng Cheng Laboratory, Shenzhen, 518055, China.
| | - Mingwen Shao
- College of Computer Science and Technology, China University of Petroleum, Qingdao 266580, China.
| | - Shanghui Lu
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China.
| | - Shuilin Liao
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China.
| | - Dong Ouyang
- Faculty of Innovation Engineering, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China.
| |
Collapse
|
30
|
Alleman K, Knecht E, Huang J, Zhang L, Lam S, DeCuypere M. Multimodal Deep Learning-Based Prognostication in Glioma Patients: A Systematic Review. Cancers (Basel) 2023; 15:cancers15020545. [PMID: 36672494 PMCID: PMC9856816 DOI: 10.3390/cancers15020545] [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: 09/30/2022] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 01/18/2023] Open
Abstract
Malignant brain tumors pose a substantial burden on morbidity and mortality. As clinical data collection improves, along with the capacity to analyze it, novel predictive clinical tools may improve prognosis prediction. Deep learning (DL) holds promise for integrating clinical data of various modalities. A systematic review of the DL-based prognostication of gliomas was performed using the Embase (Elsevier), PubMed MEDLINE (National library of Medicine), and Scopus (Elsevier) databases, in accordance with PRISMA guidelines. All included studies focused on the prognostication of gliomas, and predicted overall survival (13 studies, 81%), overall survival as well as genotype (2 studies, 12.5%), and response to immunotherapy (1 study, 6.2%). Multimodal analyses were varied, with 6 studies (37.5%) combining MRI with clinical data; 6 studies (37.5%) integrating MRI with histologic, clinical, and biomarker data; 3 studies (18.8%) combining MRI with genomic data; and 1 study (6.2%) combining histologic imaging with clinical data. Studies that compared multimodal models to unimodal-only models demonstrated improved predictive performance. The risk of bias was mixed, most commonly due to inconsistent methodological reporting. Overall, the use of multimodal data in DL assessments of gliomas leads to a more accurate overall survival prediction. However, due to data limitations and a lack of transparency in model and code reporting, the full extent of multimodal DL as a resource for brain tumor patients has not yet been realized.
Collapse
Affiliation(s)
- Kaitlyn Alleman
- Chicago Medical School, Rosalind Franklin University of Science and Medicine, Chicago, IL 60064, USA
| | - Erik Knecht
- Chicago Medical School, Rosalind Franklin University of Science and Medicine, Chicago, IL 60064, USA
| | - Jonathan Huang
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Lu Zhang
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Sandi Lam
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael DeCuypere
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence:
| |
Collapse
|
31
|
Ghaffari Laleh N, Truhn D, Veldhuizen GP, Han T, van Treeck M, Buelow RD, Langer R, Dislich B, Boor P, Schulz V, Kather JN. Adversarial attacks and adversarial robustness in computational pathology. Nat Commun 2022; 13:5711. [PMID: 36175413 PMCID: PMC9522657 DOI: 10.1038/s41467-022-33266-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022] Open
Abstract
Artificial Intelligence (AI) can support diagnostic workflows in oncology by aiding diagnosis and providing biomarkers directly from routine pathology slides. However, AI applications are vulnerable to adversarial attacks. Hence, it is essential to quantify and mitigate this risk before widespread clinical use. Here, we show that convolutional neural networks (CNNs) are highly susceptible to white- and black-box adversarial attacks in clinically relevant weakly-supervised classification tasks. Adversarially robust training and dual batch normalization (DBN) are possible mitigation strategies but require precise knowledge of the type of attack used in the inference. We demonstrate that vision transformers (ViTs) perform equally well compared to CNNs at baseline, but are orders of magnitude more robust to white- and black-box attacks. At a mechanistic level, we show that this is associated with a more robust latent representation of clinically relevant categories in ViTs compared to CNNs. Our results are in line with previous theoretical studies and provide empirical evidence that ViTs are robust learners in computational pathology. This implies that large-scale rollout of AI models in computational pathology should rely on ViTs rather than CNN-based classifiers to provide inherent protection against perturbation of the input data, especially adversarial attacks. Artificial Intelligence can support diagnostic workflows in oncology, but they are vulnerable to adversarial attacks. Here, the authors show that convolutional neural networks are highly susceptible to white- and black-box adversarial attacks in clinically relevant classification tasks.
Collapse
Affiliation(s)
- Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Gregory Patrick Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tianyu Han
- Department of Physics of Molecular Imaging Systems, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany
| | - Roman D Buelow
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland.,Institute of Pathology and Molecular Pathology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Bastian Dislich
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Volkmar Schulz
- Department of Physics of Molecular Imaging Systems, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.,Physics Institute III B, RWTH Aachen University, Aachen, Germany.,Fraunhofer Institute for Digital Medicine MEVIS, Aachen, Germany.,Hyperion Hybrid Imaging Systems GmbH, Aachen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany. .,Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany. .,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany. .,Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK. .,Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
32
|
Li T, Huang H, Zhang S, Zhang Y, Jing H, Sun T, Zhang X, Lu L, Zhang M. Predictive models based on machine learning for bone metastasis in patients with diagnosed colorectal cancer. Front Public Health 2022; 10:984750. [PMID: 36203663 PMCID: PMC9531117 DOI: 10.3389/fpubh.2022.984750] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/25/2022] [Indexed: 01/25/2023] Open
Abstract
Background This study aimed to develop an artificial intelligence predictive model for predicting the probability of developing BM in CRC patients. Methods From SEER database, 50,566 CRC patients were identified between January 2015 and December 2019 without missing data. SVM and LR models were trained and tested on the dataset. Accuracy, area under the curve (AUC), and IDI were used to evaluate and compare the models. Results For bone metastases in the entire cohort, SVM model with poly as kernel function presents the best performance, whose accuracy is 0.908, recall is 0.838, and AUC is 0.926, outperforming LR model. The top three most important factors affecting the model's prediction of BM include extraosseous metastases (EM), CEA, and size. Conclusion Our study developed an SVM model with poly as kernel function for predicting BM in CRC patients. SVM model could improve personalized clinical decision-making, help rationalize the bone metastasis screening process, and reduce the burden on healthcare systems and patients.
Collapse
Affiliation(s)
- Tianhao Li
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Honghong Huang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Shuocun Zhang
- Department of General Surgery, Tianjin Hongqiao Hospital, Tianjin, China
| | - Yongdan Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,Tianjin Institute of Coloproctology, Tianjin, China
| | - Haoren Jing
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,Tianjin Institute of Coloproctology, Tianjin, China
| | - Tianwei Sun
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Xipeng Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,Tianjin Institute of Coloproctology, Tianjin, China,The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China,Nankai University School of Medicine, Nankai University, Tianjin, China,*Correspondence: Xipeng Zhang
| | - Liangfu Lu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China,Liangfu Lu
| | - Mingqing Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China,Tianjin Institute of Coloproctology, Tianjin, China,The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China,Nankai University School of Medicine, Nankai University, Tianjin, China,Mingqing Zhang
| |
Collapse
|
33
|
Qiao Y, Zhao L, Luo C, Luo Y, Wu Y, Li S, Bu D, Zhao Y. Multi-modality artificial intelligence in digital pathology. Brief Bioinform 2022; 23:6702380. [PMID: 36124675 PMCID: PMC9677480 DOI: 10.1093/bib/bbac367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022] Open
Abstract
In common medical procedures, the time-consuming and expensive nature of obtaining test results plagues doctors and patients. Digital pathology research allows using computational technologies to manage data, presenting an opportunity to improve the efficiency of diagnosis and treatment. Artificial intelligence (AI) has a great advantage in the data analytics phase. Extensive research has shown that AI algorithms can produce more up-to-date and standardized conclusions for whole slide images. In conjunction with the development of high-throughput sequencing technologies, algorithms can integrate and analyze data from multiple modalities to explore the correspondence between morphological features and gene expression. This review investigates using the most popular image data, hematoxylin-eosin stained tissue slide images, to find a strategic solution for the imbalance of healthcare resources. The article focuses on the role that the development of deep learning technology has in assisting doctors' work and discusses the opportunities and challenges of AI.
Collapse
Affiliation(s)
- Yixuan Qiao
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lianhe Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| | - Chunlong Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufan Luo
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Wu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Shengtong Li
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dechao Bu
- Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Yi Zhao
- Corresponding authors: Yi Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences; Shandong First Medical University & Shandong Academy of Medical Sciences. Tel.: +86 10 6260 0822; Fax: +86 10 6260 1356; E-mail: ; Lianhe Zhao, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences. Tel.: +86 18513983324; E-mail:
| |
Collapse
|
34
|
Vilgrain I, Martin DK. Letter re: Integration of deep learning-based image analysis and genomic data in cancer pathology: A systematic review: Label-free diagnostic technique to differentiate cancer cells from healthy cells. Eur J Cancer 2022; 172:400-402. [PMID: 35717368 DOI: 10.1016/j.ejca.2022.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Isabelle Vilgrain
- Univ. Grenoble Alpes, UMRS INSERM-CEA-UGA-CNRS U1292, Biology and Biotechnology for Health, Institute of Interdisciplinary Research of Grenoble, Endothelial Cell Junctions in Vascular Disease and Vascular Engineering Team, Grenoble 38054, France.
| | - Donald K Martin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC-SyNaBi, 38000 Grenoble, France.
| |
Collapse
|
35
|
Schneider L, Krieghoff-Henning E, Laiouar-Pedari S, Kuntz S, Hekler A, Kather JN, Gaiser T, Fröhling S, Brinker TJ. Response to letter entitled: Re: Integration of deep learning-based image analysis and genomic data in cancer pathology: A systematic review. Eur J Cancer 2022; 172:403-404. [PMID: 35781181 DOI: 10.1016/j.ejca.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Lucas Schneider
- 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
| | - 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
| | - 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 Tumor 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 Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
36
|
Active surveillance for non-muscle-invasive bladder cancer: fallacy or opportunity? Curr Opin Urol 2022; 32:567-574. [DOI: 10.1097/mou.0000000000001028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|