101
|
Valieris R, Martins L, Defelicibus A, Bueno AP, de Toledo Osorio CAB, Carraro D, Dias-Neto E, Rosales RA, de Figueiredo JMB, Silva ITD. Weakly-supervised deep learning models enable HER2-low prediction from H &E stained slides. Breast Cancer Res 2024; 26:124. [PMID: 39160593 PMCID: PMC11331614 DOI: 10.1186/s13058-024-01863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2)-low breast cancer has emerged as a new subtype of tumor, for which novel antibody-drug conjugates have shown beneficial effects. Assessment of HER2 requires several immunohistochemistry tests with an additional in situ hybridization test if a case is classified as HER2 2+. Therefore, novel cost-effective methods to speed up the HER2 assessment are highly desirable. METHODS We used a self-supervised attention-based weakly supervised method to predict HER2-low directly from 1437 histopathological images from 1351 breast cancer patients. We built six distinct models to explore the ability of classifiers to distinguish between the HER2-negative, HER2-low, and HER2-high classes in different scenarios. The attention-based model was used to comprehend the decision-making process aimed at relevant tissue regions. RESULTS Our results indicate that the effectiveness of classification models hinges on the consistency and dependability of assay-based tests for HER2, as the outcomes from these tests are utilized as the baseline truth for training our models. Through the use of explainable AI, we reveal histologic patterns associated with the HER2 subtypes. CONCLUSION Our findings offer a demonstration of how deep learning technologies can be applied to identify HER2 subgroup statuses, potentially enriching the toolkit available for clinical decision-making in oncology.
Collapse
Affiliation(s)
- Renan Valieris
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Luan Martins
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
- Institute of Mathematics and Computer Sciences, Universidade de São Paulo, São Carlos, São Paulo, 13566-590, Brazil
| | - Alexandre Defelicibus
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Adriana Passos Bueno
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
- Department of Pathology, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | | | - Dirce Carraro
- Laboratory of Genomics and Molecular Biology, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Emmanuel Dias-Neto
- Laboratory Medical Genomics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Rafael A Rosales
- Departamento de Computação e Matemática, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14040-901, Brazil
| | | | - Israel Tojal da Silva
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil.
| |
Collapse
|
102
|
Reitsam NG, Grosser B, Steiner DF, Grozdanov V, Wulczyn E, L'Imperio V, Plass M, Müller H, Zatloukal K, Muti HS, Kather JN, Märkl B. Converging deep learning and human-observed tumor-adipocyte interaction as a biomarker in colorectal cancer. COMMUNICATIONS MEDICINE 2024; 4:163. [PMID: 39147895 PMCID: PMC11327259 DOI: 10.1038/s43856-024-00589-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Tumor-Adipose-Feature (TAF) as well as SARIFA (Stroma AReactive Invasion Front Areas) are two histologic features/biomarkers linking tumor-associated adipocytes to poor outcomes in colorectal cancer (CRC) patients. Whereas TAF was identified by deep learning (DL) algorithms, SARIFA was established as a human-observed histopathologic biomarker. METHODS To study the overlap between TAF and SARIFA, we performed a systematic pathological review of TAF based on all published image tiles. Additionally, we analyzed the presence/absence of TAF in SARIFA-negative CRC cases to elucidate the biologic and prognostic role of a direct tumor-adipocyte contact. TCGA-CRC gene expression data is investigated to assess the association of FABP4 (fatty-acid binding protein 4) and CD36 (fatty-acid translocase) with both TAF and CRC prognosis. RESULTS By investigating the TAF/SARIFA overlap, we show that many TAF patches correspond to the recently described SARIFA-phenomenon. Even though there is a pronounced morphological and biological overlap, there are differences in the concepts. The presence of TAF in SARIFA-negative CRCs is not associated with poor outcomes in this cohort, potentially highlighting the importance of a direct tumor-adipocyte interaction. Upregulation of FABP4 and CD36 gene expression seem both linked to a poor prognosis in CRC. CONCLUSIONS By proving the substantial overlap between human-observed SARIFA and DL-based TAF as morphologic biomarkers, we demonstrate that linking DL-based image features to independently developed histopathologic biomarkers is a promising tool in the identification of clinically and biologically meaningful biomarkers. Adipocyte-tumor-cell interactions seem to be crucial in CRC, which should be considered as biomarkers for further investigations.
Collapse
Affiliation(s)
- Nic G Reitsam
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany.
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany.
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
| | - Bianca Grosser
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | | | | | - Ellery Wulczyn
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, University of Milano-Bicocca, IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Markus Plass
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Heimo Müller
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Kurt Zatloukal
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Hannah S Muti
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Bruno Märkl
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| |
Collapse
|
103
|
Sui Z, Li Z, Sun W. Exploit Spatially Resolved Transcriptomic Data to Infer Cellular Features from Pathology Imaging Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606654. [PMID: 39149252 PMCID: PMC11326158 DOI: 10.1101/2024.08.05.606654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Digital pathology is a rapidly advancing field where deep learning methods can be employed to extract meaningful imaging features. However, the efficacy of training deep learning models is often hindered by the scarcity of annotated pathology images, particularly images with detailed annotations for small image patches or tiles. To overcome this challenge, we propose an innovative approach that leverages paired spatially resolved transcriptomic data to annotate pathology images. We demonstrate the feasibility of this approach and introduce a novel transfer-learning neural network model, STpath (Spatial Transcriptomics and pathology images), designed to predict cell type proportions or classify tumor microenvironments. Our findings reveal that the features from pre-trained deep learning models are associated with cell type identities in pathology image patches. Evaluating STpath using three distinct breast cancer datasets, we observe its promising performance despite the limited training data. STpath excels in samples with variable cell type proportions and high-resolution pathology images. As the influx of spatially resolved transcriptomic data continues, we anticipate ongoing updates to STpath, evolving it into an invaluable AI tool for assisting pathologists in various diagnostic tasks.
Collapse
Affiliation(s)
- Zhining Sui
- Department of Biostatistics and Computational Biology, University of Rochester, 265 Crittenden Blvd. Rochester, 14642, NY, USA
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Avenue, 77030, TX, USA
| | - Wei Sun
- Biostatistics Program, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, 98109, WA, USA
- Department of Biostatistics, University of Washington, 3980 15th Avenue NE, 98195, WA, USA
| |
Collapse
|
104
|
Cai C, Zhou Y, Jiao Y, Li L, Xu J. Prognostic Analysis Combining Histopathological Features and Clinical Information to Predict Colorectal Cancer Survival from Whole-Slide Images. Dig Dis Sci 2024; 69:2985-2995. [PMID: 38837111 DOI: 10.1007/s10620-024-08501-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor within the digestive tract with both a high incidence rate and mortality. Early detection and intervention could improve patient clinical outcomes and survival. METHODS This study computationally investigates a set of prognostic tissue and cell features from diagnostic tissue slides. With the combination of clinical prognostic variables, the pathological image features could predict the prognosis in CRC patients. Our CRC prognosis prediction pipeline sequentially consisted of three modules: (1) A MultiTissue Net to delineate outlines of different tissue types within the WSI of CRC for further ROI selection by pathologists. (2) Development of three-level quantitative image metrics related to tissue compositions, cell shape, and hidden features from a deep network. (3) Fusion of multi-level features to build a prognostic CRC model for predicting survival for CRC. RESULTS Experimental results suggest that each group of features has a particular relationship with the prognosis of patients in the independent test set. In the fusion features combination experiment, the accuracy rate of predicting patients' prognosis and survival status is 81.52%, and the AUC value is 0.77. CONCLUSION This paper constructs a model that can predict the postoperative survival of patients by using image features and clinical information. Some features were found to be associated with the prognosis and survival of patients.
Collapse
Affiliation(s)
- Chengfei Cai
- School of Automation, Nanjing University of Information Science and Technology, Nanjing, 210044, China.
- College of Information Engineering, Taizhou University, Taizhou, 225300, China.
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China.
| | - Yangshu Zhou
- Department of Pathology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280, China
| | - Yiping Jiao
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| | - Liang Li
- Department of Pathology, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Jun Xu
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| |
Collapse
|
105
|
Javed S, Mahmood A, Qaiser T, Werghi N, Rajpoot N. Unsupervised mutual transformer learning for multi-gigapixel Whole Slide Image classification. Med Image Anal 2024; 96:103203. [PMID: 38810517 DOI: 10.1016/j.media.2024.103203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/30/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
The classification of gigapixel Whole Slide Images (WSIs) is an important task in the emerging area of computational pathology. There has been a surge of interest in deep learning models for WSI classification with clinical applications such as cancer detection or prediction of cellular mutations. Most supervised methods require expensive and labor-intensive manual annotations by expert pathologists. Weakly supervised Multiple Instance Learning (MIL) methods have recently demonstrated excellent performance; however, they still require large-scale slide-level labeled training datasets that require a careful inspection of each slide by an expert pathologist. In this work, we propose a fully unsupervised WSI classification algorithm based on mutual transformer learning. The instances (i.e., patches) from gigapixel WSIs are transformed into a latent space and then inverse-transformed to the original space. Using the transformation loss, pseudo labels are generated and cleaned using a transformer label cleaner. The proposed transformer-based pseudo-label generator and cleaner modules mutually train each other iteratively in an unsupervised manner. A discriminative learning mechanism is introduced to improve normal versus cancerous instance labeling. In addition to the unsupervised learning, we demonstrate the effectiveness of the proposed framework for weakly supervised learning and cancer subtype classification as downstream analysis. Extensive experiments on four publicly available datasets show better performance of the proposed algorithm compared to the existing state-of-the-art methods.
Collapse
Affiliation(s)
- Sajid Javed
- Department of Computer Science, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, United Arab Emirates.
| | - Arif Mahmood
- Department of Computer Science, Information Technology University, Lahore, Pakistan.
| | - Talha Qaiser
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
| | - Naoufel Werghi
- Department of Computer Science, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, United Arab Emirates
| | - Nasir Rajpoot
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK; Department of Pathology, University Hospitals Coventry and Warwickshire, Walsgrave, Coventry, CV2 2DX, UK; The Alan Turing Institute, London, NW1 2DB, UK
| |
Collapse
|
106
|
Li X, Zhang Y, Ge F. A mutual reconstruction network model for few-shot classification of histological images: addressing interclass similarity and intraclass diversity. Quant Imaging Med Surg 2024; 14:5443-5459. [PMID: 39144045 PMCID: PMC11320516 DOI: 10.21037/qims-24-253] [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: 02/06/2024] [Accepted: 06/17/2024] [Indexed: 08/16/2024]
Abstract
Background The automated classification of histological images is crucial for the diagnosis of cancer. The limited availability of well-annotated datasets, especially for rare cancers, poses a significant challenge for deep learning methods due to the small number of relevant images. This has led to the development of few-shot learning approaches, which bear considerable clinical importance, as they are designed to overcome the challenges of data scarcity in deep learning for histological image classification. Traditional methods often ignore the challenges of intraclass diversity and interclass similarities in histological images. To address this, we propose a novel mutual reconstruction network model, aimed at meeting these challenges and improving the few-shot classification performance of histological images. Methods The key to our approach is the extraction of subtle and discriminative features. We introduce a feature enhancement module (FEM) and a mutual reconstruction module to increase differences between classes while reducing variance within classes. First, we extract features of support and query images using a feature extractor. These features are then processed by the FEM, which uses a self-attention mechanism for self-reconstruction of features, enhancing the learning of detailed features. These enhanced features are then input into the mutual reconstruction module. This module uses enhanced support features to reconstruct enhanced query features and vice versa. The classification of query samples is based on weighted calculations of the distances between query features and reconstructed query features and between support features and reconstructed support features. Results We extensively evaluated our model using a specially created few-shot histological image dataset. The results showed that in a 5-way 10-shot setup, our model achieved an impressive accuracy of 92.09%. This is a 23.59% improvement in accuracy compared to the model-agnostic meta-learning (MAML) method, which does not focus on fine-grained attributes. In the more challenging, 5-way 1-shot setting, our model also performed well, demonstrating a 18.52% improvement over the ProtoNet, which does not address this challenge. Additional ablation studies indicated the effectiveness and complementary nature of each module and confirmed our method's ability to parse small differences between classes and large variations within classes in histological images. These findings strongly support the superiority of our proposed method in the few-shot classification of histological images. Conclusions The mutual reconstruction network provides outstanding performance in the few-shot classification of histological images, successfully overcoming the challenges of similarities between classes and diversity within classes. This marks a significant advancement in the automated classification of histological images.
Collapse
Affiliation(s)
- Xiangbo Li
- Huitong College, Beijing Normal University, Zhuhai, China
| | - Yinghui Zhang
- College of Education for the Future, Beijing Normal University, Zhuhai, China
| | - Fengxiang Ge
- Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China
| |
Collapse
|
107
|
Paverd H, Zormpas-Petridis K, Clayton H, Burge S, Crispin-Ortuzar M. Radiology and multi-scale data integration for precision oncology. NPJ Precis Oncol 2024; 8:158. [PMID: 39060351 PMCID: PMC11282284 DOI: 10.1038/s41698-024-00656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
In this Perspective paper we explore the potential of integrating radiological imaging with other data types, a critical yet underdeveloped area in comparison to the fusion of other multi-omic data. Radiological images provide a comprehensive, three-dimensional view of cancer, capturing features that would be missed by biopsies or other data modalities. This paper explores the complexities and challenges of incorporating medical imaging into data integration models, in the context of precision oncology. We present the different categories of imaging-omics integration and discuss recent progress, highlighting the opportunities that arise from bringing together spatial data on different scales.
Collapse
Affiliation(s)
- Hania Paverd
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | | | - Hannah Clayton
- Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Sarah Burge
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Mireia Crispin-Ortuzar
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
108
|
Faa G, Coghe F, Pretta A, Castagnola M, Van Eyken P, Saba L, Scartozzi M, Fraschini M. Artificial Intelligence Models for the Detection of Microsatellite Instability from Whole-Slide Imaging of Colorectal Cancer. Diagnostics (Basel) 2024; 14:1605. [PMID: 39125481 PMCID: PMC11311951 DOI: 10.3390/diagnostics14151605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
With the advent of whole-slide imaging (WSI), a technology that can digitally scan whole slides in high resolution, pathology is undergoing a digital revolution. Detecting microsatellite instability (MSI) in colorectal cancer is crucial for proper treatment, as it identifies patients responsible for immunotherapy. Even though universal testing for MSI is recommended, particularly in patients affected by colorectal cancer (CRC), many patients remain untested, and they reside mainly in low-income countries. A critical need exists for accessible, low-cost tools to perform MSI pre-screening. Here, the potential predictive role of the most relevant artificial intelligence-driven models in predicting microsatellite instability directly from histology alone is discussed, focusing on CRC. The role of deep learning (DL) models in identifying the MSI status is here analyzed in the most relevant studies reporting the development of algorithms trained to this end. The most important performance and the most relevant deficiencies are discussed for every AI method. The models proposed for algorithm sharing among multiple research and clinical centers, including federal learning (FL) and swarm learning (SL), are reported. According to all the studies reported here, AI models are valuable tools for predicting MSI status on WSI alone in CRC. The use of digitized H&E-stained sections and a trained algorithm allow the extraction of relevant molecular information, such as MSI status, in a short time and at a low cost. The possible advantages related to introducing DL methods in routine surgical pathology are underlined here, and the acceleration of the digital transformation of pathology departments and services is recommended.
Collapse
Affiliation(s)
- Gavino Faa
- Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, 09123 Cagliari, Italy;
| | - Ferdinando Coghe
- UOC Laboratorio Analisi, AOU of Cagliari, 09123 Cagliari, Italy;
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (A.P.); (M.S.)
| | - Massimo Castagnola
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Peter Van Eyken
- Division of Pathology, Genk Regional Hospital, 3600 Genk, Belgium;
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, University of Cagliari, 40138 Cagliari, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (A.P.); (M.S.)
| | - Matteo Fraschini
- Dipartimento di Ingegneria Elettrica ed Elettronica, University of Cagliari, 09123 Cagliari, Italy
| |
Collapse
|
109
|
Jaume G, Peeters T, Song AH, Pettit R, Williamson DFK, Oldenburg L, Vaidya A, de Brot S, Chen RJ, Thiran JP, Le LP, Gerber G, Mahmood F. AI-driven Discovery of Morphomolecular Signatures in Toxicology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604355. [PMID: 39091765 PMCID: PMC11291055 DOI: 10.1101/2024.07.19.604355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Early identification of drug toxicity is essential yet challenging in drug development. At the preclinical stage, toxicity is assessed with histopathological examination of tissue sections from animal models to detect morphological lesions. To complement this analysis, toxicogenomics is increasingly employed to understand the mechanism of action of the compound and ultimately identify lesion-specific safety biomarkers for which in vitro assays can be designed. However, existing works that aim to identify morphological correlates of expression changes rely on qualitative or semi-quantitative morphological characterization and remain limited in scale or morphological diversity. Artificial intelligence (AI) offers a promising approach for quantitatively modeling this relationship at an unprecedented scale. Here, we introduce GEESE, an AI model designed to impute morphomolecular signatures in toxicology data. Our model was trained to predict 1,536 gene targets on a cohort of 8,231 hematoxylin and eosin-stained liver sections from Rattus norvegicus across 127 preclinical toxicity studies. The model, evaluated on 2,002 tissue sections from 29 held-out studies, can yield pseudo-spatially resolved gene expression maps, which we correlate with six key drug-induced liver injuries (DILI). From the resulting 25 million lesion-expression pairs, we established quantitative relations between up and downregulated genes and lesions. Validation of these signatures against toxicogenomic databases, pathway enrichment analyses, and human hepatocyte cell lines asserted their relevance. Overall, our study introduces new methods for characterizing toxicity at an unprecedented scale and granularity, paving the way for AI-driven discovery of toxicity biomarkers.
Collapse
Affiliation(s)
- Guillaume Jaume
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Thomas Peeters
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Signal Processing Laboratory, EPFL, Lausanne, Switzerland
| | - Andrew H. Song
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | - Rowland Pettit
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Drew F. K. Williamson
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lukas Oldenburg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Anurag Vaidya
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Simone de Brot
- Institute of Animal Pathology, Vetsuisse, University of Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Switzerland
| | - Richard J. Chen
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
| | | | - Long Phi Le
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| | - Georg Gerber
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Health Sciences and Technology, Harvard-MIT, Cambridge, MA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA
| |
Collapse
|
110
|
Park IG, Yoon SJ, Won SM, Oh KK, Hyun JY, Suk KT, Lee U. Gut microbiota-based machine-learning signature for the diagnosis of alcohol-associated and metabolic dysfunction-associated steatotic liver disease. Sci Rep 2024; 14:16122. [PMID: 38997279 PMCID: PMC11245548 DOI: 10.1038/s41598-024-60768-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/26/2024] [Indexed: 07/14/2024] Open
Abstract
Alcoholic-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD) show a high prevalence rate worldwide. As gut microbiota represents current state of ALD and MASLD via gut-liver axis, typical characteristics of gut microbiota can be used as a potential diagnostic marker in ALD and MASLD. Machine learning (ML) algorithms improve diagnostic performance in various diseases. Using gut microbiota-based ML algorithms, we evaluated the diagnostic index for ALD and MASLD. Fecal 16S rRNA sequencing data of 263 ALD (control, elevated liver enzyme [ELE], cirrhosis, and hepatocellular carcinoma [HCC]) and 201 MASLD (control and ELE) subjects were collected. For external validation, 126 ALD and 84 MASLD subjects were recruited. Four supervised ML algorithms (support vector machine, random forest, multilevel perceptron, and convolutional neural network) were used for classification with 20, 40, 60, and 80 features, in which three nonsupervised ML algorithms (independent component analysis, principal component analysis, linear discriminant analysis, and random projection) were used for feature reduction. A total of 52 combinations of ML algorithms for each pair of subgroups were performed with 60 hyperparameter variations and Stratified ShuffleSplit tenfold cross validation. The ML models of the convolutional neural network combined with principal component analysis achieved areas under the receiver operating characteristic curve (AUCs) > 0.90. In ALD, the diagnostic AUC values of the ML strategy (vs. control) were 0.94, 0.97, and 0.96 for ELE, cirrhosis, and liver cancer, respectively. The AUC value (vs. control) for MASLD (ELE) was 0.93. In the external validation, the AUC values of ALD and MASLD (vs control) were > 0.90 and 0.88, respectively. The gut microbiota-based ML strategy can be used for the diagnosis of ALD and MASLD.ClinicalTrials.gov NCT04339725.
Collapse
Affiliation(s)
- In-Gyu Park
- Department of Electrical Engineering, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Sung-Min Won
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ji Ye Hyun
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, 24253, Republic of Korea.
- Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea.
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Gyo-dong, Chuncheon, 24253, Republic of Korea.
| |
Collapse
|
111
|
Ding GY, Tan WM, Lin YP, Ling Y, Huang W, Zhang S, Shi JY, Luo RK, Ji Y, Wang XY, Zhou J, Fan J, Cai MY, Yan B, Gao Q. Mining the interpretable prognostic features from pathological image of intrahepatic cholangiocarcinoma using multi-modal deep learning. BMC Med 2024; 22:282. [PMID: 38972973 PMCID: PMC11229270 DOI: 10.1186/s12916-024-03482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND The advances in deep learning-based pathological image analysis have invoked tremendous insights into cancer prognostication. Still, lack of interpretability remains a significant barrier to clinical application. METHODS We established an integrative prognostic neural network for intrahepatic cholangiocarcinoma (iCCA), towards a comprehensive evaluation of both architectural and fine-grained information from whole-slide images. Then, leveraging on multi-modal data, we conducted extensive interrogative approaches to the models, to extract and visualize the morphological features that most correlated with clinical outcome and underlying molecular alterations. RESULTS The models were developed and optimized on 373 iCCA patients from our center and demonstrated consistent accuracy and robustness on both internal (n = 213) and external (n = 168) cohorts. The occlusion sensitivity map revealed that the distribution of tertiary lymphoid structures, the geometric traits of the invasive margin, the relative composition of tumor parenchyma and stroma, the extent of necrosis, the presence of the disseminated foci, and the tumor-adjacent micro-vessels were the determining architectural features that impacted on prognosis. Quantifiable morphological vector extracted by CellProfiler demonstrated that tumor nuclei from high-risk patients exhibited significant larger size, more distorted shape, with less prominent nuclear envelope and textural contrast. The multi-omics data (n = 187) further revealed key molecular alterations left morphological imprints that could be attended by the network, including glycolysis, hypoxia, apical junction, mTORC1 signaling, and immune infiltration. CONCLUSIONS We proposed an interpretable deep-learning framework to gain insights into the biological behavior of iCCA. Most of the significant morphological prognosticators perceived by the network are comprehensible to human minds.
Collapse
Affiliation(s)
- Guang-Yu Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Wei-Min Tan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China
| | - You-Pei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Yu Ling
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Jie-Yi Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Rong-Kui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Ying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Mu-Yan Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No.651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Bo Yan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, No.2005, Song Hu Road, Shanghai, 200433, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, No.180, Feng Lin Road, Shanghai, 200032, China.
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
112
|
Osorio P, Jimenez-Perez G, Montalt-Tordera J, Hooge J, Duran-Ballester G, Singh S, Radbruch M, Bach U, Schroeder S, Siudak K, Vienenkoetter J, Lawrenz B, Mohammadi S. Latent Diffusion Models with Image-Derived Annotations for Enhanced AI-Assisted Cancer Diagnosis in Histopathology. Diagnostics (Basel) 2024; 14:1442. [PMID: 39001331 PMCID: PMC11241396 DOI: 10.3390/diagnostics14131442] [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: 05/03/2024] [Revised: 06/19/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Artificial Intelligence (AI)-based image analysis has immense potential to support diagnostic histopathology, including cancer diagnostics. However, developing supervised AI methods requires large-scale annotated datasets. A potentially powerful solution is to augment training data with synthetic data. Latent diffusion models, which can generate high-quality, diverse synthetic images, are promising. However, the most common implementations rely on detailed textual descriptions, which are not generally available in this domain. This work proposes a method that constructs structured textual prompts from automatically extracted image features. We experiment with the PCam dataset, composed of tissue patches only loosely annotated as healthy or cancerous. We show that including image-derived features in the prompt, as opposed to only healthy and cancerous labels, improves the Fréchet Inception Distance (FID) by 88.6. We also show that pathologists find it challenging to detect synthetic images, with a median sensitivity/specificity of 0.55/0.55. Finally, we show that synthetic data effectively train AI models.
Collapse
Affiliation(s)
- Pedro Osorio
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | | | | | - Jens Hooge
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | | | - Shivam Singh
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | - Moritz Radbruch
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | - Ute Bach
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | | | - Krystyna Siudak
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | | | - Bettina Lawrenz
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | - Sadegh Mohammadi
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| |
Collapse
|
113
|
Jung JO, Pisula JI, Beyerlein X, Lukomski L, Knipper K, Abu Hejleh AP, Fuchs HF, Tolkach Y, Chon SH, Nienhüser H, Büchler MW, Bruns CJ, Quaas A, Bozek K, Popp F, Schmidt T. Deep Learning Histology for Prediction of Lymph Node Metastases and Tumor Regression after Neoadjuvant FLOT Therapy of Gastroesophageal Adenocarcinoma. Cancers (Basel) 2024; 16:2445. [PMID: 39001507 PMCID: PMC11240557 DOI: 10.3390/cancers16132445] [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: 06/04/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The aim of this study was to establish a deep learning prediction model for neoadjuvant FLOT chemotherapy response. The neural network utilized clinical data and visual information from whole-slide images (WSIs) of therapy-naïve gastroesophageal cancer biopsies. METHODS This study included 78 patients from the University Hospital of Cologne and 59 patients from the University Hospital of Heidelberg used as external validation. RESULTS After surgical resection, 33 patients from Cologne (42.3%) were ypN0 and 45 patients (57.7%) were ypN+, while 23 patients from Heidelberg (39.0%) were ypN0 and 36 patients (61.0%) were ypN+ (p = 0.695). The neural network had an accuracy of 92.1% to predict lymph node metastasis and the area under the curve (AUC) was 0.726. A total of 43 patients from Cologne (55.1%) had less than 50% residual vital tumor (RVT) compared to 34 patients from Heidelberg (57.6%, p = 0.955). The model was able to predict tumor regression with an error of ±14.1% and an AUC of 0.648. CONCLUSIONS This study demonstrates that visual features extracted by deep learning from therapy-naïve biopsies of gastroesophageal adenocarcinomas correlate with positive lymph nodes and tumor regression. The results will be confirmed in prospective studies to achieve early allocation of patients to the most promising treatment.
Collapse
Affiliation(s)
- Jin-On Jung
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Juan I. Pisula
- Data Science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Xenia Beyerlein
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Leandra Lukomski
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Karl Knipper
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Aram P. Abu Hejleh
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Hans F. Fuchs
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Yuri Tolkach
- Institute of Pathology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Henrik Nienhüser
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Markus W. Büchler
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Christiane J. Bruns
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Katarzyna Bozek
- Data Science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Felix Popp
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Thomas Schmidt
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| |
Collapse
|
114
|
Wu Z, Yang Y, Chen M, Zha Y. Matrix metalloproteinase 9 expression and glioblastoma survival prediction using machine learning on digital pathological images. Sci Rep 2024; 14:15065. [PMID: 38956384 PMCID: PMC11220146 DOI: 10.1038/s41598-024-66105-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: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
This study aimed to apply pathomics to predict Matrix metalloproteinase 9 (MMP9) expression in glioblastoma (GBM) and investigate the underlying molecular mechanisms associated with pathomics. Here, we included 127 GBM patients, 78 of whom were randomly allocated to the training and test cohorts for pathomics modeling. The prognostic significance of MMP9 was assessed using Kaplan-Meier and Cox regression analyses. PyRadiomics was used to extract the features of H&E-stained whole slide images. Feature selection was performed using the maximum relevance and minimum redundancy (mRMR) and recursive feature elimination (RFE) algorithms. Prediction models were created using support vector machines (SVM) and logistic regression (LR). The performance was assessed using ROC analysis, calibration curve assessment, and decision curve analysis. MMP9 expression was elevated in patients with GBM. This was an independent prognostic factor for GBM. Six features were selected for the pathomics model. The area under the curves (AUCs) of the training and test subsets were 0.828 and 0.808, respectively, for the SVM model and 0.778 and 0.754, respectively, for the LR model. The C-index and calibration plots exhibited effective estimation abilities. The pathomics score calculated using the SVM model was highly correlated with overall survival time. These findings indicate that MMP9 plays a crucial role in GBM development and prognosis. Our pathomics model demonstrated high efficacy for predicting MMP9 expression levels and prognosis of patients with GBM.
Collapse
Affiliation(s)
- Zijun Wu
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Maojuan Chen
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Yunfei Zha
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
115
|
Lecuelle J, Truntzer C, Basile D, Laghi L, Greco L, Ilie A, Rageot D, Emile JF, Bibeau F, Taïeb J, Derangere V, Lepage C, Ghiringhelli F. Machine learning evaluation of immune infiltrate through digital tumour score allows prediction of survival outcome in a pooled analysis of three international stage III colon cancer cohorts. EBioMedicine 2024; 105:105207. [PMID: 38880067 PMCID: PMC11233898 DOI: 10.1016/j.ebiom.2024.105207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND T-cell immune infiltrates are robust prognostic variables in localised colon cancer. Evaluation of prognosis using artificial intelligence is an emerging field. We evaluated whether machine learning analysis improved prediction of patient outcome in comparison with analysis of T cell infiltrate only or in association with clinical variables. METHODS We used data from two phase III clinical trials (Prodige-13 and PETACC08) and one retrospective Italian cohort (HARMONY). Cohorts were split into training (N = 692), internal validation (N = 297) and external validation (N = 672) sets. Tumour slides were stained with CD3mAb. CD3 Machine Learning (CD3ML) score was computed using graphical parameters within the tumour tiles obtained from CD3 slides. CD3 infiltrates in tumour core and invasive margin were automatically detected. Associations of CD3 infiltrates and CD3ML with 5-year Disease-Free Survival (DFS) were examined using univariate and multivariable survival models by Cox regression. FINDINGS CD3 density both in the invasive margin and the tumour core were significantly associated with DFS in the different sets. Similarly, CD3ML score was significantly associated with DFS in all sets. CD3 assessment did not provide added value on top of CD3ML assessment (Likelihood Ratio Test (LRT), p = 0.13). In contrast, CD3ML improved prediction of DFS when combined with a clinical risk stage (LRT, p = 0.001). Stratified by clinical risk score (High or Low), patients with low CD3ML score had better DFS. INTERPRETATION In all tested sets, machine learning analysis of tumour cells improved prediction of prognosis compared to clinical parameters. Adding tumour-infiltrating lymphocytes assessment did not improve prognostic determination. FUNDING This research received no external funding.
Collapse
Affiliation(s)
- Julie Lecuelle
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Caroline Truntzer
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Debora Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, Crotone, Italy
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luana Greco
- Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alis Ilie
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - David Rageot
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Jean-François Emile
- Paris-Saclay University, Versailles SQY University (UVSQ), EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Ambroise Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| | - Fréderic Bibeau
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Normandie Université, Caen, France; Department of Pathology, Besançon University Hospital, Besançon, France
| | - Julien Taïeb
- Institut du Cancer Paris Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique, Sorbonne Université, Université Sorbonne Paris Cité, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Valentin Derangere
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France
| | - Come Lepage
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Fédération Francophone de Cancérologie Digestive, Centre de Randomisation Gestion Analyse, EPICAD LNC 1231, Dijon, France; Service d'Hépato-gastroentérologie et Oncologie digestive, CHU de Dijon, France
| | - François Ghiringhelli
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France.
| |
Collapse
|
116
|
Cheng W, Deng L, Liu W. A commentary on 'A CT-based multitask deep learning model for predicting tumor stroma ratio and treatment outcomes in patients with colorectal cancer: a multicenter cohort study'. Int J Surg 2024; 110:4496-4497. [PMID: 38661554 PMCID: PMC11254226 DOI: 10.1097/js9.0000000000001431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024]
Affiliation(s)
| | | | - Wen Liu
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
117
|
Laury AR, Zheng S, Aho N, Fallegger R, Hänninen S, Saez-Rodriguez J, Tanevski J, Youssef O, Tang J, Carpén OM. Opening the Black Box: Spatial Transcriptomics and the Relevance of Artificial Intelligence-Detected Prognostic Regions in High-Grade Serous Carcinoma. Mod Pathol 2024; 37:100508. [PMID: 38704029 DOI: 10.1016/j.modpat.2024.100508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/04/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Image-based deep learning models are used to extract new information from standard hematoxylin and eosin pathology slides; however, biological interpretation of the features detected by artificial intelligence (AI) remains a challenge. High-grade serous carcinoma of the ovary (HGSC) is characterized by aggressive behavior and chemotherapy resistance, but also exhibits striking variability in outcome. Our understanding of this disease is limited, partly due to considerable tumor heterogeneity. We previously trained an AI model to identify HGSC tumor regions that are highly associated with outcome status but are indistinguishable by conventional morphologic methods. Here, we applied spatially resolved transcriptomics to further profile the AI-identified tumor regions in 16 patients (8 per outcome group) and identify molecular features related to disease outcome in patients who underwent primary debulking surgery and platinum-based chemotherapy. We examined formalin-fixed paraffin-embedded tissue from (1) regions identified by the AI model as highly associated with short or extended chemotherapy response, and (2) background tumor regions (not identified by the AI model as highly associated with outcome status) from the same tumors. We show that the transcriptomic profiles of AI-identified regions are more distinct than background regions from the same tumors, are superior in predicting outcome, and differ in several pathways including those associated with chemoresistance in HGSC. Further, we find that poor outcome and good outcome regions are enriched by different tumor subpopulations, suggesting distinctive interaction patterns. In summary, our work presents proof of concept that AI-guided spatial transcriptomic analysis improves recognition of biologic features relevant to patient outcomes.
Collapse
Affiliation(s)
- Anna Ray Laury
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.
| | - Shuyu Zheng
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Niina Aho
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Robin Fallegger
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Satu Hänninen
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Jovan Tanevski
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany; Department of Knowledge Technologies, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Omar Youssef
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinical and Chemical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Jing Tang
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Olli Mikael Carpén
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| |
Collapse
|
118
|
Shahamatdar S, Saeed-Vafa D, Linsley D, Khalil F, Lovinger K, Li L, McLeod HT, Ramachandran S, Serre T. Deceptive learning in histopathology. Histopathology 2024; 85:116-132. [PMID: 38556922 PMCID: PMC11162337 DOI: 10.1111/his.15180] [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: 02/01/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
AIMS Deep learning holds immense potential for histopathology, automating tasks that are simple for expert pathologists and revealing novel biology for tasks that were previously considered difficult or impossible to solve by eye alone. However, the extent to which the visual strategies learned by deep learning models in histopathological analysis are trustworthy or not has yet to be systematically analysed. Here, we systematically evaluate deep neural networks (DNNs) trained for histopathological analysis in order to understand if their learned strategies are trustworthy or deceptive. METHODS AND RESULTS We trained a variety of DNNs on a novel data set of 221 whole-slide images (WSIs) from lung adenocarcinoma patients, and evaluated their effectiveness at (1) molecular profiling of KRAS versus EGFR mutations, (2) determining the primary tissue of a tumour and (3) tumour detection. While DNNs achieved above-chance performance on molecular profiling, they did so by exploiting correlations between histological subtypes and mutations, and failed to generalise to a challenging test set obtained through laser capture microdissection (LCM). In contrast, DNNs learned robust and trustworthy strategies for determining the primary tissue of a tumour as well as detecting and localising tumours in tissue. CONCLUSIONS Our work demonstrates that DNNs hold immense promise for aiding pathologists in analysing tissue. However, they are also capable of achieving seemingly strong performance by learning deceptive strategies that leverage spurious correlations, and are ultimately unsuitable for research or clinical work. The framework we propose for model evaluation and interpretation is an important step towards developing reliable automated systems for histopathological analysis.
Collapse
Affiliation(s)
- Sahar Shahamatdar
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
- The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Daryoush Saeed-Vafa
- Department of Anatomic Pathology, H. Lee Moffitt Cancer and Research Institute, Tampa, FL, USA
| | - Drew Linsley
- Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Department of Cognitive Linguistic & Psychological Sciences, Brown University, Providence, RI, USA
| | - Farah Khalil
- Department of Anatomic Pathology, H. Lee Moffitt Cancer and Research Institute, Tampa, FL, USA
| | - Katherine Lovinger
- Department of Molecular Biology, H. Lee Moffitt Cancer and Research Institute, Tampa, FL, USA
| | - Lester Li
- University of Rochester, Rochester, NY, USA
| | | | - Sohini Ramachandran
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI, USA
- The Data Science Initiative, Brown University, Providence, RI, USA
| | - Thomas Serre
- Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Department of Cognitive Linguistic & Psychological Sciences, Brown University, Providence, RI, USA
| |
Collapse
|
119
|
Hoang DT, Shulman ED, Turakulov R, Abdullaev Z, Singh O, Campagnolo EM, Lalchungnunga H, Stone EA, Nasrallah MP, Ruppin E, Aldape K. Prediction of DNA methylation-based tumor types from histopathology in central nervous system tumors with deep learning. Nat Med 2024; 30:1952-1961. [PMID: 38760587 DOI: 10.1038/s41591-024-02995-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024]
Abstract
Precision in the diagnosis of diverse central nervous system (CNS) tumor types is crucial for optimal treatment. DNA methylation profiles, which capture the methylation status of thousands of individual CpG sites, are state-of-the-art data-driven means to enhance diagnostic accuracy but are also time consuming and not widely available. Here, to address these limitations, we developed Deep lEarning from histoPathoLOgy and methYlation (DEPLOY), a deep learning model that classifies CNS tumors to ten major categories from histopathology. DEPLOY integrates three distinct components: the first classifies CNS tumors directly from slide images ('direct model'), the second initially generates predictions for DNA methylation beta values, which are subsequently used for tumor classification ('indirect model'), and the third classifies tumor types directly from routinely available patient demographics. First, we find that DEPLOY accurately predicts beta values from histopathology images. Second, using a ten-class model trained on an internal dataset of 1,796 patients, we predict the tumor categories in three independent external test datasets including 2,156 patients, achieving an overall accuracy of 95% and balanced accuracy of 91% on samples that are predicted with high confidence. These results showcase the potential future use of DEPLOY to assist pathologists in diagnosing CNS tumors within a clinically relevant short time frame.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rust Turakulov
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Omkar Singh
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emma M Campagnolo
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - H Lalchungnunga
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Eric A Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - MacLean P Nasrallah
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
120
|
Raufeisen J, Xie K, Hörst F, Braunschweig T, Li J, Kleesiek J, Röhrig R, Egger J, Leibe B, Hölzle F, Hermans A, Puladi B. Cyto R-CNN and CytoNuke Dataset: Towards reliable whole-cell segmentation in bright-field histological images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 252:108215. [PMID: 38781811 DOI: 10.1016/j.cmpb.2024.108215] [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: 02/06/2024] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Cell segmentation in bright-field histological slides is a crucial topic in medical image analysis. Having access to accurate segmentation allows researchers to examine the relationship between cellular morphology and clinical observations. Unfortunately, most segmentation methods known today are limited to nuclei and cannot segment the cytoplasm. METHODS We present a new network architecture Cyto R-CNN that is able to accurately segment whole cells (with both the nucleus and the cytoplasm) in bright-field images. We also present a new dataset CytoNuke, consisting of multiple thousand manual annotations of head and neck squamous cell carcinoma cells. Utilizing this dataset, we compared the performance of Cyto R-CNN to other popular cell segmentation algorithms, including QuPath's built-in algorithm, StarDist, Cellpose and a multi-scale Attention Deeplabv3+. To evaluate segmentation performance, we calculated AP50, AP75 and measured 17 morphological and staining-related features for all detected cells. We compared these measurements to the gold standard of manual segmentation using the Kolmogorov-Smirnov test. RESULTS Cyto R-CNN achieved an AP50 of 58.65% and an AP75 of 11.56% in whole-cell segmentation, outperforming all other methods (QuPath 19.46/0.91%; StarDist 45.33/2.32%; Cellpose 31.85/5.61%, Deeplabv3+ 3.97/1.01%). Cell features derived from Cyto R-CNN showed the best agreement to the gold standard (D¯=0.15) outperforming QuPath (D¯=0.22), StarDist (D¯=0.25), Cellpose (D¯=0.23) and Deeplabv3+ (D¯=0.33). CONCLUSION Our newly proposed Cyto R-CNN architecture outperforms current algorithms in whole-cell segmentation while providing more reliable cell measurements than any other model. This could improve digital pathology workflows, potentially leading to improved diagnosis. Moreover, our published dataset can be used to develop further models in the future.
Collapse
Affiliation(s)
- Johannes Raufeisen
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Institute of Medical Informatics, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany
| | - Kunpeng Xie
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Institute of Medical Informatics, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany
| | - Fabian Hörst
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131 Essen, Germany; Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Hufelandstr. 55, 45147 Essen, Germany
| | - Till Braunschweig
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany; Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Institute of Pathology, LMU Munich, Thalkirchner Str. 36, 80337 Munich, Germany
| | - Jianning Li
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131 Essen, Germany; Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Hufelandstr. 55, 45147 Essen, Germany
| | - Jens Kleesiek
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131 Essen, Germany; Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Hufelandstr. 55, 45147 Essen, Germany; Department of Physics, TU Dortmund University, August-Schmidt-Str. 4, 44227 Dortmund, Germany
| | - Rainer Röhrig
- Institute of Medical Informatics, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany
| | - Jan Egger
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131 Essen, Germany; Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Hufelandstr. 55, 45147 Essen, Germany; Center for Virtual and Extended Reality in Medicine (ZvRM), University Hospital Essen, University Medicine Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Bastian Leibe
- Visual Computing Institute (Computer Vision), RWTH Aachen University, Mies-van-der-Rohe Str. 15, 52074 Aachen, Germany
| | - Frank Hölzle
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany
| | - Alexander Hermans
- Visual Computing Institute (Computer Vision), RWTH Aachen University, Mies-van-der-Rohe Str. 15, 52074 Aachen, Germany
| | - Behrus Puladi
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Institute of Medical Informatics, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Pauwelsstraße 30, 52074 Aachen, Germany.
| |
Collapse
|
121
|
Schäfer R, Nicke T, Höfener H, Lange A, Merhof D, Feuerhake F, Schulz V, Lotz J, Kiessling F. Overcoming data scarcity in biomedical imaging with a foundational multi-task model. NATURE COMPUTATIONAL SCIENCE 2024; 4:495-509. [PMID: 39030386 PMCID: PMC11288886 DOI: 10.1038/s43588-024-00662-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/17/2024] [Indexed: 07/21/2024]
Abstract
Foundational models, pretrained on a large scale, have demonstrated substantial success across non-medical domains. However, training these models typically requires large, comprehensive datasets, which contrasts with the smaller and more specialized datasets common in biomedical imaging. Here we propose a multi-task learning strategy that decouples the number of training tasks from memory requirements. We trained a universal biomedical pretrained model (UMedPT) on a multi-task database including tomographic, microscopic and X-ray images, with various labeling strategies such as classification, segmentation and object detection. The UMedPT foundational model outperformed ImageNet pretraining and previous state-of-the-art models. For classification tasks related to the pretraining database, it maintained its performance with only 1% of the original training data and without fine-tuning. For out-of-domain tasks it required only 50% of the original training data. In an external independent validation, imaging features extracted using UMedPT proved to set a new standard for cross-center transferability.
Collapse
Affiliation(s)
- Raphael Schäfer
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Till Nicke
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Henning Höfener
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Annkristin Lange
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Dorit Merhof
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
- Institute of Image Analysis and Computer Vision, Faculty of Informatics and Data Science, University of Regensburg, Regensburg, Germany
| | - Friedrich Feuerhake
- Institute for Pathology, Hannover Medical School, Hanover, Germany
- Institute for Neuropathology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Volkmar Schulz
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Johannes Lotz
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany.
| | - Fabian Kiessling
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany.
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
122
|
Bülow RD, Lan YC, Amann K, Boor P. [Artificial intelligence in kidney transplant pathology]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:277-283. [PMID: 38598097 DOI: 10.1007/s00292-024-01324-7] [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] [Accepted: 03/12/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Artificial intelligence (AI) systems have showed promising results in digital pathology, including digital nephropathology and specifically also kidney transplant pathology. AIM Summarize the current state of research and limitations in the field of AI in kidney transplant pathology diagnostics and provide a future outlook. MATERIALS AND METHODS Literature search in PubMed and Web of Science using the search terms "deep learning", "transplant", and "kidney". Based on these results and studies cited in the identified literature, a selection was made of studies that have a histopathological focus and use AI to improve kidney transplant diagnostics. RESULTS AND CONCLUSION Many studies have already made important contributions, particularly to the automation of the quantification of some histopathological lesions in nephropathology. This likely can be extended to automatically quantify all relevant lesions for a kidney transplant, such as Banff lesions. Important limitations and challenges exist in the collection of representative data sets and the updates of Banff classification, making large-scale studies challenging. The already positive study results make future AI support in kidney transplant pathology appear likely.
Collapse
Affiliation(s)
- Roman David Bülow
- Institut für Pathologie, Sektion Nephropathologie, Universitätsklinikum RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Deutschland
| | - Yu-Chia Lan
- Institut für Pathologie, Sektion Nephropathologie, Universitätsklinikum RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Deutschland
| | - Kerstin Amann
- Abteilung Nephropathologie, Institut für Pathologie, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Deutschland
| | - Peter Boor
- Institut für Pathologie, Sektion Nephropathologie, Universitätsklinikum RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Deutschland.
- Medizinische Klinik II, Universitätsklinikum RWTH Aachen, Aachen, Deutschland.
| |
Collapse
|
123
|
Nap R, Aburidi M, Marcia R. Contrastive Pre-Training and Multiple Instance Learning for Predicting Tumor Microsatellite Instability. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-7. [PMID: 40039070 DOI: 10.1109/embc53108.2024.10782037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Accurate classification between tumor MicroSatellite Stability (MSS) and Instability (MSI) is crucial in gastrointestinal (GI) cancer prognosis and treatment. In this paper, we present a novel two-stage weakly supervised methodology, leveraging the synergy of Multiple Instance Learning (MIL) and a unique Contrastive Clustering Network (CCNet), aimed at enhancing MSI prediction in Whole Slide Image (WSI) analysis of GI cancers. In our framework, we utilize a contrastive learning-based feature extractor, coupled with MIL's efficient labeling. Our approach shows notable improvement in MSI classification, outperforming existing methods. Experiments using colorectal cancer and stomach adenocarcinoma datasets demonstrate the model's efficacy and generalizability, marking an advance in computational pathology and cancer diagnostics. Furthermore, we explored the efficacy of transfer learning using our model, examining the performance of pretrained feature extractors from ImageNet and STAD datasets. Our framework outperforms existing methods when pretrained on STAD and transferred to CRC data.Clinical relevance- The potential to enhance the accuracy of gastrointestinal cancer diagnosis and prognosis through advanced machine learning techniques. By leveraging transfer learning and weakly supervised frameworks clinicians can benefit from improved MSI prediction in histopathological images, aiding in personalized treatment strategies and patient outcomes.
Collapse
|
124
|
Shephard AJ, Bashir RMS, Mahmood H, Jahanifar M, Minhas F, Raza SEA, McCombe KD, Craig SG, James J, Brooks J, Nankivell P, Mehanna H, Khurram SA, Rajpoot NM. A fully automated and explainable algorithm for predicting malignant transformation in oral epithelial dysplasia. NPJ Precis Oncol 2024; 8:137. [PMID: 38942998 PMCID: PMC11213925 DOI: 10.1038/s41698-024-00624-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 05/29/2024] [Indexed: 06/30/2024] Open
Abstract
Oral epithelial dysplasia (OED) is a premalignant histopathological diagnosis given to lesions of the oral cavity. Its grading suffers from significant inter-/intra-observer variability, and does not reliably predict malignancy progression, potentially leading to suboptimal treatment decisions. To address this, we developed an artificial intelligence (AI) algorithm, that assigns an Oral Malignant Transformation (OMT) risk score based on the Haematoxylin and Eosin (H&E) stained whole slide images (WSIs). Our AI pipeline leverages an in-house segmentation model to detect and segment both nuclei and epithelium. Subsequently, a shallow neural network utilises interpretable morphological and spatial features, emulating histological markers, to predict progression. We conducted internal cross-validation on our development cohort (Sheffield; n = 193 cases) and independent validation on two external cohorts (Birmingham and Belfast; n = 89 cases). On external validation, the proposed OMTscore achieved an AUROC = 0.75 (Recall = 0.92) in predicting OED progression, outperforming other grading systems (Binary: AUROC = 0.72, Recall = 0.85). Survival analyses showed the prognostic value of our OMTscore (C-index = 0.60, p = 0.02), compared to WHO (C-index = 0.64, p = 0.003) and binary grades (C-index = 0.65, p < 0.001). Nuclear analyses elucidated the presence of peri-epithelial and intra-epithelial lymphocytes in highly predictive patches of transforming cases (p < 0.001). This is the first study to propose a completely automated, explainable, and externally validated algorithm for predicting OED transformation. Our algorithm shows comparable-to-human-level performance, offering a promising solution to the challenges of grading OED in routine clinical practice.
Collapse
Affiliation(s)
- Adam J Shephard
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | | | - Hanya Mahmood
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Mostafa Jahanifar
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Fayyaz Minhas
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Shan E Ahmed Raza
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Kris D McCombe
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Stephanie G Craig
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jacqueline James
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jill Brooks
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Paul Nankivell
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Syed Ali Khurram
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Nasir M Rajpoot
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK.
| |
Collapse
|
125
|
Darbandsari A, Farahani H, Asadi M, Wiens M, Cochrane D, Khajegili Mirabadi A, Jamieson A, Farnell D, Ahmadvand P, Douglas M, Leung S, Abolmaesumi P, Jones SJM, Talhouk A, Kommoss S, Gilks CB, Huntsman DG, Singh N, McAlpine JN, Bashashati A. AI-based histopathology image analysis reveals a distinct subset of endometrial cancers. Nat Commun 2024; 15:4973. [PMID: 38926357 PMCID: PMC11208496 DOI: 10.1038/s41467-024-49017-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Endometrial cancer (EC) has four molecular subtypes with strong prognostic value and therapeutic implications. The most common subtype (NSMP; No Specific Molecular Profile) is assigned after exclusion of the defining features of the other three molecular subtypes and includes patients with heterogeneous clinical outcomes. In this study, we employ artificial intelligence (AI)-powered histopathology image analysis to differentiate between p53abn and NSMP EC subtypes and consequently identify a sub-group of NSMP EC patients that has markedly inferior progression-free and disease-specific survival (termed 'p53abn-like NSMP'), in a discovery cohort of 368 patients and two independent validation cohorts of 290 and 614 from other centers. Shallow whole genome sequencing reveals a higher burden of copy number abnormalities in the 'p53abn-like NSMP' group compared to NSMP, suggesting that this group is biologically distinct compared to other NSMP ECs. Our work demonstrates the power of AI to detect prognostically different and otherwise unrecognizable subsets of EC where conventional and standard molecular or pathologic criteria fall short, refining image-based tumor classification. This study's findings are applicable exclusively to females.
Collapse
Affiliation(s)
- Amirali Darbandsari
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Hossein Farahani
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maryam Asadi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Wiens
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Dawn Cochrane
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | | | - Amy Jamieson
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Pouya Ahmadvand
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Maxwell Douglas
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Samuel Leung
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Purang Abolmaesumi
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Steven J M Jones
- Michael Smith Genome Sciences Center, British Columbia Cancer Research Center, Vancouver, BC, Canada
| | - Aline Talhouk
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - Naveena Singh
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver General Hospital, Vancouver, BC, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | - Ali Bashashati
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
126
|
Song J, Zhang J, Liu G, Guo Z, Liao H, Feng W, Lin W, Li L, Zhang Y, Yang Y, Liu B, Luo R, Chen H, Wang S, Liu JH. PET/CT deep learning prognosis for treatment decision support in esophageal squamous cell carcinoma. Insights Imaging 2024; 15:161. [PMID: 38913225 PMCID: PMC11196479 DOI: 10.1186/s13244-024-01737-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVES The clinical decision-making regarding choosing surgery alone (SA) or surgery followed by postoperative adjuvant chemotherapy (SPOCT) in esophageal squamous cell carcinoma (ESCC) remains controversial. We aim to propose a pre-therapy PET/CT image-based deep learning approach to improve the survival benefit and clinical management of ESCC patients. METHODS This retrospective multicenter study included 837 ESCC patients from three institutions. Prognostic biomarkers integrating six networks were developed to build an ESCC prognosis (ESCCPro) model and predict the survival probability of ESCC patients treated with SA and SPOCT. Patients who did not undergo surgical resection were in a control group. Overall survival (OS) was the primary end-point event. The expected improvement in survival prognosis with the application of ESCCPro to assign treatment protocols was estimated by comparing the survival of patients in each subgroup. Seven clinicians with varying experience evaluated how ESCCPro performed in assisting clinical decision-making. RESULTS In this retrospective multicenter study, patients receiving SA had a median OS 9.2 months longer than controls. No significant differences in survival were found between SA patients with predicted poor outcomes and the controls (p > 0.05). It was estimated that if ESCCPro was used to determine SA and SPOCT eligibility, the median OS in the ESCCPro-recommended SA group and SPOCT group would have been 15.3 months and 24.9 months longer, respectively. In addition, ESCCPro also significantly improved prognosis accuracy, certainty, and the efficiency of clinical experts. CONCLUSION ESCCPro assistance improved the survival benefit of ESCC patients and the clinical decision-making among the two treatment approaches. CRITICAL RELEVANCE STATEMENT The ESCCPro model for treatment decision-making is promising to improve overall survival in ESCC patients undergoing surgical resection and patients undergoing surgery followed by postoperative adjuvant chemotherapy. KEY POINTS ESCC is associated with a poor prognosis and unclear ideal treatments. ESCCPro predicts the survival of patients with ESCC and the expected benefit from SA. ESCCPro improves clinicians' stratification of patients' prognoses.
Collapse
Affiliation(s)
- Jiangdian Song
- School of Health Management, China Medical University, Shenyang, China.
| | - Jie Zhang
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Guichao Liu
- The Fifth Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
| | - Zhexu Guo
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors China Medical University, Ministry of Education, Shenyang, China
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, China
| | - Hongxian Liao
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Wenhui Feng
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Wenxiang Lin
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Lei Li
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yi Zhang
- Department of Medical Imaging, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Yuxiang Yang
- Department of Radiology, The Second People's Hospital of Xiangzhou, Zhuhai, China
| | - Bin Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, Hong Kong
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Siyun Wang
- Department of Oncology, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Jian-Hua Liu
- Department of Oncology, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
127
|
Pattilachan TM, Christodoulou M, Ross S. Diagnosis to dissection: AI's role in early detection and surgical intervention for gastric cancer. J Robot Surg 2024; 18:259. [PMID: 38900376 DOI: 10.1007/s11701-024-02005-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024]
Abstract
Gastric cancer remains a formidable health challenge worldwide; early detection and effective surgical intervention are critical for improving patient outcomes. This comprehensive review explores the evolving landscape of gastric cancer management, emphasizing the significant contributions of artificial intelligence (AI) in revolutionizing both diagnostic and therapeutic approaches. Despite advancements in the medical field, the subtle nature of early gastric cancer symptoms often leads to late-stage diagnoses, where survival rates are notably decreased. Historically, the treatment of gastric cancer has transitioned from palliative care to surgical resection, evolving further with the introduction of minimally invasive surgical (MIS) techniques. In the current era, AI has emerged as a transformative force, enhancing the precision of early gastric cancer detection through sophisticated image analysis, and supporting surgical decision-making with predictive modeling and real-time preop-, intraop-, and postoperative guidance. However, the deployment of AI in healthcare raises significant ethical, legal, and practical challenges, including the necessity for ongoing professional education and the development of standardized protocols to ensure patient safety and the effective use of AI technologies. Future directions point toward a synergistic integration of AI with clinical best practices, promising a new era of personalized, efficient, and safer gastric cancer management.
Collapse
Affiliation(s)
- Tara Menon Pattilachan
- AdventHealth Tampa, Surgery College of Medicine, Digestive Health Institute, University of Central Florida (UCF), 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Maria Christodoulou
- AdventHealth Tampa, Surgery College of Medicine, Digestive Health Institute, University of Central Florida (UCF), 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA
| | - Sharona Ross
- AdventHealth Tampa, Surgery College of Medicine, Digestive Health Institute, University of Central Florida (UCF), 3000 Medical Park Drive, Suite #500, Tampa, FL, 33613, USA.
| |
Collapse
|
128
|
Zeng Z, Zhu Q. Progress and prospects of biomarker-based targeted therapy and immune checkpoint inhibitors in advanced gastric cancer. Front Oncol 2024; 14:1382183. [PMID: 38947886 PMCID: PMC11211377 DOI: 10.3389/fonc.2024.1382183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Gastric cancer and gastroesophageal junction cancer represent the leading cause of tumor-related death worldwide. Although advances in immunotherapy and molecular targeted therapy have expanded treatment options, they have not significantly altered the prognosis for patients with unresectable or metastatic gastric cancer. A minority of patients, particularly those with PD-L1-positive, HER-2-positive, or MSI-high tumors, may benefit more from immune checkpoint inhibitors and/or HER-2-directed therapies in advanced stages. However, for those lacking specific targets and unique molecular features, conventional chemotherapy remains the only recommended effective and durable regimen. In this review, we summarize the roles of various signaling pathways and further investigate the available targets. Then, the current results of phase II/III clinical trials in advanced gastric cancer, along with the superiorities and limitations of the existing biomarkers, are specifically discussed. Finally, we will offer our insights in precision treatment pattern when encountering the substantial challenges.
Collapse
Affiliation(s)
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
129
|
Reitsam NG, Enke JS, Vu Trung K, Märkl B, Kather JN. Artificial Intelligence in Colorectal Cancer: From Patient Screening over Tailoring Treatment Decisions to Identification of Novel Biomarkers. Digestion 2024; 105:331-344. [PMID: 38865982 PMCID: PMC11457979 DOI: 10.1159/000539678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Artificial intelligence (AI) is increasingly entering and transforming not only medical research but also clinical practice. In the last 10 years, new AI methods have enabled computers to perform visual tasks, reaching high performance and thereby potentially supporting and even outperforming human experts. This is in particular relevant for colorectal cancer (CRC), which is the 3rd most common cancer type in general, as along the CRC patient journey many complex visual tasks need to be performed: from endoscopy over imaging to histopathology; the screening, diagnosis, and treatment of CRC involve visual image analysis tasks. SUMMARY In all these clinical areas, AI models have shown promising results by supporting physicians, improving accuracy, and providing new biological insights and biomarkers. By predicting prognostic and predictive biomarkers from routine images/slides, AI models could lead to an improved patient stratification for precision oncology approaches in the near future. Moreover, it is conceivable that AI models, in particular together with innovative techniques such as single-cell or spatial profiling, could help identify novel clinically as well as biologically meaningful biomarkers that could pave the way to new therapeutic approaches. KEY MESSAGES Here, we give a comprehensive overview of AI in colorectal cancer, describing and discussing these developments as well as the next steps which need to be taken to incorporate AI methods more broadly into the clinical care of CRC.
Collapse
Affiliation(s)
- Nic Gabriel Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany,
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany,
| | - Johanna Sophie Enke
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Kien Vu Trung
- Division of Gastroenterology, Medical Department II, University of Leipzig Medical Center, Leipzig, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
130
|
Rumberger JL, Greenwald NF, Ranek JS, Boonrat P, Walker C, Franzen J, Varra SR, Kong A, Sowers C, Liu CC, Averbukh I, Piyadasa H, Vanguri R, Nederlof I, Wang XJ, Van Valen D, Kok M, Hollmann TJ, Kainmueller D, Angelo M. Automated classification of cellular expression in multiplexed imaging data with Nimbus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597062. [PMID: 38895405 PMCID: PMC11185540 DOI: 10.1101/2024.06.02.597062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Multiplexed imaging offers a powerful approach to characterize the spatial topography of tissues in both health and disease. To analyze such data, the specific combination of markers that are present in each cell must be enumerated to enable accurate phenotyping, a process that often relies on unsupervised clustering. We constructed the Pan-Multiplex (Pan-M) dataset containing 197 million distinct annotations of marker expression across 15 different cell types. We used Pan-M to create Nimbus, a deep learning model to predict marker positivity from multiplexed image data. Nimbus is a pre-trained model that uses the underlying images to classify marker expression across distinct cell types, from different tissues, acquired using different microscope platforms, without requiring any retraining. We demonstrate that Nimbus predictions capture the underlying staining patterns of the full diversity of markers present in Pan-M. We then show how Nimbus predictions can be integrated with downstream clustering algorithms to robustly identify cell subtypes in image data. We have open-sourced Nimbus and Pan-M to enable community use at https://github.com/angelolab/Nimbus-Inference.
Collapse
Affiliation(s)
- J. Lorenz Rumberger
- Max-Delbruck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Humboldt-Universität zu Berlin, Faculty of Mathematics and Natural Sciences, Berlin, Germany
- Helmholtz Imaging
| | - Noah F. Greenwald
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Jolene S. Ranek
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Potchara Boonrat
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Cameron Walker
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Jannik Franzen
- Max-Delbruck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Helmholtz Imaging
- Charité University Medicine, Berlin, Germany
| | | | - Alex Kong
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Cameron Sowers
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Candace C. Liu
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Inna Averbukh
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Hadeesha Piyadasa
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Rami Vanguri
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Iris Nederlof
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Xuefei Julie Wang
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA, USA
| | - David Van Valen
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Travis J. Hollmann
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Dagmar Kainmueller
- Max-Delbruck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Helmholtz Imaging
- Potsdam University, Digital Engineering Faculty, Germany
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, California, USA
| |
Collapse
|
131
|
Kurz A, Müller H, Kather JN, Schneider L, Bucher TC, Brinker TJ. 3-Dimensional Reconstruction From Histopathological Sections: A Systematic Review. J Transl Med 2024; 104:102049. [PMID: 38513977 DOI: 10.1016/j.labinv.2024.102049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Although pathological tissue analysis is typically performed on single 2-dimensional (2D) histologic reference slides, 3-dimensional (3D) reconstruction from a sequence of histologic sections could provide novel opportunities for spatial analysis of the extracted tissue. In this review, we analyze recent works published after 2018 and report information on the extracted tissue types, the section thickness, and the number of sections used for reconstruction. By analyzing the technological requirements for 3D reconstruction, we observe that software tools exist, both free and commercial, which include the functionality to perform 3D reconstruction from a sequence of histologic images. Through the analysis of the most recent works, we provide an overview of the workflows and tools that are currently used for 3D reconstruction from histologic sections and address points for future work, such as a missing common file format or computer-aided analysis of the reconstructed model.
Collapse
Affiliation(s)
- Alexander Kurz
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heimo Müller
- Diagnostics and Research Institute for Pathology, Medical University of Graz, Graz, Austria
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea C Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
132
|
Perez-Lopez R, Ghaffari Laleh N, Mahmood F, Kather JN. A guide to artificial intelligence for cancer researchers. Nat Rev Cancer 2024; 24:427-441. [PMID: 38755439 DOI: 10.1038/s41568-024-00694-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Artificial intelligence (AI) has been commoditized. It has evolved from a specialty resource to a readily accessible tool for cancer researchers. AI-based tools can boost research productivity in daily workflows, but can also extract hidden information from existing data, thereby enabling new scientific discoveries. Building a basic literacy in these tools is useful for every cancer researcher. Researchers with a traditional biological science focus can use AI-based tools through off-the-shelf software, whereas those who are more computationally inclined can develop their own AI-based software pipelines. In this article, we provide a practical guide for non-computational cancer researchers to understand how AI-based tools can benefit them. We convey general principles of AI for applications in image analysis, natural language processing and drug discovery. In addition, we give examples of how non-computational researchers can get started on the journey to productively use AI in their own work.
Collapse
Affiliation(s)
- Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Narmin Ghaffari Laleh
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, 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
|
133
|
Xu Z, Li W, Dong X, Chen Y, Zhang D, Wang J, Zhou L, He G. Precision medicine in colorectal cancer: Leveraging multi-omics, spatial omics, and artificial intelligence. Clin Chim Acta 2024; 559:119686. [PMID: 38663471 DOI: 10.1016/j.cca.2024.119686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths. Recent advancements in genomic technologies and analytical approaches have revolutionized CRC research, enabling precision medicine. This review highlights the integration of multi-omics, spatial omics, and artificial intelligence (AI) in advancing precision medicine for CRC. Multi-omics approaches have uncovered molecular mechanisms driving CRC progression, while spatial omics have provided insights into the spatial heterogeneity of gene expression in CRC tissues. AI techniques have been utilized to analyze complex datasets, identify new treatment targets, and enhance diagnosis and prognosis. Despite the tumor's heterogeneity and genetic and epigenetic complexity, the fusion of multi-omics, spatial omics, and AI shows the potential to overcome these challenges and advance precision medicine in CRC. The future lies in integrating these technologies to provide deeper insights and enable personalized therapies for CRC patients.
Collapse
Affiliation(s)
- Zishan Xu
- Department of Pathology, Xinxiang Medical University, Xinxiang 453000, China
| | - Wei Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453000, China
| | - Xiangyang Dong
- Department of Pathology, Xinxiang Medical University, Xinxiang 453000, China
| | - Yingying Chen
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453000, China
| | - Dan Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang 453000, China
| | - Jingnan Wang
- Xinxiang Medical University SanQuan Medical College, Xinxiang 453003, China
| | - Lin Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Guoyang He
- Department of Pathology, Xinxiang Medical University, Xinxiang 453000, China.
| |
Collapse
|
134
|
Hilgers L, Ghaffari Laleh N, West NP, Westwood A, Hewitt KJ, Quirke P, Grabsch HI, Carrero ZI, Matthaei E, Loeffler CML, Brinker TJ, Yuan T, Brenner H, Brobeil A, Hoffmeister M, Kather JN. Automated curation of large-scale cancer histopathology image datasets using deep learning. Histopathology 2024; 84:1139-1153. [PMID: 38409878 DOI: 10.1111/his.15159] [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: 09/19/2023] [Revised: 12/29/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Artificial intelligence (AI) has numerous applications in pathology, supporting diagnosis and prognostication in cancer. However, most AI models are trained on highly selected data, typically one tissue slide per patient. In reality, especially for large surgical resection specimens, dozens of slides can be available for each patient. Manually sorting and labelling whole-slide images (WSIs) is a very time-consuming process, hindering the direct application of AI on the collected tissue samples from large cohorts. In this study we addressed this issue by developing a deep-learning (DL)-based method for automatic curation of large pathology datasets with several slides per patient. METHODS We collected multiple large multicentric datasets of colorectal cancer histopathological slides from the United Kingdom (FOXTROT, N = 21,384 slides; CR07, N = 7985 slides) and Germany (DACHS, N = 3606 slides). These datasets contained multiple types of tissue slides, including bowel resection specimens, endoscopic biopsies, lymph node resections, immunohistochemistry-stained slides, and tissue microarrays. We developed, trained, and tested a deep convolutional neural network model to predict the type of slide from the slide overview (thumbnail) image. The primary statistical endpoint was the macro-averaged area under the receiver operating curve (AUROCs) for detection of the type of slide. RESULTS In the primary dataset (FOXTROT), with an AUROC of 0.995 [95% confidence interval [CI]: 0.994-0.996] the algorithm achieved a high classification performance and was able to accurately predict the type of slide from the thumbnail image alone. In the two external test cohorts (CR07, DACHS) AUROCs of 0.982 [95% CI: 0.979-0.985] and 0.875 [95% CI: 0.864-0.887] were observed, which indicates the generalizability of the trained model on unseen datasets. With a confidence threshold of 0.95, the model reached an accuracy of 94.6% (7331 classified cases) in CR07 and 85.1% (2752 classified cases) for the DACHS cohort. CONCLUSION Our findings show that using the low-resolution thumbnail image is sufficient to accurately classify the type of slide in digital pathology. This can support researchers to make the vast resource of existing pathology archives accessible to modern AI models with only minimal manual annotations.
Collapse
Affiliation(s)
- Lars Hilgers
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Alice Westwood
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Katherine J Hewitt
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heike I Grabsch
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Emylou Matthaei
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
135
|
Duwe G, Mercier D, Wiesmann C, Kauth V, Moench K, Junker M, Neumann CCM, Haferkamp A, Dengel A, Höfner T. Challenges and perspectives in use of artificial intelligence to support treatment recommendations in clinical oncology. Cancer Med 2024; 13:e7398. [PMID: 38923826 PMCID: PMC11196383 DOI: 10.1002/cam4.7398] [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: 01/24/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Artificial intelligence (AI) promises to be the next revolutionary step in modern society. Yet, its role in all fields of industry and science need to be determined. One very promising field is represented by AI-based decision-making tools in clinical oncology leading to more comprehensive, personalized therapy approaches. In this review, the authors provide an overview on all relevant technical applications of AI in oncology, which are required to understand the future challenges and realistic perspectives for decision-making tools. In recent years, various applications of AI in medicine have been developed focusing on the analysis of radiological and pathological images. AI applications encompass large amounts of complex data supporting clinical decision-making and reducing errors by objectively quantifying all aspects of the data collected. In clinical oncology, almost all patients receive a treatment recommendation in a multidisciplinary cancer conference at the beginning and during their treatment periods. These highly complex decisions are based on a large amount of information (of the patients and of the various treatment options), which need to be analyzed and correctly classified in a short time. In this review, the authors describe the technical and medical requirements of AI to address these scientific challenges in a multidisciplinary manner. Major challenges in the use of AI in oncology and decision-making tools are data security, data representation, and explainability of AI-based outcome predictions, in particular for decision-making processes in multidisciplinary cancer conferences. Finally, limitations and potential solutions are described and compared for current and future research attempts.
Collapse
Affiliation(s)
- Gregor Duwe
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Dominique Mercier
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Crispin Wiesmann
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Verena Kauth
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Kerstin Moench
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Markus Junker
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Christopher C. M. Neumann
- Department of Hematology, Oncology and Tumor ImmunologyCharité‐Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt‐Universität zu BerlinBerlinGermany
| | - Axel Haferkamp
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Andreas Dengel
- Research Unit Smart Data and Knowledge ServicesGerman Research Center for Artificial IntelligenceKaiserslauternGermany
| | - Thomas Höfner
- Department of Urology and Pediatric UrologyUniversity Medical Center, Johannes Gutenberg UniversityMainzGermany
- Department of Urology, Ordensklinikum Linz ElisabethinenLinzAustria
| |
Collapse
|
136
|
Gustav M, Reitsam NG, Carrero ZI, Loeffler CML, van Treeck M, Yuan T, West NP, Quirke P, Brinker TJ, Brenner H, Favre L, Märkl B, Stenzinger A, Brobeil A, Hoffmeister M, Calderaro J, Pujals A, Kather JN. Deep learning for dual detection of microsatellite instability and POLE mutations in colorectal cancer histopathology. NPJ Precis Oncol 2024; 8:115. [PMID: 38783059 PMCID: PMC11116442 DOI: 10.1038/s41698-024-00592-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/14/2024] [Indexed: 05/25/2024] Open
Abstract
In the spectrum of colorectal tumors, microsatellite-stable (MSS) tumors with DNA polymerase ε (POLE) mutations exhibit a hypermutated profile, holding the potential to respond to immunotherapy similarly to their microsatellite-instable (MSI) counterparts. Yet, due to their rarity and the associated testing costs, systematic screening for these mutations is not commonly pursued. Notably, the histopathological phenotype resulting from POLE mutations is theorized to resemble that of MSI. This resemblance not only could facilitate their detection by a transformer-based Deep Learning (DL) system trained on MSI pathology slides, but also indicates the possibility for MSS patients with POLE mutations to access enhanced treatment options, which might otherwise be overlooked. To harness this potential, we trained a Deep Learning classifier on a large dataset with the ground truth for microsatellite status and subsequently validated its capabilities for MSI and POLE detection across three external cohorts. Our model accurately identified MSI status in both the internal and external resection cohorts using pathology images alone. Notably, with a classification threshold of 0.5, over 75% of POLE driver mutant patients in the external resection cohorts were flagged as "positive" by a DL system trained on MSI status. In a clinical setting, deploying this DL model as a preliminary screening tool could facilitate the efficient identification of clinically relevant MSI and POLE mutations in colorectal tumors, in one go.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Titus J Brinker
- Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Loëtitia Favre
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Anaïs Pujals
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
137
|
Ahn B, Moon D, Kim HS, Lee C, Cho NH, Choi HK, Kim D, Lee JY, Nam EJ, Won D, An HJ, Kwon SY, Shin SJ, Jung HR, Kwon D, Park H, Kim M, Cha YJ, Park H, Lee Y, Noh S, Lee YM, Choi SE, Kim JM, Sung SH, Park E. Histopathologic image-based deep learning classifier for predicting platinum-based treatment responses in high-grade serous ovarian cancer. Nat Commun 2024; 15:4253. [PMID: 38762636 PMCID: PMC11102549 DOI: 10.1038/s41467-024-48667-6] [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: 07/20/2023] [Accepted: 05/09/2024] [Indexed: 05/20/2024] Open
Abstract
Platinum-based chemotherapy is the cornerstone treatment for female high-grade serous ovarian carcinoma (HGSOC), but choosing an appropriate treatment for patients hinges on their responsiveness to it. Currently, no available biomarkers can promptly predict responses to platinum-based treatment. Therefore, we developed the Pathologic Risk Classifier for HGSOC (PathoRiCH), a histopathologic image-based classifier. PathoRiCH was trained on an in-house cohort (n = 394) and validated on two independent external cohorts (n = 284 and n = 136). The PathoRiCH-predicted favorable and poor response groups show significantly different platinum-free intervals in all three cohorts. Combining PathoRiCH with molecular biomarkers provides an even more powerful tool for the risk stratification of patients. The decisions of PathoRiCH are explained through visualization and a transcriptomic analysis, which bolster the reliability of our model's decisions. PathoRiCH exhibits better predictive performance than current molecular biomarkers. PathoRiCH will provide a solid foundation for developing an innovative tool to transform the current diagnostic pipeline for HGSOC.
Collapse
Affiliation(s)
- Byungsoo Ahn
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Damin Moon
- Artificial Intelligence Research Center, JLK Inc., Seoul, South Korea
| | - Hyun-Soo Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Chung Lee
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Nam Hoon Cho
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Heung-Kook Choi
- Artificial Intelligence Research Center, JLK Inc., Seoul, South Korea
| | - Dongmin Kim
- Artificial Intelligence Research Center, JLK Inc., Seoul, South Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ji Nam
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Dongju Won
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Jung An
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Sun Young Kwon
- Department of Pathology, Keimyung University School of Medicine, Daegu, South Korea
| | - Su-Jin Shin
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Ra Jung
- Department of Pathology, Keimyung University School of Medicine, Daegu, South Korea
| | - Dohee Kwon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Heejung Park
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Milim Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyunjin Park
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yangkyu Lee
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Songmi Noh
- Department of Diagnostic Pathology, Gangnam CHA Medical Center, CHA University College of Medicine, Seoul, South Korea
| | - Yong-Moon Lee
- Department of Pathology, Dankook University School of Medicine, Cheonan, South Korea
| | - Sung-Eun Choi
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Ji Min Kim
- Department of Pathology, Ewha Womans University, Seoul, South Korea
| | - Sun Hee Sung
- Department of Pathology, Ewha Womans University, Seoul, South Korea
| | - Eunhyang Park
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
138
|
Han Z, Zhang Z, Yang X, Li Z, Sang S, Islam MT, Guo AA, Li Z, Wang X, Wang J, Zhang T, Sun Z, Yu L, Wang W, Xiong W, Li G, Jiang Y. Development and interpretation of a pathomics-driven ensemble model for predicting the response to immunotherapy in gastric cancer. J Immunother Cancer 2024; 12:e008927. [PMID: 38749538 PMCID: PMC11097892 DOI: 10.1136/jitc-2024-008927] [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] [Accepted: 04/24/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Only a subset of patients with gastric cancer experience long-term benefits from immune checkpoint inhibitors (ICIs). Currently, there is a deficiency in precise predictive biomarkers for ICI efficacy. The aim of this study was to develop and validate a pathomics-driven ensemble model for predicting the response to ICIs in gastric cancer, using H&E-stained whole slide images (WSI). METHODS This multicenter study retrospectively collected and analyzed H&E-stained WSIs and clinical data from 584 patients with gastric cancer. An ensemble model, integrating four classifiers: least absolute shrinkage and selection operator, k-nearest neighbors, decision trees, and random forests, was developed and validated using pathomics features, with the objective of predicting the therapeutic efficacy of immune checkpoint inhibition. Model performance was evaluated using metrics including the area under the curve (AUC), sensitivity, and specificity. Additionally, SHAP (SHapley Additive exPlanations) analysis was used to explain the model's predicted values as the sum of the attribution values for each input feature. Pathogenomics analysis was employed to explain the molecular mechanisms underlying the model's predictions. RESULTS Our pathomics-driven ensemble model effectively stratified the response to ICIs in training cohort (AUC 0.985 (95% CI 0.971 to 0.999)), which was further validated in internal validation cohort (AUC 0.921 (95% CI 0.839 to 0.999)), as well as in external validation cohort 1 (AUC 0.914 (95% CI 0.837 to 0.990)), and external validation cohort 2 (0.927 (95% CI 0.802 to 0.999)). The univariate Cox regression analysis revealed that the prediction signature of pathomics-driven ensemble model was a prognostic factor for progression-free survival in patients with gastric cancer who underwent immunotherapy (p<0.001, HR 0.35 (95% CI 0.24 to 0.50)), and remained an independent predictor after multivariable Cox regression adjusted for clinicopathological variables, (including sex, age, carcinoembryonic antigen, carbohydrate antigen 19-9, therapy regime, line of therapy, differentiation, location and programmed death ligand 1 (PD-L1) expression in all patients (p<0.001, HR 0.34 (95% CI 0.24 to 0.50)). Pathogenomics analysis suggested that the ensemble model is driven by molecular-level immune, cancer, metabolism-related pathways, and was correlated with the immune-related characteristics, including immune score, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data score, and tumor purity. CONCLUSIONS Our pathomics-driven ensemble model exhibited high accuracy and robustness in predicting the response to ICIs using WSIs. Therefore, it could serve as a novel and valuable tool to facilitate precision immunotherapy.
Collapse
Affiliation(s)
- Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Zhicheng Zhang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
- JancsiLab, JancsiTech, Hongkong, China
| | - Xianqi Yang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhe Li
- School of Computer Science and Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Shengtian Sang
- Department of Radiology, Stanford University School of Medicine, Stanford, California, USA
| | - Md Tauhidul Islam
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Alyssa A Guo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Zihan Li
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Xiaoyan Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
| | - Lequan Yu
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wei Wang
- Department of Gastric Surgery, and State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wenjun Xiong
- Department of Gastrointestinal Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine,Southern Medical University, Guangzhou, Guangdong, China
- School of Clinical Medicine, Tsinghua University, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
139
|
Plekhanov AA, Kozlov DS, Shepeleva AA, Kiseleva EB, Shimolina LE, Druzhkova IN, Plekhanova MA, Karabut MM, Gubarkova EV, Gavrina AI, Krylov DP, Sovetsky AA, Gamayunov SV, Kuznetsova DS, Zaitsev VY, Sirotkina MA, Gladkova ND. Tissue Elasticity as a Diagnostic Marker of Molecular Mutations in Morphologically Heterogeneous Colorectal Cancer. Int J Mol Sci 2024; 25:5337. [PMID: 38791375 PMCID: PMC11120711 DOI: 10.3390/ijms25105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
The presence of molecular mutations in colorectal cancer (CRC) is a decisive factor in selecting the most effective first-line therapy. However, molecular analysis is routinely performed only in a limited number of patients with remote metastases. We propose to use tissue stiffness as a marker of the presence of molecular mutations in CRC samples. For this purpose, we applied compression optical coherence elastography (C-OCE) to calculate stiffness values in regions corresponding to specific CRC morphological patterns (n = 54). In parallel to estimating stiffness, molecular analysis from the same zones was performed to establish their relationships. As a result, a high correlation between the presence of KRAS/NRAS/BRAF driver mutations and high stiffness values was revealed regardless of CRC morphological pattern type. Further, we proposed threshold stiffness values for label-free targeted detection of molecular alterations in CRC tissues: for KRAS, NRAS, or BRAF driver mutation-above 803 kPa (sensitivity-91%; specificity-80%; diagnostic accuracy-85%), and only for KRAS driver mutation-above 850 kPa (sensitivity-90%; specificity-88%; diagnostic accuracy-89%). To conclude, C-OCE estimation of tissue stiffness can be used as a clinical diagnostic tool for preliminary screening of genetic burden in CRC tissues.
Collapse
Affiliation(s)
- Anton A. Plekhanov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Dmitry S. Kozlov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Anastasia A. Shepeleva
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
| | - Elena B. Kiseleva
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Liubov E. Shimolina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Irina N. Druzhkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Maria A. Plekhanova
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
- Nizhny Novgorod City Polyclinic #1, 5 Marshala Zhukova Sq., 603107 Nizhny Novgorod, Russia
| | - Maria M. Karabut
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Ekaterina V. Gubarkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Alena I. Gavrina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Dmitry P. Krylov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Alexander A. Sovetsky
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanova St., 603950 Nizhny Novgorod, Russia
| | - Sergey V. Gamayunov
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
| | - Daria S. Kuznetsova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Vladimir Y. Zaitsev
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanova St., 603950 Nizhny Novgorod, Russia
| | - Marina A. Sirotkina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Natalia D. Gladkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| |
Collapse
|
140
|
Ren Z, Yang K, Zhu L, Yin D, Zhou Y. Regulatory T cells as crucial trigger and potential target for hyperprogressive disease subsequent to PD-1/PD-L1 blockade for cancer treatment. Int Immunopharmacol 2024; 132:111934. [PMID: 38574701 DOI: 10.1016/j.intimp.2024.111934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
PD-1/PD-L1 blockade therapy has brought great success to cancer treatment. Nevertheless, limited beneficiary populations and even hyperprogressive disease (HPD) greatly constrain the application of PD-1/PD-L1 inhibitors in clinical treatment. HPD is a special pattern of disease progression with rapid tumor growth and even serious consequences of patient death, which requires urgent attention. Among the many predisposing causes of HPD, regulatory T cells (Tregs) are suspected because they are amplified in cases of HPD. Tregs express PD-1 thus PD-1/PD-L1 blockade therapy may have an impact on Tregs which leads to HPD. Tregs are a subset of CD4+ T cells expressing FoxP3 and play critical roles in suppressing immunity. Tregs migrate toward tumors in the presence of chemokines to suppress antitumor immune responses, causing cancer cells to grow and proliferate. Studies have shown that deleting Tregs could enhance the efficacy of PD-1/PD-L1 blockade therapy and reduce the occurrence of HPD. This suggests that immunotherapy combined with Treg depletion may be an effective means of avoiding HPD. In this review, we summarized the immunosuppressive-related functions of Tregs in antitumor therapy and focused on advances in therapy combining Tregs depletion with PD-1/PD-L1 blockade in clinical studies. Moreover, we provided an outlook on Treg-targeted HPD early warning for PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Zhe Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Kaiqing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Detao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
141
|
Umemoto M, Mariya T, Nambu Y, Nagata M, Horimai T, Sugita S, Kanaseki T, Takenaka Y, Shinkai S, Matsuura M, Iwasaki M, Hirohashi Y, Hasegawa T, Torigoe T, Fujino Y, Saito T. Prediction of Mismatch Repair Status in Endometrial Cancer from Histological Slide Images Using Various Deep Learning-Based Algorithms. Cancers (Basel) 2024; 16:1810. [PMID: 38791889 PMCID: PMC11119770 DOI: 10.3390/cancers16101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The application of deep learning algorithms to predict the molecular profiles of various cancers from digital images of hematoxylin and eosin (H&E)-stained slides has been reported in recent years, mainly for gastric and colon cancers. In this study, we investigated the potential use of H&E-stained endometrial cancer slide images to predict the associated mismatch repair (MMR) status. H&E-stained slide images were collected from 127 cases of the primary lesion of endometrial cancer. After digitization using a Nanozoomer virtual slide scanner (Hamamatsu Photonics), we segmented the scanned images into 5397 tiles of 512 × 512 pixels. The MMR proteins (PMS2, MSH6) were immunohistochemically stained, classified into MMR proficient/deficient, and annotated for each case and tile. We trained several neural networks, including convolutional and attention-based networks, using tiles annotated with the MMR status. Among the tested networks, ResNet50 exhibited the highest area under the receiver operating characteristic curve (AUROC) of 0.91 for predicting the MMR status. The constructed prediction algorithm may be applicable to other molecular profiles and useful for pre-screening before implementing other, more costly genetic profiling tests.
Collapse
Affiliation(s)
- Mina Umemoto
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Tasuku Mariya
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Yuta Nambu
- Department of Media Architecture, Future University Hakodate, Hakodate 041-8655, Japan; (Y.N.); (M.N.); (Y.F.)
| | - Mai Nagata
- Department of Media Architecture, Future University Hakodate, Hakodate 041-8655, Japan; (Y.N.); (M.N.); (Y.F.)
| | | | - Shintaro Sugita
- Department of Surgical Pathology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (S.S.); (T.H.)
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (T.K.); (Y.H.); (T.T.)
| | - Yuka Takenaka
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Shota Shinkai
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Motoki Matsuura
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Masahiro Iwasaki
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (T.K.); (Y.H.); (T.T.)
| | - Tadashi Hasegawa
- Department of Surgical Pathology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (S.S.); (T.H.)
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (T.K.); (Y.H.); (T.T.)
| | - Yuichi Fujino
- Department of Media Architecture, Future University Hakodate, Hakodate 041-8655, Japan; (Y.N.); (M.N.); (Y.F.)
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University of Medicine, Sapporo 060-8556, Japan; (M.U.); (Y.T.); (S.S.); (M.M.); (M.I.); (T.S.)
| |
Collapse
|
142
|
Zhang X, Zhao Z, Wang R, Chen H, Zheng X, Liu L, Lan L, Li P, Wu S, Cao Q, Luo R, Hu W, Lyu S, Zhang Z, Xie D, Ye Y, Wang Y, Cai M. A multicenter proof-of-concept study on deep learning-based intraoperative discrimination of primary central nervous system lymphoma. Nat Commun 2024; 15:3768. [PMID: 38704409 PMCID: PMC11069536 DOI: 10.1038/s41467-024-48171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
Accurate intraoperative differentiation of primary central nervous system lymphoma (PCNSL) remains pivotal in guiding neurosurgical decisions. However, distinguishing PCNSL from other lesions, notably glioma, through frozen sections challenges pathologists. Here we sought to develop and validate a deep learning model capable of precisely distinguishing PCNSL from non-PCNSL lesions, especially glioma, using hematoxylin and eosin (H&E)-stained frozen whole-slide images. Also, we compared its performance against pathologists of varying expertise. Additionally, a human-machine fusion approach integrated both model and pathologic diagnostics. In external cohorts, LGNet achieved AUROCs of 0.965 and 0.972 in distinguishing PCNSL from glioma and AUROCs of 0.981 and 0.993 in differentiating PCNSL from non-PCNSL lesions. Outperforming several pathologists, LGNet significantly improved diagnostic performance, further augmented to some extent by fusion approach. LGNet's proficiency in frozen section analysis and its synergy with pathologists indicate its valuable role in intraoperative diagnosis, particularly in discriminating PCNSL from glioma, alongside other lesions.
Collapse
Affiliation(s)
- Xinke Zhang
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zihan Zhao
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ruixuan Wang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haohua Chen
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xueyi Zheng
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lili Liu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lilong Lan
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Peng Li
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shuyang Wu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qinghua Cao
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Rongzhen Luo
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shanshan Lyu
- Department of Pathology, Guangdong Provincial People's Hospital, Guangzhou, 510080, China
| | - Zhengyu Zhang
- Department of Pathology, Nanfang Hospital, Soutern Medical University, Guangzhou, 510515, China
| | - Dan Xie
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Soutern Medical University, Guangzhou, 510515, China.
| | - Yu Wang
- Department of Pathology, Zhujiang Hospital, Soutern Medical University, Guangzhou, 510280, China.
| | - Muyan Cai
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
143
|
Li D, Zhang J, Guo W, Ma K, Qin Z, Zhang J, Chen L, Xiong L, Huang J, Wan C, Huang P. A diagnostic strategy for pulmonary fat embolism based on routine H&E staining using computational pathology. Int J Legal Med 2024; 138:849-858. [PMID: 37999766 DOI: 10.1007/s00414-023-03136-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023]
Abstract
Pulmonary fat embolism (PFE) as a cause of death often occurs in trauma cases such as fractures and soft tissue contusions. Traditional PFE diagnosis relies on subjective methods and special stains like oil red O. This study utilizes computational pathology, combining digital pathology and deep learning algorithms, to precisely quantify fat emboli in whole slide images using conventional hematoxylin-eosin (H&E) staining. The results demonstrate deep learning's ability to identify fat droplet morphology in lung microvessels, achieving an area under the receiver operating characteristic (ROC) curve (AUC) of 0.98. The AI-quantified fat globules generally matched the Falzi scoring system with oil red O staining. The relative quantity of fat emboli against lung area was calculated by the algorithm, determining a diagnostic threshold of 8.275% for fatal PFE. A diagnostic strategy based on this threshold achieved a high AUC of 0.984, similar to manual identification with special stains but surpassing H&E staining. This demonstrates computational pathology's potential as an affordable, rapid, and precise method for fatal PFE diagnosis in forensic practice.
Collapse
Affiliation(s)
- Dechan Li
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Ji Zhang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Wenqing Guo
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
- Department of Forensic Pathology, Shanxi Medical University, Taiyuan, China
| | - Kaijun Ma
- Shanghai Key Laboratory of Crime Scene Evidence, Institute of Criminal Science and Technology, Shanghai Municipal Public Security Bureau, Shanghai, China
| | - Zhiqiang Qin
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Jianhua Zhang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Liqin Chen
- Department of Forensic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Ling Xiong
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Jiang Huang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China.
| | - Changwu Wan
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China.
| | - Ping Huang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China.
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China.
| |
Collapse
|
144
|
Kazemi A, Rasouli-Saravani A, Gharib M, Albuquerque T, Eslami S, Schüffler PJ. A systematic review of machine learning-based tumor-infiltrating lymphocytes analysis in colorectal cancer: Overview of techniques, performance metrics, and clinical outcomes. Comput Biol Med 2024; 173:108306. [PMID: 38554659 DOI: 10.1016/j.compbiomed.2024.108306] [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: 10/14/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
The incidence of colorectal cancer (CRC), one of the deadliest cancers around the world, is increasing. Tissue microenvironment (TME) features such as tumor-infiltrating lymphocytes (TILs) can have a crucial impact on diagnosis or decision-making for treating patients with CRC. While clinical studies showed that TILs improve the host immune response, leading to a better prognosis, inter-observer agreement for quantifying TILs is not perfect. Incorporating machine learning (ML) based applications in clinical routine may promote diagnosis reliability. Recently, ML has shown potential for making progress in routine clinical procedures. We aim to systematically review the TILs analysis based on ML in CRC histological images. Deep learning (DL) and non-DL techniques can aid pathologists in identifying TILs, and automated TILs are associated with patient outcomes. However, a large multi-institutional CRC dataset with a diverse and multi-ethnic population is necessary to generalize ML methods.
Collapse
Affiliation(s)
- Azar Kazemi
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany.
| | - Ashkan Rasouli-Saravani
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Gharib
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Saeid Eslami
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Informatics, University of Amsterdam, Amsterdam, the Netherlands.
| | - Peter J Schüffler
- Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany; TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; Munich Center for Machine Learning, Munich, Germany; Munich Data Science Institute, Munich, Germany.
| |
Collapse
|
145
|
Eisenstein M. AI assistance for planning cancer treatment. Nature 2024; 629:S14-S16. [PMID: 38811703 DOI: 10.1038/d41586-024-01431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
|
146
|
Yilmaz F, Brickman A, Najdawi F, Yakirevich E, Egger R, Resnick MB. Advancing Artificial Intelligence Integration Into the Pathology Workflow: Exploring Opportunities in Gastrointestinal Tract Biopsies. J Transl Med 2024; 104:102043. [PMID: 38431118 DOI: 10.1016/j.labinv.2024.102043] [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: 10/30/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
This review aims to present a comprehensive overview of the current landscape of artificial intelligence (AI) applications in the analysis of tubular gastrointestinal biopsies. These publications cover a spectrum of conditions, ranging from inflammatory ailments to malignancies. Moving beyond the conventional diagnosis based on hematoxylin and eosin-stained whole-slide images, the review explores additional implications of AI, including its involvement in interpreting immunohistochemical results, molecular subtyping, and the identification of cellular spatial biomarkers. Furthermore, the review examines how AI can contribute to enhancing the quality and control of diagnostic processes, introducing new workflow options, and addressing the limitations and caveats associated with current AI platforms in this context.
Collapse
Affiliation(s)
- Fazilet Yilmaz
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Arlen Brickman
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Fedaa Najdawi
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Evgeny Yakirevich
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | | | - Murray B Resnick
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island.
| |
Collapse
|
147
|
Schüffler P, Steiger K, Mogler C. [Artificial intelligence for pathology-how, where, and why?]. PATHOLOGIE (HEIDELBERG, GERMANY) 2024; 45:198-202. [PMID: 38472382 PMCID: PMC11045628 DOI: 10.1007/s00292-024-01314-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Artificial intelligence promises many innovations and simplifications in pathology, but also raises just as many questions and uncertainties. In this article, we provide a brief overview of the current status, the goals already achieved by existing algorithms, and the remaining challenges.
Collapse
Affiliation(s)
- Peter Schüffler
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland.
- TUM School of Computation, Information and Technology, Technische Universität München, München, Deutschland.
- Munich Center for Machine Learning (MCML), München, Deutschland.
| | - Katja Steiger
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland
| | - Carolin Mogler
- Institut für Pathologie, TUM School of Medicine and Health, Technische Universität München, München, Deutschland
| |
Collapse
|
148
|
Lotter W, Hassett MJ, Schultz N, Kehl KL, Van Allen EM, Cerami E. Artificial Intelligence in Oncology: Current Landscape, Challenges, and Future Directions. Cancer Discov 2024; 14:711-726. [PMID: 38597966 PMCID: PMC11131133 DOI: 10.1158/2159-8290.cd-23-1199] [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: 10/12/2023] [Revised: 01/29/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Artificial intelligence (AI) in oncology is advancing beyond algorithm development to integration into clinical practice. This review describes the current state of the field, with a specific focus on clinical integration. AI applications are structured according to cancer type and clinical domain, focusing on the four most common cancers and tasks of detection, diagnosis, and treatment. These applications encompass various data modalities, including imaging, genomics, and medical records. We conclude with a summary of existing challenges, evolving solutions, and potential future directions for the field. SIGNIFICANCE AI is increasingly being applied to all aspects of oncology, where several applications are maturing beyond research and development to direct clinical integration. This review summarizes the current state of the field through the lens of clinical translation along the clinical care continuum. Emerging areas are also highlighted, along with common challenges, evolving solutions, and potential future directions for the field.
Collapse
Affiliation(s)
- William Lotter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael J. Hassett
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth L. Kehl
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M. Van Allen
- Harvard Medical School, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ethan Cerami
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
149
|
Zhao Y, Wang W, Ji Y, Guo Y, Duan J, Liu X, Yan D, Liang D, Li W, Zhang Z, Li ZC. Computational Pathology for Prediction of Isocitrate Dehydrogenase Gene Mutation from Whole Slide Images in Adult Patients with Diffuse Glioma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:747-758. [PMID: 38325551 DOI: 10.1016/j.ajpath.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
Isocitrate dehydrogenase gene (IDH) mutation is one of the most important molecular markers of glioma. Accurate detection of IDH status is a crucial step for integrated diagnosis of adult-type diffuse gliomas. Herein, a clustering-based hybrid of a convolutional neural network and a vision transformer deep learning model was developed to detect IDH mutation status from annotation-free hematoxylin and eosin-stained whole slide pathologic images of 2275 adult patients with diffuse gliomas. For comparison, a pure convolutional neural network, a pure vision transformer, and a classic multiple-instance learning model were also assessed. The hybrid model achieved an area under the receiver operating characteristic curve of 0.973 in the validation set and 0.953 in the external test set, outperforming the other models. The hybrid model's ability in IDH detection between difficult subgroups with different IDH status but shared histologic features, achieving areas under the receiver operating characteristic curve ranging from 0.850 to 0.985 in validation and test sets. These data suggest that the proposed hybrid model has a potential to be used as a computational pathology tool for preliminary rapid detection of IDH mutation from whole slide images in adult patients with diffuse gliomas.
Collapse
Affiliation(s)
- Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuchen Ji
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial Hospital, Zhengzhou, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dong Liang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China; National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China; National Innovation Center for Advanced Medical Devices, Shenzhen, China.
| |
Collapse
|
150
|
Liu X, Hu W, Diao S, Abera DE, Racoceanu D, Qin W. Multi-scale feature fusion for prediction of IDH1 mutations in glioma histopathological images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 248:108116. [PMID: 38518408 DOI: 10.1016/j.cmpb.2024.108116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/30/2024] [Accepted: 03/02/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND AND OBJECTIVE Mutations in isocitrate dehydrogenase 1 (IDH1) play a crucial role in the prognosis, diagnosis, and treatment of gliomas. However, current methods for determining its mutation status, such as immunohistochemistry and gene sequencing, are difficult to implement widely in routine clinical diagnosis. Recent studies have shown that using deep learning methods based on pathological images of glioma can predict the mutation status of the IDH1 gene. However, our research focuses on utilizing multi-scale information in pathological images to improve the accuracy of predicting IDH1 gene mutations, thereby providing an accurate and cost-effective prediction method for routine clinical diagnosis. METHODS In this paper, we propose a multi-scale fusion gene identification network (MultiGeneNet). The network first uses two feature extractors to obtain feature maps at different scale images, and then by employing a bilinear pooling layer based on Hadamard product to realize the fusion of multi-scale features. Through fully exploiting the complementarity among features at different scales, we are able to obtain a more comprehensive and rich representation of multi-scale features. RESULTS Based on the Hematoxylin and Eosin stained pathological section dataset of 296 patients, our method achieved an accuracy of 83.575 % and an AUC of 0.886, thus significantly outperforming other single-scale methods. CONCLUSIONS Our method can be deployed in medical aid systems at very low cost, serving as a diagnostic or prognostic tool for glioma patients in medically underserved areas.
Collapse
Affiliation(s)
- Xiang Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wanming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, China
| | - Songhui Diao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Deboch Eyob Abera
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Daniel Racoceanu
- Sorbonne University, Inria, CNRS, Inserm, AP-HP, Inria, Paris Brain Institute - ICM, F-75013 Paris, France
| | - Wenjian Qin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|