1
|
Zhou J, Yang K, Lu M, Fu P, Chen Y, Chen L. Higher density of compact B cell clusters in invasive front may contribute to better prognosis in pancreatic ductal adenocarcinoma. Discov Oncol 2025; 16:555. [PMID: 40246809 PMCID: PMC12006623 DOI: 10.1007/s12672-025-02260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
While the correlation between T cells and patient survival was widely investigated, the clinical significance of CD20+ B cells in pancreatic ductal adenocarcinoma (PDAC) is less clear. We hypothesized that the spatial pattern of B cells within tumor microenvironment (TME) are more informative, which may reveal the prognostic significance for PDAC patients. Therefore, we developed a computer-based workflow to analyze CD20+ B cells in whole slide images (WSI) from 45 cases of PDAC patients. Depending on this workflow, annotations of each case which were created by pathologists were subdivided for three regions, including invasive front (IF), cancer center (CT) and cancer island (CI) to explore the association between the spatial pattern of CD20+ B cells and patient prognosis outcomes. After that, occupancy rate (as area under curve, occupancy AUC), fractal dimension differences (ΔFD), cluster density and coverage ratio were used to quantify the spatial pattern of B cells in TME. We observed B cells were distributed across different regions, manifesting in both clustered and dispersed patterns. Compared to features of B cells spatial distribution in CT region, B cells in IF region exhibited higher occupancy AUC (p = 0.00004), cluster density (p = 0.000002) and coverage ratio (p = 0.000884). Patients with longer survivals had smaller ΔFD (p = 0.05), higher B-cell cluster density (p = 0.003) and lower coverage ratio (p = 0.02) in IF region. Our study indicated the spatial distribution of B cells in IF and CT was different and the higher density of compact B-cell clusters in IF region may be associated with better prognosis in PDAC.
Collapse
Affiliation(s)
- Junwen Zhou
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hung Hom, HK, China
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Kunping Yang
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, China
| | - Mei Lu
- Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, Fujian, China
| | - Peiling Fu
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Yupeng Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China.
| |
Collapse
|
2
|
Cui X, Song J, Li Q, Ren J. Identification of biomarkers and target drugs for melanoma: a topological and deep learning approach. Front Genet 2025; 16:1471037. [PMID: 40098976 PMCID: PMC11911340 DOI: 10.3389/fgene.2025.1471037] [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: 07/26/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Melanoma, a highly aggressive malignancy characterized by rapid metastasis and elevated mortality rates, predominantly originates in cutaneous tissues. While surgical interventions, immunotherapy, and targeted therapies have advanced, the prognosis for advanced-stage melanoma remains dismal. Globally, melanoma incidence continues to rise, with the United States alone reporting over 100,000 new cases and 7,000 deaths annually. Despite the exponential growth of tumor data facilitated by next-generation sequencing (NGS), current analytical approaches predominantly emphasize single-gene analyses, neglecting critical insights into complex gene interaction networks. This study aims to address this gap by systematically exploring immune gene regulatory dynamics in melanoma progression. Methods We developed a bidirectional, weighted, signed, and directed topological immune gene regulatory network to compare transcriptional landscapes between benign melanocytic nevi and cutaneous melanoma. Advanced network analysis tools were employed to identify structural disparities and functional module shifts. Key driver genes were validated through topological centrality metrics. Additionally, deep learning models were implemented to predict drug-target interactions, leveraging molecular features derived from network analyses. Results Significant topological divergences emerged between nevi and melanoma networks, with dominant functional modules transitioning from cell cycle regulation in benign lesions to DNA repair and cell migration pathways in malignant tumors. A group of genes, including AURKA, CCNE1, APEX2, and EXOC8, were identified as potential orchestrators of immune microenvironment remodeling during malignant transformation. The deep learning framework successfully predicted 23 clinically actionable drug candidates targeting these molecular drivers. Discussion The observed module shift from cell cycle to invasion-related pathways provides mechanistic insights into melanoma progression, suggesting early therapeutic targeting of DNA repair machinery might mitigate metastatic potential. The identified hub genes, particularly AURKA and DDX19B, represent novel candidates for immunomodulatory interventions. Our computational drug prediction strategy bridges molecular network analysis with clinical translation, offering a paradigm for precision oncology in melanoma. Future studies should validate these targets in preclinical models and explore network-based biomarkers for early detection.
Collapse
Affiliation(s)
- Xiwei Cui
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jipeng Song
- Comprehensive Ward of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieyi Ren
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Feng X, Shi Y, Wu M, Cui G, Du Y, Yang J, Xu Y, Wang W, Liu F. Predicting the efficacy of neoadjuvant chemotherapy in breast cancer patients based on ultrasound longitudinal temporal depth network fusion model. Breast Cancer Res 2025; 27:30. [PMID: 40016785 PMCID: PMC11869678 DOI: 10.1186/s13058-025-01971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE The aim of this study was to develop and validate a deep learning radiomics (DLR) model based on longitudinal ultrasound data and clinical features to predict pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC) in breast cancer patients. METHODS Between January 2018 and June 2023, 312 patients with histologically confirmed breast cancer were enrolled and randomly assigned to a training cohort (n = 219) and a test cohort (n = 93) in a 7:3 ratio. Next, pre-NAC and post-treatment 2-cycle ultrasound images were collected, and radiomics and deep learning features were extracted from NAC pre-treatment (Pre), post-treatment 2 cycle (Post), and Delta (pre-NAC-NAC 2 cycle) images. In the training cohort, to filter features, the intraclass correlation coefficient test, the Boruta algorithm, and the least absolute shrinkage and selection operator (LASSO) logistic regression were used. Single-modality models (Pre, Post, and Delta) were constructed based on five machine-learning classifiers. Finally, based on the classifier with the optimal predictive performance, the DLR model was constructed by combining Pre, Post, and Delta ultrasound features and was subsequently combined with clinical features to develop a combined model (Integrated). The discriminative power, predictive performance, and clinical utility of the models were further evaluated in the test cohort. Furthermore, patients were assigned into three subgroups, including the HR+/HER2-, HER2+, and TNBC subgroups, according to molecular typing to validate the predictability of the model across the different subgroups. RESULTS After feature screening, 16, 13, and 10 features were selected to construct the Pre model, Post model, and Delta model based on the five machine learning classifiers, respectively. The three single-modality models based on the XGBoost classifier displayed optimal predictive performance. Meanwhile, the DLR model (AUC of 0.827) was superior to the single-modality model (Pre, Post, and Delta AUCs of 0.726, 0.776, and 0.710, respectively) in terms of prediction performance. Moreover, multivariate logistic regression analysis identified Her-2 status and histological grade as independent risk factors for NAC response in breast cancer. In both the training and test cohorts, the Integrated model, which included Pre, Post, and Delta ultrasound features and clinical features, exhibited the highest predictive ability, with AUC values of 0.924 and 0.875, respectively. Likewise, the Integrated model displayed the highest predictive performance across the different subgroups. CONCLUSION The Integrated model, which incorporated pre-NAC treatment and early treatment ultrasound data and clinical features, accurately predicted pCR after NAC in breast cancer patients and provided valuable insights for personalized treatment strategies, allowing for timely adjustment of chemotherapy regimens.
Collapse
Affiliation(s)
- Xiaodan Feng
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yan Shi
- Department of Ultrasonography, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, China
| | - Meng Wu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Guanghe Cui
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yao Du
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Jie Yang
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yuyuan Xu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Wenjuan Wang
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Feifei Liu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China.
| |
Collapse
|
4
|
Pan L, Liang Q, Zeng W, Peng Y, Zhao Z, Liang Y, Luo J, Wang X, Peng S. Feature-interactive Siamese graph encoder-based image analysis to predict STAS from histopathology images in lung cancer. NPJ Precis Oncol 2024; 8:285. [PMID: 39706875 DOI: 10.1038/s41698-024-00771-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024] Open
Abstract
Spread through air spaces (STAS) is a distinct invasion pattern in lung cancer, crucial for prognosis assessment and guiding surgical decisions. Histopathology is the gold standard for STAS detection, yet traditional methods are subjective, time-consuming, and prone to misdiagnosis, limiting large-scale applications. We present VERN, an image analysis model utilizing a feature-interactive Siamese graph encoder to predict STAS from lung cancer histopathological images. VERN captures spatial topological features with feature sharing and skip connections to enhance model training. Using 1,546 histopathology slides, we built a large single-cohort STAS lung cancer dataset. VERN achieved an AUC of 0.9215 in internal validation and AUCs of 0.8275 and 0.8829 in frozen and paraffin-embedded test sections, respectively, demonstrating clinical-grade performance. Validated on a single-cohort and three external datasets, VERN showed robust predictive performance and generalizability, providing an open platform ( http://plr.20210706.xyz:5000/ ) to enhance STAS diagnosis efficiency and accuracy.
Collapse
Affiliation(s)
- Liangrui Pan
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Qingchun Liang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China
| | - Wenwu Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Yijun Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Zhenyu Zhao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiyi Liang
- Oncology Department and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Hunan, China.
| | - Xiang Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China.
| |
Collapse
|
5
|
Hosseini MS, Bejnordi BE, Trinh VQH, Chan L, Hasan D, Li X, Yang S, Kim T, Zhang H, Wu T, Chinniah K, Maghsoudlou S, Zhang R, Zhu J, Khaki S, Buin A, Chaji F, Salehi A, Nguyen BN, Samaras D, Plataniotis KN. Computational pathology: A survey review and the way forward. J Pathol Inform 2024; 15:100357. [PMID: 38420608 PMCID: PMC10900832 DOI: 10.1016/j.jpi.2023.100357] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/02/2024] Open
Abstract
Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.
Collapse
Affiliation(s)
- Mahdi S. Hosseini
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | | | - Vincent Quoc-Huy Trinh
- Institute for Research in Immunology and Cancer of the University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Lyndon Chan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Danial Hasan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Xingwen Li
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Stephen Yang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Taehyo Kim
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Haochen Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Theodore Wu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Kajanan Chinniah
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Sina Maghsoudlou
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ryan Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jiadai Zhu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Samir Khaki
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Andrei Buin
- Huron Digitial Pathology, St. Jacobs, ON N0B 2N0, Canada
| | - Fatemeh Chaji
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ala Salehi
- Department of Electrical and Computer Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Bich Ngoc Nguyen
- University of Montreal Hospital Center, Montreal, QC H2X 0C2, Canada
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11794, United States
| | - Konstantinos N. Plataniotis
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| |
Collapse
|
6
|
Lapuente-Santana Ó, Kant J, Eduati F. Integrating histopathology and transcriptomics for spatial tumor microenvironment profiling in a melanoma case study. NPJ Precis Oncol 2024; 8:254. [PMID: 39511301 PMCID: PMC11543820 DOI: 10.1038/s41698-024-00749-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024] Open
Abstract
Local structures formed by cells in the tumor microenvironment (TME) play an important role in tumor development and treatment response. This study introduces SPoTLIghT, a computational framework providing a quantitative description of the tumor architecture from hematoxylin and eosin (H&E) slides. We trained a weakly supervised machine learning model on melanoma patients linking tile-level imaging features extracted from H&E slides to sample-level cell type quantifications derived from RNA-sequencing data. Using this model, SPoTLIghT provides spatial cellular maps for any H&E image, and converts them in graphs to derive 96 interpretable features capturing TME cellular organization. We show how SPoTLIghT's spatial features can distinguish microenvironment subtypes and reveal nuanced immune infiltration structures not apparent in molecular data alone. Finally, we use SPoTLIghT to effectively predict patients' prognosis in an independent melanoma cohort. SPoTLIghT enhances computational histopathology providing a quantitative and interpretable characterization of the spatial contexture of tumors.
Collapse
Affiliation(s)
- Óscar Lapuente-Santana
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Joan Kant
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Princess Margaret Cancer Research Centre, University Health Network, Toronto, Canada
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
7
|
Krishnan SN, Ji S, Elhossiny AM, Rao A, Frankel TL, Rao A. Proximogram-A multi-omics network-based framework to capture tissue heterogeneity integrating single-cell omics and spatial profiling. Comput Biol Med 2024; 182:109082. [PMID: 39255657 DOI: 10.1016/j.compbiomed.2024.109082] [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: 02/05/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024]
Abstract
The increasing availability of patient-derived multimodal biological data for various diseases has opened up avenues for finding the optimal methods for jointly leveraging the information extracted in a customizable and scalable manner. Here, we propose the Proximogram, a graph-based representation that provides a joint construct for embedding independently obtained omics and spatial data. To evaluate the representation, we generated proximograms from 2 distinct biological sources, namely, multiplexed immunofluorescence images and single-cell RNA-seq data obtained from patients across two pancreatic diseases that include normal and chronic Pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). The generated proximograms were used as inputs to 2 distinct graph deep-learning models. The improved classification results over simpler spatial-data-based input graphs point to the increased discriminatory power obtained by integrating structural information from single-cell ligand-receptor signaling data and the spatial architecture of cells in each disease class, which can help point to markers of high diagnostic significance.
Collapse
Affiliation(s)
- Santhoshi N Krishnan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Sunjong Ji
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Ahmed M Elhossiny
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Azimi M, Cho S, Bozkurt E, McDonough E, Kisakol B, Matveeva A, Salvucci M, Dussmann H, McDade S, Firat C, Urganci N, Shia J, Longley DB, Ginty F, Prehn JH. Spatial effects of infiltrating T cells on neighbouring cancer cells and prognosis in stage III CRC patients. J Pathol 2024; 264:148-159. [PMID: 39092716 DOI: 10.1002/path.6327] [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: 01/29/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring cancers, but prognostic biomarkers identifying patients at risk of recurrence are still lacking. In this study, we aimed to investigate in more detail the spatial relationship between intratumoural T cells, cancer cells, and cancer cell hallmarks as prognostic biomarkers in stage III colorectal cancer patients. We conducted multiplexed imaging of 56 protein markers at single-cell resolution on resected fixed tissue from stage III CRC patients who received adjuvant 5-fluorouracil (5FU)-based chemotherapy. Images underwent segmentation for tumour, stroma, and immune cells, and cancer cell 'state' protein marker expression was quantified at a cellular level. We developed a Python package for estimation of spatial proximity, nearest neighbour analysis focusing on cancer cell-T-cell interactions at single-cell level. In our discovery cohort (Memorial Sloan Kettering samples), we processed 462 core samples (total number of cells: 1,669,228) from 221 adjuvant 5FU-treated stage III patients. The validation cohort (Huntsville Clearview Cancer Center samples) consisted of 272 samples (total number of cells: 853,398) from 98 stage III CRC patients. While there were trends for an association between the percentage of cytotoxic T cells (across the whole cancer core), it did not reach significance (discovery cohort: p = 0.07; validation cohort: p = 0.19). We next utilised our region-based nearest neighbour approach to determine the spatial relationships between cytotoxic T cells, helper T cells, and cancer cell clusters. In both cohorts, we found that shorter distance between cytotoxic T cells, T helper cells, and cancer cells was significantly associated with increased disease-free survival. An unsupervised trained model that clustered patients based on the median distance between immune cells and cancer cells, as well as protein expression profiles, successfully classified patients into low-risk and high-risk groups (discovery cohort: p = 0.01; validation cohort: p = 0.003). © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mohammadreza Azimi
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Sanghee Cho
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Emir Bozkurt
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth McDonough
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Batuhan Kisakol
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Anna Matveeva
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Manuela Salvucci
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Simon McDade
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Canan Firat
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Nil Urganci
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Jinru Shia
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Daniel B Longley
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Fiona Ginty
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Jochen Hm Prehn
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
9
|
Mi H, Sivagnanam S, Ho WJ, Zhang S, Bergman D, Deshpande A, Baras AS, Jaffee EM, Coussens LM, Fertig EJ, Popel AS. Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology. Brief Bioinform 2024; 25:bbae421. [PMID: 39179248 PMCID: PMC11343572 DOI: 10.1093/bib/bbae421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
| | - Won Jin Ho
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Daniel Bergman
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Atul Deshpande
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Alexander S Baras
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Pathology, Johns Hopkins University School of Medicine, MD 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lisa M Coussens
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR 97201, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
| |
Collapse
|
10
|
Mentucci FM, Romero Nuñez EA, Ercole A, Silvetti V, Dal Col J, Lamberti MJ. Impact of Genomic Mutation on Melanoma Immune Microenvironment and IFN-1 Pathway-Driven Therapeutic Responses. Cancers (Basel) 2024; 16:2568. [PMID: 39061208 PMCID: PMC11274745 DOI: 10.3390/cancers16142568] [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: 06/24/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The BRAFV600E mutation, found in approximately 50% of melanoma cases, plays a crucial role in the activation of the MAPK/ERK signaling pathway, which promotes tumor cell proliferation. This study aimed to evaluate its impact on the melanoma immune microenvironment and therapeutic responses, particularly focusing on immunogenic cell death (ICD), a pivotal cytotoxic process triggering anti-tumor immune responses. Through comprehensive in silico analysis of the Cancer Genome Atlas data, we explored the association between the BRAFV600E mutation, immune subtype dynamics, and tumor mutation burden (TMB). Our findings revealed that the mutation correlated with a lower TMB, indicating a reduced generation of immunogenic neoantigens. Investigation into immune subtypes reveals an exacerbation of immunosuppression mechanisms in BRAFV600E-mutated tumors. To assess the response to ICD inducers, including doxorubicin and Me-ALA-based photodynamic therapy (PDT), compared to the non-ICD inducer cisplatin, we used distinct melanoma cell lines with wild-type BRAF (SK-MEL-2) and BRAFV600E mutation (SK-MEL-28, A375). We demonstrated a differential response to PDT between the WT and BRAFV600E cell lines. Further transcriptomic analysis revealed upregulation of IFNAR1, IFNAR2, and CXCL10 genes associated with the BRAFV600E mutation, suggesting their involvement in ICD. Using a gene reporter assay, we showed that PDT robustly activated the IFN-1 pathway through cGAS-STING signaling. Collectively, our results underscore the complex interplay between the BRAFV600E mutation and immune responses, suggesting a putative correlation between tumors carrying the mutation and their responsiveness to therapies inducing the IFN-1 pathway, such as the ICD inducer PDT, possibly mediated by the elevated expression of IFNAR1/2 receptors.
Collapse
Affiliation(s)
- Fátima María Mentucci
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Elisa Ayelén Romero Nuñez
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Agustina Ercole
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Valentina Silvetti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy;
| | - María Julia Lamberti
- Departamento de Biología Molecular, INBIAS, Universidad Nacional de Río Cuarto, Río Cuarto X5800BIA, Argentina; (F.M.M.); (V.S.)
| |
Collapse
|
11
|
Magness A, Colliver E, Enfield KSS, Lee C, Shimato M, Daly E, Moore DA, Sivakumar M, Valand K, Levi D, Hiley CT, Hobson PS, van Maldegem F, Reading JL, Quezada SA, Downward J, Sahai E, Swanton C, Angelova M. Deep cell phenotyping and spatial analysis of multiplexed imaging with TRACERx-PHLEX. Nat Commun 2024; 15:5135. [PMID: 38879602 PMCID: PMC11180132 DOI: 10.1038/s41467-024-48870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/16/2024] [Indexed: 06/19/2024] Open
Abstract
The growing scale and dimensionality of multiplexed imaging require reproducible and comprehensive yet user-friendly computational pipelines. TRACERx-PHLEX performs deep learning-based cell segmentation (deep-imcyto), automated cell-type annotation (TYPEx) and interpretable spatial analysis (Spatial-PHLEX) as three independent but interoperable modules. PHLEX generates single-cell identities, cell densities within tissue compartments, marker positivity calls and spatial metrics such as cellular barrier scores, along with summary graphs and spatial visualisations. PHLEX was developed using imaging mass cytometry (IMC) in the TRACERx study, validated using published Co-detection by indexing (CODEX), IMC and orthogonal data and benchmarked against state-of-the-art approaches. We evaluated its use on different tissue types, tissue fixation conditions, image sizes and antibody panels. As PHLEX is an automated and containerised Nextflow pipeline, manual assessment, programming skills or pathology expertise are not essential. PHLEX offers an end-to-end solution in a growing field of highly multiplexed data and provides clinically relevant insights.
Collapse
Grants
- RF\ERE\231118 Royal Society
- C416/A21999 Cancer Research UK (CRUK)
- CC2041 Wellcome Trust
- CC2041 Arthritis Research UK
- 838540 EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- RF\ERE\210216 Royal Society
- CC2040 Arthritis Research UK
- Wellcome Trust
- 101079113 EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- Francis Crick Institute (Francis Crick Institute Limited)
- Wellcome Trust (Wellcome)
- The TRACERx study (Clinicaltrials.gov no: NCT01888601) is sponsored by University College London (UCL/12/0279) and has been approved by an independent Research Ethics Committee (13/LO/1546). TRACERx is funded by Cancer Research UK (C11496/A17786) and coordinated through the Cancer Research UK and UCL Cancer Trials Centre which has a core grant from CRUK (C444/A15953). We gratefully acknowledge the patients and relatives who participated in TRACERx and PEACE studies. We thank all site personnel, investigators, funders and industry partners that supported the generation of the data within this study. This work was supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (CC2041), the UK Medical Research Council (CC2041), and the Wellcome Trust (CC2041). This work was also supported by the Cancer Research UK Lung Cancer Centre of Excellence and the CRUK City of London Centre Award (C7893/A26233) as well as the UCL Experimental Cancer Medicine Centre. This work was supported by funding as part of a research collaboration with Bristol Myers Squibb. This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement No. 101018670). For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission. C.S. is a Royal Society Napier Research Professor (RSRP\R\210001). His work is supported by the Francis Crick Institute that receives its core funding from Cancer Research UK (CC2041), the UK Medical Research Council (CC2041), and the Wellcome Trust (CC2041). C.S. is funded by Cancer Research UK (TRACERx (C11496/A17786), PEACE (C416/A21999) and CRUK Cancer Immunotherapy Catalyst Network); Cancer Research UK Lung Cancer Centre of Excellence (C11496/A30025); the Rosetrees Trust, Butterfield and Stoneygate Trusts; NovoNordisk Foundation (ID16584); Royal Society Professorship Enhancement Award (RP/EA/180007 & RF\ERE\231118)); National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre; the Cancer Research UK-University College London Centre; Experimental Cancer Medicine Centre; the Breast Cancer Research Foundation (US; BCRF-22-157); Cancer Research UK Early Detection and Diagnosis Primer Award (Grant EDDPMA-Nov21/100034); and The Mark Foundation for Cancer Research Aspire Award (Grant 21-029-ASP) and ASPIRE II award (23-034-ASP). This work was supported by a Stand Up To Cancer‐LUNGevity-American Lung Association Lung Cancer Interception Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT23-17 to S.M. Dubinett and A.E. Spira). The indicated Stand Up To Cancer grant is administered by the American Association for Cancer Research, the Scientific Partner of SU2C. C.S. is in receipt of an ERC Advanced Grant (PROTEUS) from the European Research Council under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 835297).
- K.S.S.E was supported by the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No. 838540 and is supported by the Royal Society (RF\ERE\210216).
- F.vM. is recipient of the Amsterdam UMC fellowship and receives funding from the European Research Council under the European Union’s Horizon Europe WIDERA work programme (grant agreement No. 101079113).
- E.S. was partly funded by The Mark Foundation for Cancer Research (MFCR ASPIRE 2022- 0384). E.S. is additionally supported by the European Research Council (ERC Advanced Grant CAN_ORGANISE, Grant agreement number 101019366) and the Francis Crick Institute which receives its core funding from Cancer Research UK (CC2040), the UK Medical Research Council (CC2040), and the Wellcome Trust (CC2040).
- M.A. was supported by a fellowship from The Mark Foundation for Cancer Research.
Collapse
Affiliation(s)
- Alastair Magness
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Katey S S Enfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Claudia Lee
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Masako Shimato
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Emer Daly
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - David A Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Karishma Valand
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Dina Levi
- Flow Cytometry, The Francis Crick Institute, London, UK
| | - Crispin T Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Febe van Maldegem
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - James L Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Pre-cancer Immunology Laboratory, University College London Cancer Institute, London, UK
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research, Department of Haematology, University College London Cancer Institute, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research, Department of Haematology, University College London Cancer Institute, London, UK
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| | - Mihaela Angelova
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
12
|
Enfield KS, Colliver E, Lee C, Magness A, Moore DA, Sivakumar M, Grigoriadis K, Pich O, Karasaki T, Hobson PS, Levi D, Veeriah S, Puttick C, Nye EL, Green M, Dijkstra KK, Shimato M, Akarca AU, Marafioti T, Salgado R, Hackshaw A, Jamal-Hanjani M, van Maldegem F, McGranahan N, Glass B, Pulaski H, Walk E, Reading JL, Quezada SA, Hiley CT, Downward J, Sahai E, Swanton C, Angelova M. Spatial Architecture of Myeloid and T Cells Orchestrates Immune Evasion and Clinical Outcome in Lung Cancer. Cancer Discov 2024; 14:1018-1047. [PMID: 38581685 PMCID: PMC11145179 DOI: 10.1158/2159-8290.cd-23-1380] [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: 11/17/2023] [Revised: 02/27/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024]
Abstract
Understanding the role of the tumor microenvironment (TME) in lung cancer is critical to improving patient outcomes. We identified four histology-independent archetype TMEs in treatment-naïve early-stage lung cancer using imaging mass cytometry in the TRACERx study (n = 81 patients/198 samples/2.3 million cells). In immune-hot adenocarcinomas, spatial niches of T cells and macrophages increased with clonal neoantigen burden, whereas such an increase was observed for niches of plasma and B cells in immune-excluded squamous cell carcinomas (LUSC). Immune-low TMEs were associated with fibroblast barriers to immune infiltration. The fourth archetype, characterized by sparse lymphocytes and high tumor-associated neutrophil (TAN) infiltration, had tumor cells spatially separated from vasculature and exhibited low spatial intratumor heterogeneity. TAN-high LUSC had frequent PIK3CA mutations. TAN-high tumors harbored recently expanded and metastasis-seeding subclones and had a shorter disease-free survival independent of stage. These findings delineate genomic, immune, and physical barriers to immune surveillance and implicate neutrophil-rich TMEs in metastasis. SIGNIFICANCE This study provides novel insights into the spatial organization of the lung cancer TME in the context of tumor immunogenicity, tumor heterogeneity, and cancer evolution. Pairing the tumor evolutionary history with the spatially resolved TME suggests mechanistic hypotheses for tumor progression and metastasis with implications for patient outcome and treatment. This article is featured in Selected Articles from This Issue, p. 897.
Collapse
Affiliation(s)
- Katey S.S. Enfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Claudia Lee
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Alastair Magness
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David A. Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Kristiana Grigoriadis
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Takahiro Karasaki
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
| | - Philip S. Hobson
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Dina Levi
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Clare Puttick
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Emma L. Nye
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Mary Green
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Krijn K. Dijkstra
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Masako Shimato
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ayse U. Akarca
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Teresa Marafioti
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Allan Hackshaw
- Cancer Research UK and University College London Cancer Trials Centre, London, United Kingdom
| | | | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Febe van Maldegem
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | | | | | | | - James L. Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Pre-cancer Immunology Laboratory, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Sergio A. Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Crispin T. Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Mihaela Angelova
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
13
|
Levenson RM, Singh Y, Rieck B, Hathaway QA, Farrelly C, Rozenblit J, Prasanna P, Erickson B, Choudhary A, Carlsson G, Sarkar D. Advancing Precision Medicine: Algebraic Topology and Differential Geometry in Radiology and Computational Pathology. J Transl Med 2024; 104:102060. [PMID: 38626875 DOI: 10.1016/j.labinv.2024.102060] [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: 12/15/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 05/19/2024] Open
Abstract
Precision medicine aims to provide personalized care based on individual patient characteristics, rather than guideline-directed therapies for groups of diseases or patient demographics. Images-both radiology- and pathology-derived-are a major source of information on presence, type, and status of disease. Exploring the mathematical relationship of pixels in medical imaging ("radiomics") and cellular-scale structures in digital pathology slides ("pathomics") offers powerful tools for extracting both qualitative and, increasingly, quantitative data. These analytical approaches, however, may be significantly enhanced by applying additional methods arising from fields of mathematics such as differential geometry and algebraic topology that remain underexplored in this context. Geometry's strength lies in its ability to provide precise local measurements, such as curvature, that can be crucial for identifying abnormalities at multiple spatial levels. These measurements can augment the quantitative features extracted in conventional radiomics, leading to more nuanced diagnostics. By contrast, topology serves as a robust shape descriptor, capturing essential features such as connected components and holes. The field of topological data analysis was initially founded to explore the shape of data, with functional network connectivity in the brain being a prominent example. Increasingly, its tools are now being used to explore organizational patterns of physical structures in medical images and digitized pathology slides. By leveraging tools from both differential geometry and algebraic topology, researchers and clinicians may be able to obtain a more comprehensive, multi-layered understanding of medical images and contribute to precision medicine's armamentarium.
Collapse
Affiliation(s)
- Richard M Levenson
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, California.
| | - Yashbir Singh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| | - Bastian Rieck
- Helmholtz Munich and Technical University of Munich, Munich, Germany
| | - Quincy A Hathaway
- Department of Medical Education, West Virginia University, Morgantown, West Virginia
| | | | | | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | | | | | - Gunnar Carlsson
- Department of Mathematics, Stanford University, Stanford, California
| | - Deepa Sarkar
- Institute of Genomic Health, Ichan school of Medicine, Mount Sinai, New York
| |
Collapse
|
14
|
Schreidah CM, Gordon ER, Adeuyan O, Chen C, Lapolla BA, Kent JA, Reynolds GB, Fahmy LM, Weng C, Tatonetti NP, Chase HS, Pe’er I, Geskin LJ. Current status of artificial intelligence methods for skin cancer survival analysis: a scoping review. Front Med (Lausanne) 2024; 11:1243659. [PMID: 38711781 PMCID: PMC11070520 DOI: 10.3389/fmed.2024.1243659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/22/2024] [Indexed: 05/08/2024] Open
Abstract
Skin cancer mortality rates continue to rise, and survival analysis is increasingly needed to understand who is at risk and what interventions improve outcomes. However, current statistical methods are limited by inability to synthesize multiple data types, such as patient genetics, clinical history, demographics, and pathology and reveal significant multimodal relationships through predictive algorithms. Advances in computing power and data science enabled the rise of artificial intelligence (AI), which synthesizes vast amounts of data and applies algorithms that enable personalized diagnostic approaches. Here, we analyze AI methods used in skin cancer survival analysis, focusing on supervised learning, unsupervised learning, deep learning, and natural language processing. We illustrate strengths and weaknesses of these approaches with examples. Our PubMed search yielded 14 publications meeting inclusion criteria for this scoping review. Most publications focused on melanoma, particularly histopathologic interpretation with deep learning. Such concentration on a single type of skin cancer amid increasing focus on deep learning highlight growing areas for innovation; however, it also demonstrates opportunity for additional analysis that addresses other types of cutaneous malignancies and expands the scope of prognostication to combine both genetic, histopathologic, and clinical data. Moreover, researchers may leverage multiple AI methods for enhanced benefit in analyses. Expanding AI to this arena may enable improved survival analysis, targeted treatments, and outcomes.
Collapse
Affiliation(s)
- Celine M. Schreidah
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Emily R. Gordon
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Oluwaseyi Adeuyan
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Caroline Chen
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Brigit A. Lapolla
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, United States
| | - Joshua A. Kent
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | | | - Lauren M. Fahmy
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Chunhua Weng
- The Data Science Institute, Columbia University, New York, NY, United States
- Department of Biomedical Informatics, Columbia University, New York, NY, United States
| | - Nicholas P. Tatonetti
- The Data Science Institute, Columbia University, New York, NY, United States
- Department of Biomedical Informatics, Columbia University, New York, NY, United States
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Herbert S. Chase
- Department of Biomedical Informatics, Columbia University, New York, NY, United States
| | - Itsik Pe’er
- The Data Science Institute, Columbia University, New York, NY, United States
- Department of Systems Biology, Columbia University, New York, NY, United States
- Department of Computer Science, Columbia University, New York, NY, United States
| | - Larisa J. Geskin
- Department of Dermatology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
15
|
Bao S, Zhu S, Kolachala VL, Remedios LW, Hwang Y, Sun Y, Deng R, Cui C, Zhang R, Li Y, Li J, Roland JT, Liu Q, Lau KS, Kugathasan S, Qiu P, Wilson KT, Coburn LA, Landman BA, Huo Y. Cell Spatial Analysis in Crohn's Disease: Unveiling Local Cell Arrangement Pattern with Graph-based Signatures. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2024; 12933:1293314. [PMID: 39309687 PMCID: PMC11415268 DOI: 10.1117/12.3006675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Crohn's disease (CD) is a chronic and relapsing inflammatory condition that affects segments of the gastrointestinal tract. CD activity is determined by histological findings, particularly the density of neutrophils observed on Hematoxylin and Eosin stains (H&E) imaging. However, understanding the broader morphometry and local cell arrangement beyond cell counting and tissue morphology remains challenging. To address this, we characterize six distinct cell types from H&E images and develop a novel approach for the local spatial signature of each cell. Specifically, we create a 10-cell neighborhood matrix, representing neighboring cell arrangements for each individual cell. Utilizing t-SNE for non-linear spatial projection in scatter-plot and Kernel Density Estimation contour-plot formats, our study examines patterns of differences in the cellular environment associated with the odds ratio of spatial patterns between active CD and control groups. This analysis is based on data collected at the two research institutes. The findings reveal heterogeneous nearest-neighbor patterns, signifying distinct tendencies of cell clustering, with a particular focus on the rectum region. These variations underscore the impact of data heterogeneity on cell spatial arrangements in CD patients. Moreover, the spatial distribution disparities between the two research sites highlight the significance of collaborative efforts among healthcare organizations. All research analysis pipeline tools are available at https://github.com/MASILab/cellNN.
Collapse
Affiliation(s)
- Shunxing Bao
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sichen Zhu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Vasantha L Kolachala
- Division of Pediatric Gastroenterology, Department of Pediatrics and Pediatric Research Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Lucas W Remedios
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Yeonjoo Hwang
- Division of Pediatric Gastroenterology, Department of Pediatrics and Pediatric Research Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Yutong Sun
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ruining Deng
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Can Cui
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Rendong Zhang
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Yike Li
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jia Li
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph T Roland
- Department of Surgery, Vanderbilt University, Medical Center, Nashville, TN, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ken S Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Department of Pediatrics and Pediatric Research Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Lori A Coburn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Bennett A Landman
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Yuankai Huo
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
16
|
Wagner SJ, Matek C, Shetab Boushehri S, Boxberg M, Lamm L, Sadafi A, Winter DJE, Marr C, Peng T. Built to Last? Reproducibility and Reusability of Deep Learning Algorithms in Computational Pathology. Mod Pathol 2024; 37:100350. [PMID: 37827448 DOI: 10.1016/j.modpat.2023.100350] [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: 02/10/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Recent progress in computational pathology has been driven by deep learning. While code and data availability are essential to reproduce findings from preceding publications, ensuring a deep learning model's reusability is more challenging. For that, the codebase should be well-documented and easy to integrate into existing workflows and models should be robust toward noise and generalizable toward data from different sources. Strikingly, only a few computational pathology algorithms have been reused by other researchers so far, let alone employed in a clinical setting. To assess the current state of reproducibility and reusability of computational pathology algorithms, we evaluated peer-reviewed articles available in PubMed, published between January 2019 and March 2021, in 5 use cases: stain normalization; tissue type segmentation; evaluation of cell-level features; genetic alteration prediction; and inference of grading, staging, and prognostic information. We compiled criteria for data and code availability and statistical result analysis and assessed them in 160 publications. We found that only one-quarter (41 of 160 publications) made code publicly available. Among these 41 studies, three-quarters (30 of 41) analyzed their results statistically, half of them (20 of 41) released their trained model weights, and approximately a third (16 of 41) used an independent cohort for evaluation. Our review is intended for both pathologists interested in deep learning and researchers applying algorithms to computational pathology challenges. We provide a detailed overview of publications with published code in the field, list reusable data handling tools, and provide criteria for reproducibility and reusability.
Collapse
Affiliation(s)
- Sophia J Wagner
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Christian Matek
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Sayedali Shetab Boushehri
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany; Institute of Pathology Munich-North, Munich, Germany
| | - Lorenz Lamm
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Helmholtz Pioneer Campus, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ario Sadafi
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Dominik J E Winter
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences, Technical University of Munich, Weihenstephan, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| | - Tingying Peng
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
17
|
Zhao S, Chen DP, Fu T, Yang JC, Ma D, Zhu XZ, Wang XX, Jiao YP, Jin X, Xiao Y, Xiao WX, Zhang HY, Lv H, Madabhushi A, Yang WT, Jiang YZ, Xu J, Shao ZM. Single-cell morphological and topological atlas reveals the ecosystem diversity of human breast cancer. Nat Commun 2023; 14:6796. [PMID: 37880211 PMCID: PMC10600153 DOI: 10.1038/s41467-023-42504-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Digital pathology allows computerized analysis of tumor ecosystem using whole slide images (WSIs). Here, we present single-cell morphological and topological profiling (sc-MTOP) to characterize tumor ecosystem by extracting the features of nuclear morphology and intercellular spatial relationship for individual cells. We construct a single-cell atlas comprising 410 million cells from 637 breast cancer WSIs and dissect the phenotypic diversity within tumor, inflammatory and stroma cells respectively. Spatially-resolved analysis identifies recurrent micro-ecological modules representing locoregional multicellular structures and reveals four breast cancer ecotypes correlating with distinct molecular features and patient prognosis. Further analysis with multiomics data uncovers clinically relevant ecosystem features. High abundance of locally-aggregated inflammatory cells indicates immune-activated tumor microenvironment and favorable immunotherapy response in triple-negative breast cancers. Morphological intratumor heterogeneity of tumor nuclei correlates with cell cycle pathway activation and CDK inhibitors responsiveness in hormone receptor-positive cases. sc-MTOP enables using WSIs to characterize tumor ecosystems at the single-cell level.
Collapse
Affiliation(s)
- Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - De-Pin Chen
- Institute for Artificial Intelligence in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| | - Tong Fu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing-Cheng Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Greater Bay Area Institute of Precision Medicine, Guangzhou, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiu-Zhi Zhu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiang-Xue Wang
- Institute for Artificial Intelligence in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| | - Yi-Ping Jiao
- Institute for Artificial Intelligence in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| | - Xi Jin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wen-Xuan Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hu-Yunlong Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hong Lv
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Anant Madabhushi
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Wen-Tao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Jun Xu
- Institute for Artificial Intelligence in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
18
|
Burlingame E, Ternes L, Lin JR, Chen YA, Kim EN, Gray JW, Chang YH. 3D multiplexed tissue imaging reconstruction and optimized region of interest (ROI) selection through deep learning model of channels embedding. FRONTIERS IN BIOINFORMATICS 2023; 3:1275402. [PMID: 37928169 PMCID: PMC10620917 DOI: 10.3389/fbinf.2023.1275402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction: Tissue-based sampling and diagnosis are defined as the extraction of information from certain limited spaces and its diagnostic significance of a certain object. Pathologists deal with issues related to tumor heterogeneity since analyzing a single sample does not necessarily capture a representative depiction of cancer, and a tissue biopsy usually only presents a small fraction of the tumor. Many multiplex tissue imaging platforms (MTIs) make the assumption that tissue microarrays (TMAs) containing small core samples of 2-dimensional (2D) tissue sections are a good approximation of bulk tumors although tumors are not 2D. However, emerging whole slide imaging (WSI) or 3D tumor atlases that use MTIs like cyclic immunofluorescence (CyCIF) strongly challenge this assumption. In spite of the additional insight gathered by measuring the tumor microenvironment in WSI or 3D, it can be prohibitively expensive and time-consuming to process tens or hundreds of tissue sections with CyCIF. Even when resources are not limited, the criteria for region of interest (ROI) selection in tissues for downstream analysis remain largely qualitative and subjective as stratified sampling requires the knowledge of objects and evaluates their features. Despite the fact TMAs fail to adequately approximate whole tissue features, a theoretical subsampling of tissue exists that can best represent the tumor in the whole slide image. Methods: To address these challenges, we propose deep learning approaches to learn multi-modal image translation tasks from two aspects: 1) generative modeling approach to reconstruct 3D CyCIF representation and 2) co-embedding CyCIF image and Hematoxylin and Eosin (H&E) section to learn multi-modal mappings by a cross-domain translation for minimum representative ROI selection. Results and discussion: We demonstrate that generative modeling enables a 3D virtual CyCIF reconstruction of a colorectal cancer specimen given a small subset of the imaging data at training time. By co-embedding histology and MTI features, we propose a simple convex optimization for objective ROI selection. We demonstrate the potential application of ROI selection and the efficiency of its performance with respect to cellular heterogeneity.
Collapse
Affiliation(s)
- Erik Burlingame
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, United States
| | - Luke Ternes
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, United States
| | - Jia-Ren Lin
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, United States
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Yu-An Chen
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, United States
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Eun Na Kim
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, United States
| | - Joe W. Gray
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Young Hwan Chang
- Department of Biomedical Engineering and Computational Biology Program, Oregon Health and Science University, Portland, OR, United States
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
19
|
Fu X, Sahai E, Wilkins A. Application of digital pathology-based advanced analytics of tumour microenvironment organisation to predict prognosis and therapeutic response. J Pathol 2023; 260:578-591. [PMID: 37551703 PMCID: PMC10952145 DOI: 10.1002/path.6153] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 08/09/2023]
Abstract
In recent years, the application of advanced analytics, especially artificial intelligence (AI), to digital H&E images, and other histological image types, has begun to radically change how histological images are used in the clinic. Alongside the recognition that the tumour microenvironment (TME) has a profound impact on tumour phenotype, the technical development of highly multiplexed immunofluorescence platforms has enhanced the biological complexity that can be captured in the TME with high precision. AI has an increasingly powerful role in the recognition and quantitation of image features and the association of such features with clinically important outcomes, as occurs in distinct stages in conventional machine learning. Deep-learning algorithms are able to elucidate TME patterns inherent in the input data with minimum levels of human intelligence and, hence, have the potential to achieve clinically relevant predictions and discovery of important TME features. Furthermore, the diverse repertoire of deep-learning algorithms able to interrogate TME patterns extends beyond convolutional neural networks to include attention-based models, graph neural networks, and multimodal models. To date, AI models have largely been evaluated retrospectively, outside the well-established rigour of prospective clinical trials, in part because traditional clinical trial methodology may not always be suitable for the assessment of AI technology. However, to enable digital pathology-based advanced analytics to meaningfully impact clinical care, specific measures of 'added benefit' to the current standard of care and validation in a prospective setting are important. This will need to be accompanied by adequate measures of explainability and interpretability. Despite such challenges, the combination of expanding datasets, increased computational power, and the possibility of integration of pre-clinical experimental insights into model development means there is exciting potential for the future progress of these AI applications. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xiao Fu
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| | - Erik Sahai
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Anna Wilkins
- Tumour Cell Biology LaboratoryThe Francis Crick InstituteLondonUK
- Division of Radiotherapy and ImagingInstitute of Cancer ResearchLondonUK
- Royal Marsden Hospitals NHS TrustLondonUK
| |
Collapse
|
20
|
Lu M, Zou Y, Fu P, Li Y, Wang P, Li G, Luo S, Chen Y, Guan G, Zhang S, Chen L. The tumor-stroma ratio and the immune microenvironment improve the prognostic prediction of pancreatic ductal adenocarcinoma. Discov Oncol 2023; 14:124. [PMID: 37405518 DOI: 10.1007/s12672-023-00744-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
Tumor-infiltrating immune cells and fibroblasts are significant components of the tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC), and they participate in tumor progression as closely as tumor cells. However, the relationship between the features of the TME and patient outcomes and the interactions among TME components are still unclear. In this study, we evaluated the PDAC TME in terms of the quantity and location of cluster of differentiation (CD)4+ T cells, CD8+ T cells, macrophages, stromal maturity, and tumor-stroma ratio (TSR), as evaluated by immunohistochemical staining of serial whole-tissue sections from 116 patients with PDAC. The density of T cells and macrophages (mainly activated macrophages) was significantly higher at the invasive margins (IMs) than at the tumor center (TC). CD4+ T cells were significantly association with all the other tumor-associated immune cells (TAIs) including CD8, CD68 and CD206 positive cells. Tumors of the non-mature (intermediate and immature) stroma type harbored significantly more CD8+ T cells at the IMs and more CD68+ macrophages at the IMs and the TC. The density of CD4+, CD8+, and CD206+ cells at the TC; CD206+ cells at the IMs; and tumor-node-metastasis (TNM) staging were independent risk factors for patient outcomes, and the c-index of the risk nomogram for predicting the survival probability based on the TME features and TNM staging was 0.772 (95% confidence interval: 0.713-0.832). PDAC harbored a significantly immunosuppressive TME, of which the IMs were the hot zones for TAIs, while cells at the TC were more predictive of prognosis. Our results indicated that the model based on the features of the TME and TNM staging could predict patient outcomes.
Collapse
Affiliation(s)
- Mei Lu
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
- Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, Fujian, China
| | - Yi Zou
- Department of Pathology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peiling Fu
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Yuyang Li
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Pengcheng Wang
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Guoping Li
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Sheng Luo
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Yupeng Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Guoping Guan
- Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, Fujian, China
| | - Sheng Zhang
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China.
- Department of Pathology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
- Fujian Key Laboratory of Translational Research in Cancer and Nurodegernerative Diseases, Fuzhou, Fujian, China.
| |
Collapse
|
21
|
Clifton GT, Rothenberg M, Ascierto PA, Begley G, Cecchini M, Eder JP, Ghiringhelli F, Italiano A, Kochetkova M, Li R, Mechta-Grigoriou F, Pai SI, Provenzano P, Puré E, Ribas A, Schalper KA, Fridman WH. Developing a definition of immune exclusion in cancer: results of a modified Delphi workshop. J Immunother Cancer 2023; 11:e006773. [PMID: 37290925 PMCID: PMC10254706 DOI: 10.1136/jitc-2023-006773] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/10/2023] Open
Abstract
Checkpoint inhibitors represent an effective treatment approach for a variety of cancers through their inhibition of immune regulatory pathways within the tumor microenvironment (TME). Unfortunately only a minority of patients with cancer achieve clinical benefit from immunotherapy, with the TME emerging as an important predictor of outcomes and sensitivity to therapy. The extent and pattern of T-cell infiltration can vary prominently within/across tumors and represents a biological continuum. Three immune profiles have been identified along this continuum: 'immune-desert' or 'T-cell cold' phenotype, 'immune-active', 'inflamed', or 'T-cell hot' phenotype, and 'immune excluded' phenotype. Of the three profiles, immune excluded remains the most ill-defined with no clear, universally accepted definition even though it is commonly associated with lack of response to immune checkpoint inhibitors and poor clinical outcomes. To address this, 16 multidisciplinary cancer experts from around the world were invited to participate in a symposium using a three-round modified Delphi approach. The first round was an open-ended questionnaire distributed via email and the second was an in-person discussion of the first round results that allowed for statements to be revised as necessary to achieve a maximum consensus (75% agreement) among the rating committee (RC). The final round questionnaire was distributed to the RC via email and had a 100% completion rate. The Delphi process resulted in moving us closer to a consensus definition for immune exclusion that is practical, clinically pertinent, and applicable across a wide range of cancer histologies. A general consensus of the role of immune exclusion in resistance to checkpoint therapy and five research priorities emerged from this process. Together, these tools could help efforts designed to address the underlying mechanisms of immune exclusion that span cancer types and, ultimately, aid in the development of treatments to target these mechanisms to improve patient outcomes.
Collapse
Affiliation(s)
| | - Mace Rothenberg
- Consultant, Parthenon Therapeutics, Boston, Massachusetts, USA
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, IRCCS Fondazione "G. Pascale", Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Glenn Begley
- Parthenon Therapeutics, Boston, Massachusetts, USA
| | - Michael Cecchini
- Department of Internal Medicine, Division of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Francois Ghiringhelli
- Department of Medical Oncology, Georges François Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Antoine Italiano
- Early Phase Trial Unit, Institut Bergonié, Bordeaux 33000, France
| | - Marina Kochetkova
- Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | | | - Sara I Pai
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Paolo Provenzano
- Department of Biomedical Engineering, University of Minnesota System, Minneapolis, Minnesota, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Antoni Ribas
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Wolf Herve Fridman
- Department of Immunology, Inflammation and Cancer, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
22
|
Jiménez-Sánchez D, López-Janeiro Á, Villalba-Esparza M, Ariz M, Kadioglu E, Masetto I, Goubert V, Lozano MD, Melero I, Hardisson D, Ortiz-de-Solórzano C, de Andrea CE. Weakly supervised deep learning to predict recurrence in low-grade endometrial cancer from multiplexed immunofluorescence images. NPJ Digit Med 2023; 6:48. [PMID: 36959234 PMCID: PMC10036616 DOI: 10.1038/s41746-023-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/10/2023] [Indexed: 03/25/2023] Open
Abstract
Predicting recurrence in low-grade, early-stage endometrial cancer (EC) is both challenging and clinically relevant. We present a weakly-supervised deep learning framework, NaroNet, that can learn, without manual expert annotation, the complex tumor-immune interrelations at three levels: local phenotypes, cellular neighborhoods, and tissue areas. It uses multiplexed immunofluorescence for the simultaneous visualization and quantification of CD68 + macrophages, CD8 + T cells, FOXP3 + regulatory T cells, PD-L1/PD-1 protein expression, and tumor cells. We used 489 tumor cores from 250 patients to train a multilevel deep-learning model to predict tumor recurrence. Using a tenfold cross-validation strategy, our model achieved an area under the curve of 0.90 with a 95% confidence interval of 0.83-0.95. Our model predictions resulted in concordance for 96,8% of cases (κ = 0.88). This method could accurately assess the risk of recurrence in EC, outperforming current prognostic factors, including molecular subtyping.
Collapse
Affiliation(s)
- Daniel Jiménez-Sánchez
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Álvaro López-Janeiro
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - María Villalba-Esparza
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Mikel Ariz
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
| | - Ece Kadioglu
- Lunaphore Technologies SA, Tolochenaz, Switzerland
| | | | | | - Maria D Lozano
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - David Hardisson
- Department of Pathology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital, IdiPAZ, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Ortiz-de-Solórzano
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain
| | - Carlos E de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain.
- Center for Biomedical Research in the Cancer Network (CIBERONC), Madrid, Spain.
| |
Collapse
|
23
|
Alemanno F, Cavo M, Delle Cave D, Fachechi A, Rizzo R, D’Amone E, Gigli G, Lonardo E, Barra A, del Mercato LL. Quantifying heterogeneity to drug response in cancer-stroma kinetics. Proc Natl Acad Sci U S A 2023; 120:e2122352120. [PMID: 36897966 PMCID: PMC10089157 DOI: 10.1073/pnas.2122352120] [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: 12/13/2021] [Accepted: 02/04/2023] [Indexed: 03/12/2023] Open
Abstract
A crucial challenge in medicine is choosing which drug (or combination) will be the most advantageous for a particular patient. Usually, drug response rates differ substantially, and the reasons for this response unpredictability remain ambiguous. Consequently, it is central to classify features that contribute to the observed drug response variability. Pancreatic cancer is one of the deadliest cancers with limited therapeutic achievements due to the massive presence of stroma that generates an environment that enables tumor growth, metastasis, and drug resistance. To understand the cancer-stroma cross talk within the tumor microenvironment and to develop personalized adjuvant therapies, there is a necessity for effective approaches that offer measurable data to monitor the effect of drugs at the single-cell level. Here, we develop a computational approach, based on cell imaging, that quantifies the cellular cross talk between pancreatic tumor cells (L3.6pl or AsPC1) and pancreatic stellate cells (PSCs), coordinating their kinetics in presence of the chemotherapeutic agent gemcitabine. We report significant heterogeneity in the organization of cellular interactions in response to the drug. For L3.6pl cells, gemcitabine sensibly decreases stroma-stroma interactions but increases stroma-cancer interactions, overall enhancing motility and crowding. In the AsPC1 case, gemcitabine promotes the interactions among tumor cells, but it does not affect stroma-cancer interplay, possibly suggesting a milder effect of the drug on cell dynamics.
Collapse
Affiliation(s)
- Francesco Alemanno
- Institute of Nanotechnology, National Research Council, Lecce73100, Italy
- Dipartimento di Matematica e Fisica Ennio De Giorgi, Università del Salento, Lecce73100, Italy
| | - Marta Cavo
- Institute of Nanotechnology, National Research Council, Lecce73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, CNR, Naples80131, Italy
| | - Alberto Fachechi
- Dipartimento di Matematica Guido Castelnuovo, Sapienza Università di Roma, Rome00185, Italy
| | - Riccardo Rizzo
- Institute of Nanotechnology, National Research Council, Lecce73100, Italy
| | - Eliana D’Amone
- Institute of Nanotechnology, National Research Council, Lecce73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council, Lecce73100, Italy
- Dipartimento di Matematica e Fisica Ennio De Giorgi, Università del Salento, Lecce73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, CNR, Naples80131, Italy
| | - Adriano Barra
- Dipartimento di Matematica e Fisica Ennio De Giorgi, Università del Salento, Lecce73100, Italy
- Istituto Nazionale di Fisica Nucleare, Sezione di Lecce, Lecce73100, Italy
| | | |
Collapse
|
24
|
Bull JA, Byrne HM. Quantification of spatial and phenotypic heterogeneity in an agent-based model of tumour-macrophage interactions. PLoS Comput Biol 2023; 19:e1010994. [PMID: 36972297 PMCID: PMC10079237 DOI: 10.1371/journal.pcbi.1010994] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/06/2023] [Accepted: 03/04/2023] [Indexed: 03/29/2023] Open
Abstract
We introduce a new spatial statistic, the weighted pair correlation function (wPCF). The wPCF extends the existing pair correlation function (PCF) and cross-PCF to describe spatial relationships between points marked with combinations of discrete and continuous labels. We validate its use through application to a new agent-based model (ABM) which simulates interactions between macrophages and tumour cells. These interactions are influenced by the spatial positions of the cells and by macrophage phenotype, a continuous variable that ranges from anti-tumour to pro-tumour. By varying model parameters that regulate macrophage phenotype, we show that the ABM exhibits behaviours which resemble the 'three Es of cancer immunoediting': Equilibrium, Escape, and Elimination. We use the wPCF to analyse synthetic images generated by the ABM. We show that the wPCF generates a 'human readable' statistical summary of where macrophages with different phenotypes are located relative to both blood vessels and tumour cells. We also define a distinct 'PCF signature' that characterises each of the three Es of immunoediting, by combining wPCF measurements with the cross-PCF describing interactions between vessels and tumour cells. By applying dimension reduction techniques to this signature, we identify its key features and train a support vector machine classifier to distinguish between simulation outputs based on their PCF signature. This proof-of-concept study shows how multiple spatial statistics can be combined to analyse the complex spatial features that the ABM generates, and to partition them into interpretable groups. The intricate spatial features produced by the ABM are similar to those generated by state-of-the-art multiplex imaging techniques which distinguish the spatial distribution and intensity of multiple biomarkers in biological tissue regions. Applying methods such as the wPCF to multiplex imaging data would exploit the continuous variation in biomarker intensities and generate more detailed characterisation of the spatial and phenotypic heterogeneity in tissue samples.
Collapse
Affiliation(s)
- Joshua A. Bull
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Helen M. Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Couetil J, Liu Z, Huang K, Zhang J, Alomari AK. Predicting melanoma survival and metastasis with interpretable histopathological features and machine learning models. Front Med (Lausanne) 2023; 9:1029227. [PMID: 36687402 PMCID: PMC9853175 DOI: 10.3389/fmed.2022.1029227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/16/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Melanoma is the fifth most common cancer in US, and the incidence is increasing 1.4% annually. The overall survival rate for early-stage disease is 99.4%. However, melanoma can recur years later (in the same region of the body or as distant metastasis), and results in a dramatically lower survival rate. Currently there is no reliable method to predict tumor recurrence and metastasis on early primary tumor histological images. Methods To identify rapid, accurate, and cost-effective predictors of metastasis and survival, in this work, we applied various interpretable machine learning approaches to analyze melanoma histopathological H&E images. The result is a set of image features that can help clinicians identify high-risk-of-metastasis patients for increased clinical follow-up and precision treatment. We use simple models (i.e., logarithmic classification and KNN) and "human-interpretable" measures of cell morphology and tissue architecture (e.g., cell size, staining intensity, and cell density) to predict the melanoma survival on public and local Stage I-III cohorts as well as the metastasis risk on a local cohort. Results We use penalized survival regression to limit features available to downstream classifiers and investigate the utility of convolutional neural networks in isolating tumor regions to focus morphology extraction on only the tumor region. This approach allows us to predict survival and metastasis with a maximum F1 score of 0.72 and 0.73, respectively, and to visualize several high-risk cell morphologies. Discussion This lays the foundation for future work, which will focus on using our interpretable pipeline to predict metastasis in Stage I & II melanoma.
Collapse
Affiliation(s)
- Justin Couetil
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ziyu Liu
- Department of Statistics, Purdue University, West Lafayette, IN, United States
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ahmed K. Alomari
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
26
|
Tiwari A, Oravecz T, Dillon LA, Italiano A, Audoly L, Fridman WH, Clifton GT. Towards a consensus definition of immune exclusion in cancer. Front Immunol 2023; 14:1084887. [PMID: 37033994 PMCID: PMC10073666 DOI: 10.3389/fimmu.2023.1084887] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/14/2023] [Indexed: 04/11/2023] Open
Abstract
Background The immune cell topography of solid tumors has been increasingly recognized as an important predictive factor for progression of disease and response to immunotherapy. The distribution pattern of immune cells in solid tumors is commonly classified into three categories - namely, "Immune inflamed", "Immune desert" and "Immune excluded" - which, to some degree, connect immune cell presence and positioning within the tumor microenvironment to anti-tumor activity. Materials and methods In this review, we look at the ways immune exclusion has been defined in published literature and identify opportunities to develop consistent, quantifiable definitions, which in turn, will allow better determination of the underlying mechanisms that span cancer types and, ultimately, aid in the development of treatments to target these mechanisms. Results The definitions of tumor immune phenotypes, especially immune exclusion, have largely been conceptual. The existing literature lacks in consistency when it comes to practically defining immune exclusion, and there is no consensus on a definition. Majority of the definitions use somewhat arbitrary cut-offs in an attempt to place each tumor into a distinct phenotypic category. Tumor heterogeneity is often not accounted for, which limits the practical application of a definition. Conclusions We have identified two key issues in existing definitions of immune exclusion, establishing clinically relevant cut-offs within the spectrum of immune cell infiltration as well as tumor heterogeneity. We propose an approach to overcome these limitations, by reporting the degree of immune cell infiltration, tying cut-offs to clinically meaningful outcome measures, maximizing the number of regions of a tumor that are analyzed and reporting the degree of heterogeneity. This will allow for a consensus practical definition for operationalizing this categorization into clinical trial and signal-seeking endpoints.
Collapse
Affiliation(s)
- Ankur Tiwari
- Department of Surgery, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | | | | | | | | | - Wolf Hervé Fridman
- Centre de Recherche des Cordeliers, National Institute for Health and Medical Research (INSERM), Sorbonne Université, Université Sorbonne Paris-Cité (USPC), Université de Paris, Equipe Inflammation, Paris, France
| | - Guy Travis Clifton
- Parthenon Therapeutics, Boston, MA, United States
- *Correspondence: Guy Travis Clifton,
| |
Collapse
|
27
|
Thol K, Pawlik P, McGranahan N. Therapy sculpts the complex interplay between cancer and the immune system during tumour evolution. Genome Med 2022; 14:137. [PMID: 36476325 PMCID: PMC9730559 DOI: 10.1186/s13073-022-01138-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer development is an evolutionary process. A key selection pressure is exerted by therapy, one of the few players in cancer evolution that can be controlled. As such, an understanding of how treatment acts to sculpt the tumour and its microenvironment and how this influences a tumour's subsequent evolutionary trajectory is critical. In this review, we examine cancer evolution and intra-tumour heterogeneity in the context of therapy. We focus on how radiotherapy, chemotherapy and immunotherapy shape both tumour development and the environment in which tumours evolve and how resistance can develop or be selected for during treatment.
Collapse
Affiliation(s)
- Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Piotr Pawlik
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Genome Evolution Research Group, University College London Cancer Institute, London, UK.
| |
Collapse
|
28
|
Mi H, Sivagnanam S, Betts CB, Liudahl SM, Jaffee EM, Coussens LM, Popel AS. Quantitative Spatial Profiling of Immune Populations in Pancreatic Ductal Adenocarcinoma Reveals Tumor Microenvironment Heterogeneity and Prognostic Biomarkers. Cancer Res 2022; 82:4359-4372. [PMID: 36112643 PMCID: PMC9716253 DOI: 10.1158/0008-5472.can-22-1190] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 01/24/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with poor 5-year survival rates, necessitating identification of novel therapeutic targets. Elucidating the biology of the tumor immune microenvironment (TiME) can provide vital insights into mechanisms of tumor progression. In this study, we developed a quantitative image processing platform to analyze sequential multiplexed IHC data from archival PDAC tissue resection specimens. A 27-plex marker panel was employed to simultaneously phenotype cell populations and their functional states, followed by a computational workflow to interrogate the immune contextures of the TiME in search of potential biomarkers. The PDAC TiME reflected a low-immunogenic ecosystem with both high intratumoral and intertumoral heterogeneity. Spatial analysis revealed that the relative distance between IL10+ myelomonocytes, PD-1+ CD4+ T cells, and granzyme B+ CD8+ T cells correlated significantly with survival, from which a spatial proximity signature termed imRS was derived that correlated with PDAC patient survival. Furthermore, spatial enrichment of CD8+ T cells in lymphoid aggregates was also linked to improved survival. Altogether, these findings indicate that the PDAC TiME, generally considered immuno-dormant or immunosuppressive, is a spatially nuanced ecosystem orchestrated by ordered immune hierarchies. This new understanding of spatial complexity may guide novel treatment strategies for PDAC. SIGNIFICANCE Quantitative image analysis of PDAC specimens reveals intertumoral and intratumoral heterogeneity of immune populations and identifies spatial immune architectures that are significantly associated with disease prognosis.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | | | - Courtney B. Betts
- Department of Cell, Development, and Cancer Biology, Oregon Health and Science University, Portland, Oregon.
| | - Shannon M. Liudahl
- Department of Cell, Development, and Cancer Biology, Oregon Health and Science University, Portland, Oregon.
| | - Elizabeth M. Jaffee
- Skip Viragh Center for Pancreatic Cancer, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medicine, Baltimore, Maryland.
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Lisa M. Coussens
- Department of Cell, Development, and Cancer Biology, Oregon Health and Science University, Portland, Oregon.
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, Oregon.
- Knight Cancer Institute, Portland, Oregon.
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
29
|
Kim I, Kang K, Song Y, Kim TJ. Application of Artificial Intelligence in Pathology: Trends and Challenges. Diagnostics (Basel) 2022; 12:2794. [PMID: 36428854 PMCID: PMC9688959 DOI: 10.3390/diagnostics12112794] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Given the recent success of artificial intelligence (AI) in computer vision applications, many pathologists anticipate that AI will be able to assist them in a variety of digital pathology tasks. Simultaneously, tremendous advancements in deep learning have enabled a synergy with artificial intelligence (AI), allowing for image-based diagnosis on the background of digital pathology. There are efforts for developing AI-based tools to save pathologists time and eliminate errors. Here, we describe the elements in the development of computational pathology (CPATH), its applicability to AI development, and the challenges it faces, such as algorithm validation and interpretability, computing systems, reimbursement, ethics, and regulations. Furthermore, we present an overview of novel AI-based approaches that could be integrated into pathology laboratory workflows.
Collapse
Affiliation(s)
- Inho Kim
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Kyungmin Kang
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Youngjae Song
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Republic of Korea
| |
Collapse
|
30
|
Alsaleh L, Li C, Couetil JL, Ye Z, Huang K, Zhang J, Chen C, Johnson TS. Spatial Transcriptomic Analysis Reveals Associations between Genes and Cellular Topology in Breast and Prostate Cancers. Cancers (Basel) 2022; 14:4856. [PMID: 36230778 PMCID: PMC9562681 DOI: 10.3390/cancers14194856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cancer is the leading cause of death worldwide with breast and prostate cancer the most common among women and men, respectively. Gene expression and image features are independently prognostic of patient survival; but until the advent of spatial transcriptomics (ST), it was not possible to determine how gene expression of cells was tied to their spatial relationships (i.e., topology). METHODS We identify topology-associated genes (TAGs) that correlate with 700 image topological features (ITFs) in breast and prostate cancer ST samples. Genes and image topological features are independently clustered and correlated with each other. Themes among genes correlated with ITFs are investigated by functional enrichment analysis. RESULTS Overall, topology-associated genes (TAG) corresponding to extracellular matrix (ECM) and Collagen Type I Trimer gene ontology terms are common to both prostate and breast cancer. In breast cancer specifically, we identify the ZAG-PIP Complex as a TAG. In prostate cancer, we identify distinct TAGs that are enriched for GI dysmotility and the IgA immunoglobulin complex. We identified TAGs in every ST slide regardless of cancer type. CONCLUSIONS These TAGs are enriched for ontology terms, illustrating the biological relevance to our image topology features and their potential utility in diagnostic and prognostic models.
Collapse
Affiliation(s)
- Lujain Alsaleh
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
| | - Chen Li
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Justin L. Couetil
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Ze Ye
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
- Regenstrief Institute, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - Chao Chen
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Travis S. Johnson
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Jiang J, Tekin B, Yuan L, Armasu S, Winham SJ, Goode EL, Liu H, Huang Y, Guo R, Wang C. Computational tumor stroma reaction evaluation led to novel prognosis-associated fibrosis and molecular signature discoveries in high-grade serous ovarian carcinoma. Front Med (Lausanne) 2022; 9:994467. [PMID: 36160147 PMCID: PMC9490262 DOI: 10.3389/fmed.2022.994467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background As one of the key criteria to differentiate benign vs. malignant tumors in ovarian and other solid cancers, tumor-stroma reaction (TSR) is long observed by pathologists and has been found correlated with patient prognosis. However, paucity of study aims to overcome subjective bias or automate TSR evaluation for enabling association analysis to a large cohort. Materials and methods Serving as positive and negative sets of TSR studies, H&E slides of primary tumors of high-grade serous ovarian carcinoma (HGSOC) (n = 291) and serous borderline ovarian tumor (SBOT) (n = 15) were digitally scanned. Three pathologist-defined quantification criteria were used to characterize the extents of TSR. Scores for each criterion were annotated (0/1/2 as none-low/intermediate/high) in the training set consisting of 18,265 H&E patches. Serial of deep learning (DL) models were trained to identify tumor vs. stroma regions and predict TSR scores. After cross-validation and independent validations, the trained models were generalized to the entire HGSOC cohort and correlated with clinical characteristics. In a subset of cases tumor transcriptomes were available, gene- and pathway-level association studies were conducted with TSR scores. Results The trained models accurately identified the tumor stroma tissue regions and predicted TSR scores. Within tumor stroma interface region, TSR fibrosis scores were strongly associated with patient prognosis. Cancer signaling aberrations associated 14 KEGG pathways were also found positively correlated with TSR-fibrosis score. Conclusion With the aid of DL, TSR evaluation could be generalized to large cohort to enable prognostic association analysis and facilitate discovering novel gene and pathways associated with disease progress.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Burak Tekin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Lin Yuan
- Pathology Center, Shanghai General Hospital, Shanghai, China
| | - Sebastian Armasu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Stacey J. Winham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Ellen L. Goode
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
- Hongfang Liu,
| | - Yajue Huang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
- Yajue Huang,
| | - Ruifeng Guo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
- Ruifeng Guo,
| | - Chen Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Chen Wang,
| |
Collapse
|
32
|
Alzoubi I, Bao G, Zhang R, Loh C, Zheng Y, Cherepanoff S, Gracie G, Lee M, Kuligowski M, Alexander KL, Buckland ME, Wang X, Graeber MB. An Open-Source AI Framework for the Analysis of Single Cells in Whole-Slide Images with a Note on CD276 in Glioblastoma. Cancers (Basel) 2022; 14:3441. [PMID: 35884502 PMCID: PMC9316952 DOI: 10.3390/cancers14143441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
Routine examination of entire histological slides at cellular resolution poses a significant if not insurmountable challenge to human observers. However, high-resolution data such as the cellular distribution of proteins in tissues, e.g., those obtained following immunochemical staining, are highly desirable. Our present study extends the applicability of the PathoFusion framework to the cellular level. We illustrate our approach using the detection of CD276 immunoreactive cells in glioblastoma as an example. Following automatic identification by means of PathoFusion's bifocal convolutional neural network (BCNN) model, individual cells are automatically profiled and counted. Only discriminable cells selected through data filtering and thresholding were segmented for cell-level analysis. Subsequently, we converted the detection signals into the corresponding heatmaps visualizing the distribution of the detected cells in entire whole-slide images of adjacent H&E-stained sections using the Discrete Wavelet Transform (DWT). Our results demonstrate that PathoFusion is capable of autonomously detecting and counting individual immunochemically labelled cells with a high prediction performance of 0.992 AUC and 97.7% accuracy. The data can be used for whole-slide cross-modality analyses, e.g., relationships between immunochemical signals and anaplastic histological features. PathoFusion has the potential to be applied to additional problems that seek to correlate heterogeneous data streams and to serve as a clinically applicable, weakly supervised system for histological image analyses in (neuro)pathology.
Collapse
Affiliation(s)
- Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia; (I.A.); (G.B.); (R.Z.)
| | - Guoqing Bao
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia; (I.A.); (G.B.); (R.Z.)
| | - Rong Zhang
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia; (I.A.); (G.B.); (R.Z.)
| | - Christina Loh
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (C.L.); (Y.Z.)
| | - Yuqi Zheng
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (C.L.); (Y.Z.)
| | - Svetlana Cherepanoff
- St Vincent’s Hospital, Victoria Street, Darlinghurst, NSW 2010, Australia; (S.C.); (G.G.)
| | - Gary Gracie
- St Vincent’s Hospital, Victoria Street, Darlinghurst, NSW 2010, Australia; (S.C.); (G.G.)
| | - Maggie Lee
- Department of Neuropathology, RPA Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (M.L.); (K.L.A.); (M.E.B.)
| | - Michael Kuligowski
- Sydney Microscopy and Microanalysis, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Kimberley L. Alexander
- Department of Neuropathology, RPA Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (M.L.); (K.L.A.); (M.E.B.)
- Neurosurgery Department, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Michael E. Buckland
- Department of Neuropathology, RPA Hospital and Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; (M.L.); (K.L.A.); (M.E.B.)
| | - Xiuying Wang
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia; (I.A.); (G.B.); (R.Z.)
| | - Manuel B. Graeber
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia; (C.L.); (Y.Z.)
| |
Collapse
|
33
|
Lou P, Wang C, Guo R, Yao L, Zhang G, Yang J, Yuan Y, Dong Y, Gao Z, Gong T, Li C. HistoML, a markup language for representation and exchange of histopathological features in pathology images. Sci Data 2022; 9:387. [PMID: 35803960 PMCID: PMC9270329 DOI: 10.1038/s41597-022-01505-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
The study of histopathological phenotypes is vital for cancer research and medicine as it links molecular mechanisms to disease prognosis. It typically involves integration of heterogenous histopathological features in whole-slide images (WSI) to objectively characterize a histopathological phenotype. However, the large-scale implementation of phenotype characterization has been hindered by the fragmentation of histopathological features, resulting from the lack of a standardized format and a controlled vocabulary for structured and unambiguous representation of semantics in WSIs. To fill this gap, we propose the Histopathology Markup Language (HistoML), a representation language along with a controlled vocabulary (Histopathology Ontology) based on Semantic Web technologies. Multiscale features within a WSI, from single-cell features to mesoscopic features, could be represented using HistoML which is a crucial step towards the goal of making WSIs findable, accessible, interoperable and reusable (FAIR). We pilot HistoML in representing WSIs of kidney cancer as well as thyroid carcinoma and exemplify the uses of HistoML representations in semantic queries to demonstrate the potential of HistoML-powered applications for phenotype characterization.
Collapse
Affiliation(s)
- Peiliang Lou
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Chunbao Wang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, China
| | - Ruifeng Guo
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lixia Yao
- Department of Health Services Administration and Policy, Temple University, Philadelphia, PA, USA
| | - Guanjun Zhang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, China
| | - Jun Yang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 3, Shang Qin Road, Xi'an, Shaanxi, China
| | - Yong Yuan
- Department of Pathology, Shaanxi Provincial Tumor Hospital, Xi'an Jiaotong University, 309 Yanta West Road, Xi'an, Shaanxi, China
| | - Yuxin Dong
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Zeyu Gao
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Tieliang Gong
- Key Laboratory of Intelligent Networks and Network Security (Xi'an Jiaotong University), Ministry of Education, Xi'an, Shaanxi, 710049, China
| | - Chen Li
- National Engineering Lab for Big Data Analytics, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China.
| |
Collapse
|
34
|
Xu H, Clemenceau JR, Park S, Choi J, Lee SH, Hwang TH. Spatial heterogeneity and organization of tumor mutation burden with immune infiltrates within tumors based on whole slide images correlated with patient survival in bladder cancer. J Pathol Inform 2022; 13:100105. [PMID: 36268064 PMCID: PMC9577053 DOI: 10.1016/j.jpi.2022.100105] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Background High tumor mutation burden (TMB-H) could result in an increased number of neoepitopes from somatic mutations expressed by a patient's own tumor cell which can be recognized and targeted by neighboring tumor-infiltrating lymphocytes (TILs). Deeper understanding of spatial heterogeneity and organization of tumor cells and their neighboring immune infiltrates within tumors could provide new insights into tumor progression and treatment response. Methods Here we first developed computational approaches using whole slide images (WSIs) to predict bladder cancer patients' TMB status and TILs across tumor regions, and then investigate spatial heterogeneity and organization of regions harboring TMB-H tumor cells and TILs within tumors, as well as their prognostic utility. Results: In experiments using WSIs from The Cancer Genome Atlas (TCGA) bladder cancer (BLCA), our findings show that computational pathology can reliably predict patient-level TMB status and delineate spatial TMB heterogeneity and co-organization with TILs. TMB-H patients with low spatial heterogeneity enriched with high TILs show improved overall survival. Conclusions Computational approaches using WSIs have the potential to provide rapid and cost-effective TMB testing and TILs detection. Survival analysis illuminates potential clinical utility of spatial heterogeneity and co-organization of TMB and TILs as a prognostic biomarker in BLCA which warrants further validation in future studies.
Collapse
Affiliation(s)
- Hongming Xu
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China
- Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Jean René Clemenceau
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sunho Park
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jinhwan Choi
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St.Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Tae Hyun Hwang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
35
|
Wu L, Ye W, Liu Y, Chen D, Wang Y, Cui Y, Li Z, Li P, Li Z, Liu Z, Liu M, Liang C, Yang X, Xie Y, Wang Y. An integrated deep learning model for the prediction of pathological complete response to neoadjuvant chemotherapy with serial ultrasonography in breast cancer patients: a multicentre, retrospective study. Breast Cancer Res 2022; 24:81. [PMID: 36414984 PMCID: PMC9680135 DOI: 10.1186/s13058-022-01580-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/13/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The biological phenotype of tumours evolves during neoadjuvant chemotherapy (NAC). Accurate prediction of pathological complete response (pCR) to NAC in the early-stage or posttreatment can optimize treatment strategies or improve the breast-conserving rate. This study aimed to develop and validate an autosegmentation-based serial ultrasonography assessment system (SUAS) that incorporated serial ultrasonographic features throughout the NAC of breast cancer to predict pCR. METHODS A total of 801 patients with biopsy-proven breast cancer were retrospectively enrolled from three institutions and were split into a training cohort (242 patients), an internal validation cohort (197 patients), and two external test cohorts (212 and 150 patients). Three imaging signatures were constructed from the serial ultrasonographic features before (pretreatment signature), during the first-second cycle of (early-stage treatment signature), and after (posttreatment signature) NAC based on autosegmentation by U-net. The SUAS was constructed by subsequently integrating the pre, early-stage, and posttreatment signatures, and the incremental performance was analysed. RESULTS The SUAS yielded a favourable performance in predicting pCR, with areas under the receiver operating characteristic curve (AUCs) of 0.927 [95% confidence interval (CI) 0.891-0.963] and 0.914 (95% CI 0.853-0.976), compared with those of the clinicopathological prediction model [0.734 (95% CI 0.665-0.804) and 0.610 (95% CI 0.504-0.716)], and radiologist interpretation [0.632 (95% CI 0.570-0.693) and 0.724 (95% CI 0.644-0.804)] in the external test cohorts. Furthermore, similar results were also observed in the early-stage treatment of NAC [AUC 0.874 (0.793-0.955)-0.897 (0.851-0.943) in the external test cohorts]. CONCLUSIONS We demonstrate that autosegmentation-based SAUS integrating serial ultrasonographic features throughout NAC can predict pCR with favourable performance, which can facilitate individualized treatment strategies.
Collapse
Affiliation(s)
- Lei Wu
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China ,grid.413352.20000 0004 1760 3705Guangdong Cardiovascular Institute, 106 Zhongshan 2nd Road, Guangzhou, 510080 China
| | - Weitao Ye
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Yu Liu
- grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China ,grid.410643.4Department of Ultrasound, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China
| | - Dong Chen
- grid.452826.fDepartment of Medical Ultrasound, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Yuxiang Wang
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Yanfen Cui
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Zhenhui Li
- grid.452826.fDepartment of Radiology, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Pinxiong Li
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Zhen Li
- grid.452826.fDepartment of 3rd Breast Surgery, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Zaiyi Liu
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Min Liu
- grid.488530.20000 0004 1803 6191Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 China
| | - Changhong Liang
- grid.410643.4Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, 510080 China ,grid.410643.4Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Xiaotang Yang
- grid.263452.40000 0004 1798 4018Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013 China
| | - Yu Xie
- grid.452826.fDepartment of Radiology, Yunnan Cancer Hospital, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 China
| | - Ying Wang
- grid.470124.4Department of Medical Ultrasonics, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou, 510120 China
| |
Collapse
|
36
|
Miao R, Toth R, Zhou Y, Madabhushi A, Janowczyk A. Quick Annotator: an open-source digital pathology based rapid image annotation tool. J Pathol Clin Res 2021; 7:542-547. [PMID: 34288586 PMCID: PMC8503896 DOI: 10.1002/cjp2.229] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/16/2021] [Accepted: 05/22/2021] [Indexed: 11/23/2022]
Abstract
Image-based biomarker discovery typically requires accurate segmentation of histologic structures (e.g. cell nuclei, tubules, and epithelial regions) in digital pathology whole slide images (WSIs). Unfortunately, annotating each structure of interest is laborious and often intractable even in moderately sized cohorts. Here, we present an open-source tool, Quick Annotator (QA), designed to improve annotation efficiency of histologic structures by orders of magnitude. While the user annotates regions of interest (ROIs) via an intuitive web interface, a deep learning (DL) model is concurrently optimized using these annotations and applied to the ROI. The user iteratively reviews DL results to either (1) accept accurately annotated regions or (2) correct erroneously segmented structures to improve subsequent model suggestions, before transitioning to other ROIs. We demonstrate the effectiveness of QA over comparable manual efforts via three use cases. These include annotating (1) 337,386 nuclei in 5 pancreatic WSIs, (2) 5,692 tubules in 10 colorectal WSIs, and (3) 14,187 regions of epithelium in 10 breast WSIs. Efficiency gains in terms of annotations per second of 102×, 9×, and 39× were, respectively, witnessed while retaining f-scores >0.95, suggesting that QA may be a valuable tool for efficiently fully annotating WSIs employed in downstream biomarker studies.
Collapse
Affiliation(s)
- Runtian Miao
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | | | - Yu Zhou
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
| | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Veterans Administration Medical CenterClevelandOHUSA
| | - Andrew Janowczyk
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Precision Oncology CenterLausanne University HospitalLausanneSwitzerland
| |
Collapse
|
37
|
Xu Z, Li Y, Wang Y, Zhang S, Huang Y, Yao S, Han C, Pan X, Shi Z, Mao Y, Xu Y, Huang X, Lin H, Chen X, Liang C, Li Z, Zhao K, Zhang Q, Liu Z. A deep learning quantified stroma-immune score to predict survival of patients with stage II-III colorectal cancer. Cancer Cell Int 2021; 21:585. [PMID: 34717647 PMCID: PMC8557607 DOI: 10.1186/s12935-021-02297-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background Profound heterogeneity in prognosis has been observed in colorectal cancer (CRC) patients with intermediate levels of disease (stage II–III), advocating the identification of valuable biomarkers that could improve the prognostic stratification. This study aims to develop a deep learning-based pipeline for fully automatic quantification of immune infiltration within the stroma region on immunohistochemical (IHC) whole-slide images (WSIs) and further analyze its prognostic value in CRC. Methods Patients from two independent cohorts were divided into three groups: the development group (N = 200), the internal (N = 134), and the external validation group (N = 90). We trained a convolutional neural network for tissue classification of CD3 and CD8 stained WSIs. A scoring system, named stroma-immune score, was established by quantifying the density of CD3+ and CD8+ T-cells infiltration in the stroma region. Results Patients with higher stroma-immune scores had much longer survival. In the development group, 5-year survival rates of the low and high scores were 55.7% and 80.8% (hazard ratio [HR] for high vs. low 0.39, 95% confidence interval [CI] 0.24–0.63, P < 0.001). These results were confirmed in the internal and external validation groups with 5-year survival rates of low and high scores were 57.1% and 78.8%, 63.9% and 88.9%, respectively (internal: HR for high vs. low 0.49, 95% CI 0.28–0.88, P = 0.017; external: HR for high vs. low 0.35, 95% CI 0.15–0.83, P = 0.018). The combination of stroma-immune score and tumor-node-metastasis (TNM) stage showed better discrimination ability for survival prediction than using the TNM stage alone. Conclusions We proposed a stroma-immune score via a deep learning-based pipeline to quantify CD3+ and CD8+ T-cells densities within the stroma region on WSIs of CRC and further predict survival. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02297-w.
Collapse
Affiliation(s)
- Zeyan Xu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.,School of Medicine, South China University of Technology, Panyu District, Guangzhou, 510006, China
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yingyi Wang
- Department of Radiology, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, 519000, China
| | - Shenyan Zhang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Chu Han
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xipeng Pan
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Zhenwei Shi
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yun Mao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yao Xu
- School of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xiaomei Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510080, China
| | - Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.,School of Medicine, South China University of Technology, Panyu District, Guangzhou, 510006, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Zhenhui Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China.
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China. .,School of Medicine, South China University of Technology, Panyu District, Guangzhou, 510006, China.
| |
Collapse
|
38
|
Mi H, Bivalacqua TJ, Kates M, Seiler R, Black PC, Popel AS, Baras AS. Predictive models of response to neoadjuvant chemotherapy in muscle-invasive bladder cancer using nuclear morphology and tissue architecture. Cell Rep Med 2021; 2:100382. [PMID: 34622225 PMCID: PMC8484511 DOI: 10.1016/j.xcrm.2021.100382] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/30/2021] [Accepted: 07/29/2021] [Indexed: 12/20/2022]
Abstract
Characterizing likelihood of response to neoadjuvant chemotherapy (NAC) in muscle-invasive bladder cancer (MIBC) is an important yet unmet challenge. In this study, a machine-learning framework is developed using imaging of biopsy pathology specimens to generate models of likelihood of NAC response. Developed using cross-validation (evaluable N = 66) and an independent validation cohort (evaluable N = 56), our models achieve promising results (65%-73% accuracy). Interestingly, one model-using features derived from hematoxylin and eosin (H&E)-stained tissues in conjunction with clinico-demographic features-is able to stratify the cohort into likely responders in cross-validation and the validation cohort (response rate of 65% for predicted responder compared with the 41% baseline response rate in the validation cohort). The results suggest that computational approaches applied to routine pathology specimens of MIBC can capture differences between responders and non-responders to NAC and should therefore be considered in the future design of precision oncology for MIBC.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Trinity J. Bivalacqua
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Max Kates
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roland Seiler
- Department of Urology, University Hospital Bern, Bern, Switzerland
| | - Peter C. Black
- Department of Urologic Sciences, University of British Columbia Faculty of Medicine, Vancouver, BC, Canada
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Alexander S. Baras
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Lee K, Lockhart JH, Xie M, Chaudhary R, Slebos RJC, Flores ER, Chung CH, Tan AC. Deep Learning of Histopathology Images at the Single Cell Level. Front Artif Intell 2021; 4:754641. [PMID: 34568816 PMCID: PMC8461055 DOI: 10.3389/frai.2021.754641] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor immune microenvironment (TIME) encompasses many heterogeneous cell types that engage in extensive crosstalk among the cancer, immune, and stromal components. The spatial organization of these different cell types in TIME could be used as biomarkers for predicting drug responses, prognosis and metastasis. Recently, deep learning approaches have been widely used for digital histopathology images for cancer diagnoses and prognoses. Furthermore, some recent approaches have attempted to integrate spatial and molecular omics data to better characterize the TIME. In this review we focus on machine learning-based digital histopathology image analysis methods for characterizing tumor ecosystem. In this review, we will consider three different scales of histopathological analyses that machine learning can operate within: whole slide image (WSI)-level, region of interest (ROI)-level, and cell-level. We will systematically review the various machine learning methods in these three scales with a focus on cell-level analysis. We will provide a perspective of workflow on generating cell-level training data sets using immunohistochemistry markers to "weakly-label" the cell types. We will describe some common steps in the workflow of preparing the data, as well as some limitations of this approach. Finally, we will discuss future opportunities of integrating molecular omics data with digital histopathology images for characterizing tumor ecosystem.
Collapse
Affiliation(s)
- Kyubum Lee
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - John H. Lockhart
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Mengyu Xie
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Ritu Chaudhary
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Robbert J. C. Slebos
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Elsa R. Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Christine H. Chung
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
40
|
Xu H, Cong F, Hwang TH. Machine Learning and Artificial Intelligence-driven Spatial Analysis of the Tumor Immune Microenvironment in Pathology Slides. Eur Urol Focus 2021; 7:706-709. [PMID: 34353733 DOI: 10.1016/j.euf.2021.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/21/2021] [Indexed: 12/27/2022]
Abstract
A better understanding of the tumor immune microenvironment (TIME) could lead to accurate diagnosis, prognosis, and treatment stratification. Although molecular analyses at the tissue and/or single cell level could reveal the cellular status of the tumor microenvironment, these approaches lack information related to spatial-level cellular distribution, co-organization, and cell-cell interaction in the TIME. With the emergence of computational pathology coupled with machine learning (ML) and artificial intelligence (AI), ML- and AI-driven spatial TIME analyses of pathology images could revolutionize our understanding of the highly heterogeneous and complex molecular architecture of the TIME. In this review we highlight recent studies on spatial TIME analysis of pathology slides using state-of-the-art ML and AI algorithms. PATIENT SUMMARY: This mini-review reports recent advances in machine learning and artificial intelligence for spatial analysis of the tumor immune microenvironment in pathology slides. This information can help in understanding the spatial heterogeneity and organization of cells in patient tumors.
Collapse
Affiliation(s)
- Hongming Xu
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Fengyu Cong
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Tae Hyun Hwang
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
41
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
42
|
Klein C, Zeng Q, Arbaretaz F, Devêvre E, Calderaro J, Lomenie N, Maiuri MC. Artificial Intelligence for solid tumor diagnosis in digital pathology. Br J Pharmacol 2021; 178:4291-4315. [PMID: 34302297 DOI: 10.1111/bph.15633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 11/30/2022] Open
Abstract
Tumor diagnosis relies on the visual examination of histological slides by pathologists through a microscope eyepiece. Digital pathology, the digitalization of histological slides at high magnification with slides scanners, has raised the opportunity to extract quantitative information thanks to image analysis. In the last decade, medical image analysis has made exceptional progress due to the development of artificial intelligence (AI) algorithms. AI has been successfully used in the field of medical imaging and more recently in digital pathology. The feasibility and usefulness of AI assisted pathology tasks have been demonstrated in the very last years and we can expect those developments to be applied on routine histopathology in the future. In this review, we will describe and illustrate this technique and present the most recent applications in the field of tumor histopathology.
Collapse
Affiliation(s)
- Christophe Klein
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Qinghe Zeng
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France.,Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Floriane Arbaretaz
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Estelle Devêvre
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Julien Calderaro
- Département de pathologie, Hôpital Henri Mondor, Créteil, France
| | - Nicolas Lomenie
- Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Maria Chiara Maiuri
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
43
|
Characterizing the ecological and evolutionary dynamics of cancer. Nat Genet 2020; 52:759-767. [DOI: 10.1038/s41588-020-0668-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
|
44
|
Peng Y, Chu Y, Chen Z, Zhou W, Wan S, Xiao Y, Zhang Y, Li J. Combining texture features of whole slide images improves prognostic prediction of recurrence-free survival for cutaneous melanoma patients. World J Surg Oncol 2020; 18:130. [PMID: 32546168 PMCID: PMC7298832 DOI: 10.1186/s12957-020-01909-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/08/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Accurate prediction of recurrence-free survival (RFS) is important for the prognosis of cutaneous melanoma patients. The image-based pathological examination remains as the gold standard for diagnosis. It is of clinical interest to account for computer-aided processing of pathology image when performing prognostic analysis. METHODS We enrolled in this study a total of 152 patients from TCGA-SKCM (The Cancer Genome Atlas Skin Cutaneous Melanoma project) with complete information in recurrence-related survival time, baseline variables (clinicopathologic variables, mutation status of BRAF and NRAS genes), gene expression data, and whole slide image (WSI) features. We preprocessed WSI to segment global or nucleus areas, and extracted 3 types of texture features from each region. We performed cross validation and used multiple evaluation metrics including C-index and time-dependent AUC to determine the best model of predicting recurrence events. We further performed differential gene expression analysis between the higher and lower-risk groups within AJCC pathologic tumor stage III patients to explore the underlying molecular mechanisms driving risk stratification. RESULTS The model combining baseline variables and WSI features had the best performance among models with any other types of data integration. The prognostic risk score generated by this model could provide a higher-resolution risk stratification within pathologically defined subgroups. We found the selected image features captured important immune-related variations, such as the aberration of expression in T cell activation and proliferation gene sets, and therefore contributed to the improved prediction. CONCLUSIONS Our study provided a prognostic model based on the combination of baseline variables and computer-processed WSI features. This model provided more accurate prediction than models based on other types of data combination in recurrence-free survival analysis. TRIAL REGISTRATION This study was based on public open data from TCGA and hence the study objects were retrospectively registered.
Collapse
Affiliation(s)
- Yanbin Peng
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Yunfeng Chu
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Zhong Chen
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Wen Zhou
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Shengxiang Wan
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Yingfeng Xiao
- Department of Microsurgery, Peking University Shenzhen Hospital, Futian District, Shenzhen, 518036 China
| | - Youlong Zhang
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057 China
| | - Jialu Li
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057 China
| |
Collapse
|