1
|
Derben J, Oeruem M, Blasberg C, Hattesohl A, Jank P, Kalder M, Denkert C, Westhoff CC. Prognostic Impact of Immunophenotypic Variation in Subcapsular Sinus Endothelium of Sentinel Lymph Nodes in Invasive Breast Carcinoma. Breast Care (Basel) 2025; 20:75-87. [PMID: 40256677 PMCID: PMC12005704 DOI: 10.1159/000543600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/14/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Several studies demonstrated the de novo formation of lymphatic vessels in tumor-draining lymph nodes (LNs), partly preceding lymphatic metastases. This "lymphovascular niche" supposedly facilitates survival of metastatic tumor cells. This study aims at evaluating the previously observed immunophenotypic variations of subcapsular endothelial cells (SECs) in a larger cohort by software-assisted image analysis. Methods Suitable cases with sentinel-LN (SLN) of invasive breast cancer were identified in the Institute of Pathology, corresponding data were extracted. LN of 231 patients were stained for HE, D2-40, CD31, and Prox1. QuPath software was used for assessing the immunohistochemical stained area of endothelial cells of the subcapsular sinus. The Cutoff Finder web application was used for identification of the best cutoff for continuous parameters according to overall survival (OS). Collected data were statistically evaluated for available data. Results A larger area of CD31-positive SEC was significantly associated with worse OS (p = 0.001), as was a higher proportion of D2-40-stained subcapsular sinus (p = 0.045). Larger area of D2-40-/CD31-/Prox1-positive SEC and higher proportion of D2-40 stained subcapsular sinus were independent marker for worse OS in multivariate analysis in the whole cohort, for D2-40- and CD31-positive SECs as well as higher proportion of D2-40-stained sinus including nodal-negative status, respectively. Conclusion QuPath-assisted evaluation of immunophenotypic variation in subcapsular sinus endothelium in SLN essentially confirmed and extended our previous findings. Larger positive area of D2-40- and CD31-positive SECs emerged as a strong independent negative prognostic factor, even before evident nodal metastasis. The potential function of alterations in D2-40-/CD31-expression in SECs has yet to be elucidated.
Collapse
Affiliation(s)
- Jonas Derben
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| | - Markus Oeruem
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
- Thoraxklinik Heidelberg, Pneumology and Respiratory Medicine, Heidelberg, Germany
| | - Carolin Blasberg
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| | - Akira Hattesohl
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| | - Paul Jank
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| | - Matthias Kalder
- Department of Gynecology and Obstetrics, Breast Center Regio, Philipps University of Marburg and University Hospital Giessen and Marburg GmbH, Marburg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| | - Christina C. Westhoff
- Institute of Pathology, Philipps University of Marburg and University Hospital Marburg (UKGM) – Universitaetsklinikum Marburg, Marburg, Germany
| |
Collapse
|
2
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
3
|
Llewellyn A, Barrow-McGee R, Stevenson J, Gore J, Naidoo K. Protocol for perfusing human axillary lymph nodes ex vivo to study structure and function in real time. STAR Protoc 2025; 6:103624. [PMID: 39913290 PMCID: PMC11848449 DOI: 10.1016/j.xpro.2025.103624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/01/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Lymph nodes regulate immunity and maintain fluid balance in health and disease. Here, we present a protocol that uses normothermic perfusion to sustain patient-derived lymph nodes ex vivo for up to 24 h to study their structure and function. We describe steps for setting up both thermoregulatory and perfusion circuits, cannulating human lymph nodes, and perfusion. This protocol can be used to study how human lymph nodes change in cancer and other diseases, and/or in response to perturbations, including drugs. For complete details on the use and execution of this protocol, please refer to Barrow-McGee et al.1.
Collapse
Affiliation(s)
- Amy Llewellyn
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Rachel Barrow-McGee
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Julia Stevenson
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | - Jasmine Gore
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Kalnisha Naidoo
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK; Department of Cellular Pathology, King's College Hospital, London, UK.
| |
Collapse
|
4
|
Wang Q, Zhao F, Zhang H, Chu T, Wang Q, Pan X, Chen Y, Zhou H, Zheng T, Li Z, Lin F, Xie H, Ma H, Liu L, Zhang L, Li Q, Wang W, Dai Y, Tang R, Wang J, Yang P, Mao N. Deep learning-based multi-task prediction of response to neoadjuvant chemotherapy using multiscale whole slide images in breast cancer: A multicenter study. Chin J Cancer Res 2025; 37:28-47. [PMID: 40078559 PMCID: PMC11893347 DOI: 10.21147/j.issn.1000-9604.2025.01.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Early predicting response before neoadjuvant chemotherapy (NAC) is crucial for personalized treatment plans for locally advanced breast cancer patients. We aim to develop a multi-task model using multiscale whole slide images (WSIs) features to predict the response to breast cancer NAC more finely. Methods This work collected 1,670 whole slide images for training and validation sets, internal testing sets, external testing sets, and prospective testing sets of the weakly-supervised deep learning-based multi-task model (DLMM) in predicting treatment response and pCR to NAC. Our approach models two-by-two feature interactions across scales by employing concatenate fusion of single-scale feature representations, and controls the expressiveness of each representation via a gating-based attention mechanism. Results In the retrospective analysis, DLMM exhibited excellent predictive performance for the prediction of treatment response, with area under the receiver operating characteristic curves (AUCs) of 0.869 [95% confidence interval (95% CI): 0.806-0.933] in the internal testing set and 0.841 (95% CI: 0.814-0.867) in the external testing sets. For the pCR prediction task, DLMM reached AUCs of 0.865 (95% CI: 0.763-0.964) in the internal testing and 0.821 (95% CI: 0.763-0.878) in the pooled external testing set. In the prospective testing study, DLMM also demonstrated favorable predictive performance, with AUCs of 0.829 (95% CI: 0.754-0.903) and 0.821 (95% CI: 0.692-0.949) in treatment response and pCR prediction, respectively. DLMM significantly outperformed the baseline models in all testing sets (P<0.05). Heatmaps were employed to interpret the decision-making basis of the model. Furthermore, it was discovered that high DLMM scores were associated with immune-related pathways and cells in the microenvironment during biological basis exploration. Conclusions The DLMM represents a valuable tool that aids clinicians in selecting personalized treatment strategies for breast cancer patients.
Collapse
Affiliation(s)
- Qin Wang
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Feng Zhao
- School of Computer Science and Technology, Shandong Technology and Business University, Yantai 264005, China
| | - Haicheng Zhang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tongpeng Chu
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Qi Wang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Xipeng Pan
- School of Computer Science and Information Security, Guilin University of Electronic Technology, Guilin 541004, China
| | - Yuqian Chen
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Zhou
- School of Information and Electronic Engineering, Shandong Technology and Business University, Yantai 264005, China
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Tiantian Zheng
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Ziyin Li
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- School of Medical Imaging, Binzhou Medical University, Yantai 264003, China
| | - Fan Lin
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Haizhu Xie
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Heng Ma
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital, the Second Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China
| | - Lina Zhang
- Department of Radiology, the First Affiliated Hospital of China Medical University, Shenyang 400042, China
| | - Qin Li
- Department of Radiology, Weifang Hospital of Traditional Chinese Medicine, Weifang 262600, China
| | - Weiwei Wang
- Department of Medical Imaging, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Yi Dai
- Department of Radiology, the Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Ruijun Tang
- Department of Pathology, Guilin Traditional Chinese Medicine Hospital, Guilin 541002, China
| | - Jigang Wang
- Department of Pathology, the Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Ping Yang
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Ning Mao
- Big Data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
- Shandong Provincial Key Medical and Health Laboratory of Intelligent Diagnosis and Treatment for Women’s Diseases, Yantai Yuhuangding Hospital, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| |
Collapse
|
5
|
Budginaite E, Magee DR, Kloft M, Woodruff HC, Grabsch HI. Computational methods for metastasis detection in lymph nodes and characterization of the metastasis-free lymph node microarchitecture: A systematic-narrative hybrid review. J Pathol Inform 2024; 15:100367. [PMID: 38455864 PMCID: PMC10918266 DOI: 10.1016/j.jpi.2024.100367] [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: 12/28/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 03/09/2024] Open
Abstract
Background Histological examination of tumor draining lymph nodes (LNs) plays a vital role in cancer staging and prognostication. However, as soon as a LN is classed as metastasis-free, no further investigation will be performed and thus, potentially clinically relevant information detectable in tumor-free LNs is currently not captured. Objective To systematically study and critically assess methods for the analysis of digitized histological LN images described in published research. Methods A systematic search was conducted in several public databases up to December 2023 using relevant search terms. Studies using brightfield light microscopy images of hematoxylin and eosin or immunohistochemically stained LN tissue sections aiming to detect and/or segment LNs, their compartments or metastatic tumor using artificial intelligence (AI) were included. Dataset, AI methodology, cancer type, and study objective were compared between articles. Results A total of 7201 articles were collected and 73 articles remained for detailed analyses after article screening. Of the remaining articles, 86% aimed at LN metastasis identification, 8% aimed at LN compartment segmentation, and remaining focused on LN contouring. Furthermore, 78% of articles used patch classification and 22% used pixel segmentation models for analyses. Five out of six studies (83%) of metastasis-free LNs were performed on publicly unavailable datasets, making quantitative article comparison impossible. Conclusions Multi-scale models mimicking multiple microscopy zooms show promise for computational LN analysis. Large-scale datasets are needed to establish the clinical relevance of analyzing metastasis-free LN in detail. Further research is needed to identify clinically interpretable metrics for LN compartment characterization.
Collapse
Affiliation(s)
- Elzbieta Budginaite
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | - Maximilian Kloft
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | - Henry C. Woodruff
- Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Heike I. Grabsch
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
Li X. Deciphering cell to cell spatial relationship for pathology images using SpatialQPFs. Sci Rep 2024; 14:29585. [PMID: 39609630 PMCID: PMC11605059 DOI: 10.1038/s41598-024-81383-1] [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: 06/18/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024] Open
Abstract
Understanding spatial dynamics within tissue microenvironments is crucial for deciphering cellular interactions and molecular signaling in living systems. These spatial characteristics govern cell distribution, extracellular matrix components, and signaling molecules, influencing local biochemical and biophysical conditions. Despite significant progress in analyzing digital pathology images, current methods for capturing spatial relationships are limited. They often rely on specific spatial features that only partially describe the complex spatial distributions of cells and are frequently tied to particular outcomes within predefined model frameworks. Furthermore, these methods are typically limited to field of view analysis, which restricts their capacity to capture spatial patterns across whole-slide images, thereby limiting their ability to fully address the complexities of tissue architecture. To address these limitations, we present SpatialQPFs (Spatial Quantitative Pathology Features), an R package designed to extract interpretable spatial features from cell imaging data using spatial statistical methodologies. Leveraging segmented cell information, our package offers a comprehensive toolkit for applying a range of spatial statistical methods within a stochastic process framework, including analyses of point process data, areal data, and geostatistical data. By decoupling feature extraction from specific outcome models, SpatialQPFs enables thorough large-scale spatial analyses applicable across diverse clinical and biological contexts. This approach enhances the depth and accuracy of spatial insights derived from tissue data, empowering researchers to conduct comprehensive spatial analyses efficiently and reproducibly. By providing a flexible and robust framework for spatial feature extraction, SpatialQPFs facilitates advanced spatial analyses, paving the way for new discoveries in tissue biology and pathology. SpatialQPFs code and documentation are publicly available at https://github.com/Genentech/SpatialQPFs .
Collapse
Affiliation(s)
- Xiao Li
- Computational Science and Informatics, Roche Diagnostics Solutions, Santa Clara, CA, 95050, USA.
| |
Collapse
|
7
|
Baharun NB, Adam A, Zailani MAH, Rajpoot NM, Xu Q, Zin RRM. Automated scoring methods for quantitative interpretation of Tumour infiltrating lymphocytes (TILs) in breast cancer: a systematic review. BMC Cancer 2024; 24:1202. [PMID: 39350098 PMCID: PMC11440723 DOI: 10.1186/s12885-024-12962-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Tumour microenvironment (TME) of breast cancer mainly comprises malignant, stromal, immune, and tumour infiltrating lymphocyte (TILs). Assessment of TILs is crucial for determining the disease's prognosis. Manual TIL assessments are hampered by multiple limitations, including low precision, poor inter-observer reproducibility, and time consumption. In response to these challenges, automated scoring emerges as a promising approach. The aim of this systematic review is to assess the evidence on the approaches and performance of automated scoring methods for TILs assessment in breast cancer. This review presents a comprehensive compilation of studies related to automated scoring of TILs, sourced from four databases (Web of Science, Scopus, Science Direct, and PubMed), employing three primary keywords (artificial intelligence, breast cancer, and tumor-infiltrating lymphocytes). The PICOS framework was employed for study eligibility, and reporting adhered to the PRISMA guidelines. The initial search yielded a total of 1910 articles. Following screening and examination, 27 studies met the inclusion criteria and data were extracted for the review. The findings indicate a concentration of studies on automated TILs assessment in developed countries, specifically the United States and the United Kingdom. From the analysis, a combination of sematic segmentation and object detection (n = 10, 37%) and convolutional neural network (CNN) (n = 11, 41%), become the most frequent automated task and ML approaches applied for model development respectively. All models developed their own ground truth datasets for training and validation, and 59% of the studies assessed the prognostic value of TILs. In conclusion, this analysis contends that automated scoring methods for TILs assessment of breast cancer show significant promise for commodification and application within clinical settings.
Collapse
Affiliation(s)
- Nurkhairul Bariyah Baharun
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia.
- Department of Medical Diagnostic, Faculty of Health Sciences, Universiti Selangor, Jalan Zirkon A7/7, Seksyen 7, Shah Alam, Selangor, 40000, Malaysia.
| | - Afzan Adam
- Centre for Artificial Intelligence Technology (CAIT), Faculty of Information Science & Technology, The National University of Malaysia, Bangi, Selangor, 43600, Malaysia
| | - Mohamed Afiq Hidayat Zailani
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia
- Department of Pathology and Forensic Pathology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Malaysia
| | - Nasir M Rajpoot
- Department of Computer Science, University of Warwick, 6 Lord Bhattacharyya Way, Coventry, CV4 7EZ, UK
| | - Qiaoyi Xu
- Centre for Artificial Intelligence Technology (CAIT), Faculty of Information Science & Technology, The National University of Malaysia, Bangi, Selangor, 43600, Malaysia
| | - Reena Rahayu Md Zin
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia
| |
Collapse
|
8
|
Jiang B, Bao L, He S, Chen X, Jin Z, Ye Y. Deep learning applications in breast cancer histopathological imaging: diagnosis, treatment, and prognosis. Breast Cancer Res 2024; 26:137. [PMID: 39304962 DOI: 10.1186/s13058-024-01895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Breast cancer is the most common malignant tumor among women worldwide and remains one of the leading causes of death among women. Its incidence and mortality rates are continuously rising. In recent years, with the rapid advancement of deep learning (DL) technology, DL has demonstrated significant potential in breast cancer diagnosis, prognosis evaluation, and treatment response prediction. This paper reviews relevant research progress and applies DL models to image enhancement, segmentation, and classification based on large-scale datasets from TCGA and multiple centers. We employed foundational models such as ResNet50, Transformer, and Hover-net to investigate the performance of DL models in breast cancer diagnosis, treatment, and prognosis prediction. The results indicate that DL techniques have significantly improved diagnostic accuracy and efficiency, particularly in predicting breast cancer metastasis and clinical prognosis. Furthermore, the study emphasizes the crucial role of robust databases in developing highly generalizable models. Future research will focus on addressing challenges related to data management, model interpretability, and regulatory compliance, ultimately aiming to provide more precise clinical treatment and prognostic evaluation programs for breast cancer patients.
Collapse
Affiliation(s)
- Bitao Jiang
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China.
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China.
| | - Lingling Bao
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Songqin He
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Xiao Chen
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China
| | - Zhihui Jin
- Department of Hematology and Oncology, Beilun District People's Hospital, Ningbo, 315800, China
- Department of Hematology and Oncology, Beilun Branch of the First Affiliated Hospital of Zhejiang University, Ningbo, 315800, China
| | - Yingquan Ye
- Department of Oncology, The 906th Hospital of the Joint Logistics Force of the Chinese People's Liberation Army, Ningbo, 315100, China.
| |
Collapse
|
9
|
Thapa R, Moglad E, Afzal M, Gupta G, Bhat AA, Almalki WH, Kazmi I, Alzarea SI, Pant K, Ali H, Paudel KR, Dureja H, Singh TG, Singh SK, Dua K. ncRNAs and their impact on dopaminergic neurons: Autophagy pathways in Parkinson's disease. Ageing Res Rev 2024; 98:102327. [PMID: 38734148 DOI: 10.1016/j.arr.2024.102327] [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: 02/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Parkinson's Disease (PD) is a complex neurological illness that causes severe motor and non-motor symptoms due to a gradual loss of dopaminergic neurons in the substantia nigra. The aetiology of PD is influenced by a variety of genetic, environmental, and cellular variables. One important aspect of this pathophysiology is autophagy, a crucial cellular homeostasis process that breaks down and recycles cytoplasmic components. Recent advances in genomic technologies have unravelled a significant impact of ncRNAs on the regulation of autophagy pathways, thereby implicating their roles in PD onset and progression. They are members of a family of RNAs that include miRNAs, circRNA and lncRNAs that have been shown to play novel pleiotropic functions in the pathogenesis of PD by modulating the expression of genes linked to autophagic activities and dopaminergic neuron survival. This review aims to integrate the current genetic paradigms with the therapeutic prospect of autophagy-associated ncRNAs in PD. By synthesizing the findings of recent genetic studies, we underscore the importance of ncRNAs in the regulation of autophagy, how they are dysregulated in PD, and how they represent novel dimensions for therapeutic intervention. The therapeutic promise of targeting ncRNAs in PD is discussed, including the barriers that need to be overcome and future directions that must be embraced to funnel these ncRNA molecules for the treatment and management of PD.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
10
|
Leon-Ferre RA, Whitaker KR, Suman VJ, Hoskin T, Giridhar KV, Moore RM, Al-Jarrad A, McLaughlin SA, Northfelt DW, Hunt KN, Conners AL, Moyer A, Carter JM, Kalari K, Weinshilboum R, Wang L, Ingle JN, Knutson KL, Ansell SM, Boughey JC, Goetz MP, Villasboas JC. Pre-treatment peripheral blood immunophenotyping and response to neoadjuvant chemotherapy in operable breast cancer. Breast Cancer Res 2024; 26:97. [PMID: 38858721 PMCID: PMC11165781 DOI: 10.1186/s13058-024-01848-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Tumor immune infiltration and peripheral blood immune signatures have prognostic and predictive value in breast cancer. Whether distinct peripheral blood immune phenotypes are associated with response to neoadjuvant chemotherapy (NAC) remains understudied. METHODS Peripheral blood mononuclear cells from 126 breast cancer patients enrolled in a prospective clinical trial (NCT02022202) were analyzed using Cytometry by time-of-flight with a panel of 29 immune cell surface protein markers. Kruskal-Wallis tests or Wilcoxon rank-sum tests were used to evaluate differences in immune cell subpopulations according to breast cancer subtype and response to NAC. RESULTS There were 122 evaluable samples: 47 (38.5%) from patients with hormone receptor-positive, 39 (32%) triple-negative (TNBC), and 36 (29.5%) HER2-positive breast cancer. The relative abundances of pre-treatment peripheral blood T, B, myeloid, NK, and unclassified cells did not differ according to breast cancer subtype. In TNBC, higher pre-treatment myeloid cells were associated with lower pathologic complete response (pCR) rates. In hormone receptor-positive breast cancer, lower pre-treatment CD8 + naïve and CD4 + effector memory cells re-expressing CD45RA (TEMRA) T cells were associated with more extensive residual disease after NAC. In HER2 + breast cancer, the peripheral blood immune phenotype did not differ according to NAC response. CONCLUSIONS Pre-treatment peripheral blood immune cell populations (myeloid in TNBC; CD8 + naïve T cells and CD4 + TEMRA cells in luminal breast cancer) were associated with response to NAC in early-stage TNBC and hormone receptor-positive breast cancers, but not in HER2 + breast cancer. TRIAL REGISTRATION NCT02022202 . Registered 20 December 2013.
Collapse
Affiliation(s)
| | | | - Vera J Suman
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Tanya Hoskin
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Raymond M Moore
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Katie N Hunt
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Ann Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Krishna Kalari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Liewei Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | |
Collapse
|
11
|
Zhang C, Xu J, Tang R, Yang J, Wang W, Yu X, Shi S. Novel research and future prospects of artificial intelligence in cancer diagnosis and treatment. J Hematol Oncol 2023; 16:114. [PMID: 38012673 PMCID: PMC10680201 DOI: 10.1186/s13045-023-01514-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Research into the potential benefits of artificial intelligence for comprehending the intricate biology of cancer has grown as a result of the widespread use of deep learning and machine learning in the healthcare sector and the availability of highly specialized cancer datasets. Here, we review new artificial intelligence approaches and how they are being used in oncology. We describe how artificial intelligence might be used in the detection, prognosis, and administration of cancer treatments and introduce the use of the latest large language models such as ChatGPT in oncology clinics. We highlight artificial intelligence applications for omics data types, and we offer perspectives on how the various data types might be combined to create decision-support tools. We also evaluate the present constraints and challenges to applying artificial intelligence in precision oncology. Finally, we discuss how current challenges may be surmounted to make artificial intelligence useful in clinical settings in the future.
Collapse
Affiliation(s)
- Chaoyi Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianhui Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
12
|
Verghese G, Lennerz JK, Ruta D, Ng W, Thavaraj S, Siziopikou KP, Naidoo T, Rane S, Salgado R, Pinder SE, Grigoriadis A. Computational pathology in cancer diagnosis, prognosis, and prediction - present day and prospects. J Pathol 2023; 260:551-563. [PMID: 37580849 PMCID: PMC10785705 DOI: 10.1002/path.6163] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/12/2023] [Accepted: 06/17/2023] [Indexed: 08/16/2023]
Abstract
Computational pathology refers to applying deep learning techniques and algorithms to analyse and interpret histopathology images. Advances in artificial intelligence (AI) have led to an explosion in innovation in computational pathology, ranging from the prospect of automation of routine diagnostic tasks to the discovery of new prognostic and predictive biomarkers from tissue morphology. Despite the promising potential of computational pathology, its integration in clinical settings has been limited by a range of obstacles including operational, technical, regulatory, ethical, financial, and cultural challenges. Here, we focus on the pathologists' perspective of computational pathology: we map its current translational research landscape, evaluate its clinical utility, and address the more common challenges slowing clinical adoption and implementation. We conclude by describing contemporary approaches to drive forward these techniques. © 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)
- Gregory Verghese
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- The Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Department of PathologyMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
| | - Danny Ruta
- Guy's CancerGuy's and St Thomas’ NHS Foundation TrustLondonUK
| | - Wen Ng
- Department of Cellular PathologyGuy's and St Thomas NHS Foundation TrustLondonUK
| | - Selvam Thavaraj
- Head & Neck PathologyGuy's and St Thomas NHS Foundation TrustLondonUK
- Centre for Clinical, Oral & Translational Science, Faculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonLondonUK
| | - Kalliopi P Siziopikou
- Department of Pathology, Section of Breast PathologyNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Threnesan Naidoo
- Department of Laboratory Medicine and Pathology, Walter Sisulu University, Mthatha, Eastern CapeSouth Africa and Africa Health Research InstituteDurbanSouth Africa
| | - Swapnil Rane
- Department of PathologyTata Memorial Centre – ACTRECHBNINavi MumbaiIndia
- Computational Pathology, AI & Imaging LaboratoryTata Memorial Centre – ACTREC, HBNINavi MumbaiIndia
| | - Roberto Salgado
- Department of PathologyGZA–ZNA ZiekenhuizenAntwerpBelgium
- Division of ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Sarah E Pinder
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- Department of Cellular PathologyGuy's and St Thomas NHS Foundation TrustLondonUK
| | - Anita Grigoriadis
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- The Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
13
|
Page DB, Broeckx G, Jahangir CA, Verbandt S, Gupta RR, Thagaard J, Khiroya R, Kos Z, Abduljabbar K, Acosta Haab G, Acs B, Akturk G, Almeida JS, Alvarado-Cabrero I, Azmoudeh-Ardalan F, Badve S, Baharun NB, Bellolio ER, Bheemaraju V, Blenman KRM, Botinelly Mendonça Fujimoto L, Bouchmaa N, Burgues O, Cheang MCU, Ciompi F, Cooper LAD, Coosemans A, Corredor G, Dantas Portela FL, Deman F, Demaria S, Dudgeon SN, Elghazawy M, Ely S, Femandez-Martín C, Fineberg S, Fox SB, Gallagher WM, Giltnane JM, Gnjatic S, Gonzalez-Ericsson PI, Grigoriadis A, Halama N, Hanna MG, Harbhajanka A, Hardas A, Hart SN, Hartman J, Hewitt S, Hida AI, Horlings HM, Husain Z, Hytopoulos E, Irshad S, Janssen EAM, Kahila M, Kataoka TR, Kawaguchi K, Kharidehal D, Khramtsov AI, Kiraz U, Kirtani P, Kodach LL, Korski K, Kovács A, Laenkholm AV, Lang-Schwarz C, Larsirnont D, Lennerz JK, Lerousseau M, Li X, Ly A, Madabhushi A, Maley SK, Narasimhamurthy VM, Marks DK, McDonald ES, Mehrotra R, Michiels S, Minhas FUAA, Mittal S, Moore DA, Mushtaq S, Nighat H, Papathomas T, Penault-Llorca F, Perera RD, Pinard CJ, Pinto-Cardenas JC, Pruneri G, Pusztai L, Rahman A, Rajpoot NM, Rapoport BL, Rau TT, Reis-Filho JS, Ribeiro JM, Rimm D, Vincent-Salomon A, Salto-Tellez M, et alPage DB, Broeckx G, Jahangir CA, Verbandt S, Gupta RR, Thagaard J, Khiroya R, Kos Z, Abduljabbar K, Acosta Haab G, Acs B, Akturk G, Almeida JS, Alvarado-Cabrero I, Azmoudeh-Ardalan F, Badve S, Baharun NB, Bellolio ER, Bheemaraju V, Blenman KRM, Botinelly Mendonça Fujimoto L, Bouchmaa N, Burgues O, Cheang MCU, Ciompi F, Cooper LAD, Coosemans A, Corredor G, Dantas Portela FL, Deman F, Demaria S, Dudgeon SN, Elghazawy M, Ely S, Femandez-Martín C, Fineberg S, Fox SB, Gallagher WM, Giltnane JM, Gnjatic S, Gonzalez-Ericsson PI, Grigoriadis A, Halama N, Hanna MG, Harbhajanka A, Hardas A, Hart SN, Hartman J, Hewitt S, Hida AI, Horlings HM, Husain Z, Hytopoulos E, Irshad S, Janssen EAM, Kahila M, Kataoka TR, Kawaguchi K, Kharidehal D, Khramtsov AI, Kiraz U, Kirtani P, Kodach LL, Korski K, Kovács A, Laenkholm AV, Lang-Schwarz C, Larsirnont D, Lennerz JK, Lerousseau M, Li X, Ly A, Madabhushi A, Maley SK, Narasimhamurthy VM, Marks DK, McDonald ES, Mehrotra R, Michiels S, Minhas FUAA, Mittal S, Moore DA, Mushtaq S, Nighat H, Papathomas T, Penault-Llorca F, Perera RD, Pinard CJ, Pinto-Cardenas JC, Pruneri G, Pusztai L, Rahman A, Rajpoot NM, Rapoport BL, Rau TT, Reis-Filho JS, Ribeiro JM, Rimm D, Vincent-Salomon A, Salto-Tellez M, Saltz J, Sayed S, Siziopikou KP, Sotiriou C, Stenzinger A, Sughayer MA, Sur D, Symmans F, Tanaka S, Taxter T, Tejpar S, Teuwen J, Thompson EA, Tramm T, Tran WT, van der Laak J, van Diest PJ, Verghese GE, Viale G, Vieth M, Wahab N, Walter T, Waumans Y, Wen HY, Yang W, Yuan Y, Adams S, Bartlett JMS, Loibl S, Denkert C, Savas P, Loi S, Salgado R, Specht Stovgaard E. Spatial analyses of immune cell infiltration in cancer: current methods and future directions: A report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. J Pathol 2023; 260:514-532. [PMID: 37608771 PMCID: PMC11288334 DOI: 10.1002/path.6165] [Show More Authors] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 08/24/2023]
Abstract
Modern histologic imaging platforms coupled with machine learning methods have provided new opportunities to map the spatial distribution of immune cells in the tumor microenvironment. However, there exists no standardized method for describing or analyzing spatial immune cell data, and most reported spatial analyses are rudimentary. In this review, we provide an overview of two approaches for reporting and analyzing spatial data (raster versus vector-based). We then provide a compendium of spatial immune cell metrics that have been reported in the literature, summarizing prognostic associations in the context of a variety of cancers. We conclude by discussing two well-described clinical biomarkers, the breast cancer stromal tumor infiltrating lymphocytes score and the colon cancer Immunoscore, and describe investigative opportunities to improve clinical utility of these spatial biomarkers. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- David B Page
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Glenn Broeckx
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Centre for Oncological Research (CORE), MIPPRO, Faculty of Medicine, Antwerp University, Antwerp, Belgium
| | - Chowdhury Arif Jahangir
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Sara Verbandt
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Rajarsi R Gupta
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Jeppe Thagaard
- Technical University of Denmark, Kongens Lyngby, Denmark
- Visiopharm A/S, Hørshoim, Denmark
| | - Reena Khiroya
- Department of Cellular Pathology, University College Hospital, London, UK
| | - Zuzana Kos
- Department of Pathology and Laboratory Medicine, BC Cancer Vancouver Centre, University of British Columbia, Vancouver, BC, Canada
| | - Khalid Abduljabbar
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | | | - Balazs Acs
- Department of Oncology and Pathology, Karolinska Institutet Stockholm, Sweden
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Guray Akturk
- Translational Molecular Biomarkers, Merck & Co Inc, Kenilworth, NJ, USA
| | - Jonas S Almeida
- National Cancer Institute, Division of Cancer Epidemiology and Genetics (DCEG), Rockville, MD, USA
| | | | | | - Sunil Badve
- Pathology and Laboratory Medicine, Emory University School of Medicine, Emory University Winship Cancer Institute, Atlanta, GA, USA
| | | | - Enrique R Bellolio
- Departamento de Anatomia Patológica, Facultad de Medicina, Universidad de La Frontera, Temuco, Chile
| | | | - Kim RM Blenman
- Internal Medicine Section of Medical Oncology and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Computer Science, Yale School of Engineering and Applied Science, New Haven, CT, USA
| | | | - Najat Bouchmaa
- Institute of Biological Sciences, Faculty of Medical Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir, Morocco
| | - Octavio Burgues
- Pathology Department Hospital Cliníco Universitario de Valencia/lncliva, Valencia, Spain
| | - Maggie Chon U Cheang
- Head of Integrative Genomics Analysis in Clinical Trials, ICR-CTSU, Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Francesco Ciompi
- Radboud University Medical Center, Department of Pathology, Nijmegen, The Netherlands
| | - Lee AD Cooper
- Department of Pathology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven, Belgium
| | - Germán Corredor
- Biomedical Engineering Department Emory University, Atlanta, GA, USA
| | | | - Frederik Deman
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Pathology, Weill Cornell Medicine, New York, NY, USA
| | - Sarah N Dudgeon
- Conputational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Mahmoud Elghazawy
- University of Surrey, Guildford, UK
- Ain Shams University, Cairo, Egypt
| | - Scott Ely
- Translational Pathology, Translational Sciences and Diagnostics/Translational Medicine/R&D, Bristol Myers Squibb, Princeton, NJ, USA
| | - Claudio Femandez-Martín
- Instituto Universitario de Investigación en Tecnología Centrada en el SerHumano, HUMAN-tech, Universitat Politècnica de València, Valencia, Spain
| | - Susan Fineberg
- Montefiore Medical Center and the Albert Einstein College of Medicine, New York, NY, USA
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Sacha Gnjatic
- Department of Oncological Sciences, Medicine Hem/One, and Pathology, Tisch Cancer Institute – Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
- Breast Cancer Now Research Unit School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Niels Halama
- Translational Immunotherapy, German Cancer Research Center, Heidelberg, Germany
| | | | | | - Alexandros Hardas
- Pathobiology & Population Sciences, The Royal Veterinary College, London, UK
| | - Steven N Hart
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Johan Hartman
- Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet Stockholm, Sweden
| | - Stephen Hewitt
- Department of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Akira I Hida
- Department of Pathology, Matsuyama Shimin Hospital, Matsuyama, Japan
| | - Hugo M Horlings
- Division of Pathology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | - Sheeba Irshad
- Kings College London & Guy’s & St Thomas’ NHS Trust, London, UK
| | - Emiel AM Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, Stavanger, Norway
| | - Mohamed Kahila
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Kosuke Kawaguchi
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Durga Kharidehal
- Department of Pathology, Narayana Medical College, Nellore, India
| | - Andrey I Khramtsov
- Pathology and Laboratory Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Umay Kiraz
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, Stavanger, Norway
| | - Pawan Kirtani
- Department of Histopathology, Aakash Healthcare Super Speciality Hospital, New Delhi, India
| | - Liudmila L Kodach
- Department of Pathology, Netherlands Cancer Institute – Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Konstanty Korski
- Data, Analytics and Imaging, Product Development, F.Hoffmann-La Roche AG, Basel, Switzerland
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg Sweden
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg Gothenburg, Sweden
| | - Anne-Vibeke Laenkholm
- Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
- Surgical Pathology, University of Copenhagen, Copenhagen, Denmark
| | - Corinna Lang-Schwarz
- Institute of Pathology, Klinikum Bayreuth GmbH, Friedrich-Alexander-University Erlangen-Nuremberg, Bayreuth, Germany
| | - Denis Larsirnont
- Institut Jules Bordet Université, Libre de Bruxelles, Brussels, Belgium
| | - Jochen K Lennerz
- Center for Integrated Diagnostics, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Marvin Lerousseau
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, Paris, France
- Institut Curie, PSL University, Paris, France
- INSERM, U900, Paris, France
| | - Xiaoxian Li
- Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Amy Ly
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Anant Madabhushi
- Biomedical Engineering, Radiology and Imaging Sciences, Biomedical Informatics, Pathology, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sai K Maley
- NRG Oncology/NSABP Foundation, Pittsburgh, PA, USA
| | | | - Douglas K Marks
- Perlmutter Cancer Center, NYU Langone Health, New York NY, USA
| | - Elizabeth S McDonald
- Breast Cancer Translational Research Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Mehrotra
- Indian Cancer Genome Atlas, Pune, India
- Centre for Health, Innovation and Policy Foundation, Noida, India
| | - Stefan Michiels
- Office of Biostatistics and Epidemiology, Gustave Roussy, Oncostat Ul 018, Inserm, University Paris-Saclay, Ligue Contre le Cancer labeled Team, Villejuif France
| | - Fayyaz ul Amir Afsar Minhas
- Tissue Image Analytics Centre, Warwick Cancer Research Centre, PathLAKE Consortium, Department of Computer Science, University of Warwick, Coventry, UK
| | - Shachi Mittal
- Department of Chemical Engineering, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - David A Moore
- CRUK Lung Cancer Centre of Excellence, UCLH, London, UK
| | - Shamim Mushtaq
- Department of Biochemistry, Ziauddin University, Karachi, Pakistan
| | - Hussain Nighat
- Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Raipur, India
| | - Thomas Papathomas
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Clinical Pathology, Drammen Sykehus, Vestre Viken HF, Drammen, Norway
| | - Frederique Penault-Llorca
- Centre Jean Perrin, INSERM U1240, Imagerie Moléculaire et Stratégies Théranostiques, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Rashindrie D Perera
- School of Electrical, Mechanical and Infrastructure Engineering, University of Melbourne, Melbourne, VIC, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Christopher J Pinard
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Department of Oncology, Lakeshore Animal Health Partners, Mississauga, ON, Canada
- Centre for Advancing Responsible and Ethical Artificial Intelligence (CARE-AI), University of Guelph, Guelph, ON, Canada
| | | | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | - Laios Pusztai
- Yale Cancer Center, New Haven, CT, USA
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Arman Rahman
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Bernardo Leon Rapoport
- The Medical Oncology Centre ofRosebank Johannesburg, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Tilman T Rau
- Institute of Pathology, University Hospital Düsseldorf and Heinrich-Heine-University Düsseldorf Düsseldorf Germany
| | - Jorge S Reis-Filho
- Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York NY, USA
| | - Joana M Ribeiro
- Département de Médecine Oncologique, Institute Gustave Roussy, Villejuif France
| | - David Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theranostic Medicine, Institut Curie, University Paris-Sciences et Lettres, Paris, France
| | - Manuel Salto-Tellez
- Integrated Pathology Unit Institute of Cancer Research, London, UK
- Precision Medicine Centre, Queen’s University Belfast Belfast UK
| | - Joel Saltz
- Department of Biomedical Informatics, Stony Brook Medicine, New York NY, USA
| | - Shahin Sayed
- Department of Pathology, Aga Khan University, Nairobi, Kenya
| | - Kalliopi P Siziopikou
- Department of Pathology, Section of Breast Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Medical Oncology Department Institut Jules Bordet Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg Centers for Personalized Medicine (ZPM), Heidelberg Germany
| | | | - Daniel Sur
- Department of Medical Oncology, University of Medicine and Pharmacy “luliu Hatieganu”, Cluj-Napoca, Romania
| | - Fraser Symmans
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sabine Tejpar
- Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jonas Teuwen
- Al for Oncology Lab, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Trine Tramm
- Pathology, and Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - William T Tran
- Department of Radiation Oncology, University of Toronto and Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht Utrecht The Netherlands
- Johns Hopkins Oncology Center, Baltimore, MD, USA
| | - Gregory E Verghese
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
- Breast Cancer Now Research Unit School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology & University of Milan, Milan, Italy
| | - Michael Vieth
- Institute of Pathology, Kiinikum Bayreuth GmbH, Friedrich-Alexander-University Erlangen-Nuremberg, Bayreuth, Germany
| | - Noorul Wahab
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick Coventry, UK
| | - Thomas Walter
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, Paris, France
- Institut Curie, PSL University, Paris, France
- INSERM, U900, Paris, France
| | | | - Hannah Y Wen
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wentao Yang
- Fudan Medical University Shanghai Cancer Center, Shanghai, PR China
| | - Yinyin Yuan
- Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sylvia Adams
- Perlmutter Cancer Center, NYU Langone Health, New York NY, USA
- Department of Medicine, NYU Grossman School of Medicine, Manhattan, NY, USA
| | | | - Sibylle Loibl
- Department of Medicine and Research, German Breast Group, Neu-lsenburg Germany
| | - Carsten Denkert
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Marburg, Marburg, Germany
| | - Peter Savas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC Australia
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Elisabeth Specht Stovgaard
- Department of Pathology, Herlev and Gentofte Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|