1
|
Zhang Y, Huang Y, Xu M, Zhuang J, Zhou Z, Zheng S, Zhu B, Guan G, Chen H, Liu X. Pathomics-based machine learning models for predicting pathological complete response and prognosis in locally advanced rectal cancer patients post-neoadjuvant chemoradiotherapy: insights from two independent institutional studies. BMC Cancer 2024; 24:1580. [PMID: 39725903 DOI: 10.1186/s12885-024-13328-w] [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: 08/13/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Accurate prediction of pathological complete response (pCR) and disease-free survival (DFS) in locally advanced rectal cancer (LARC) patients undergoing neoadjuvant chemoradiotherapy (NCRT) is essential for formulating effective treatment plans. This study aimed to construct and validate the machine learning (ML) models to predict pCR and DFS using pathomics. METHOD A retrospective analysis was conducted on 294 patients who received NCRT from two independent institutions. Pathomics from pre-NCRT H&E stains were extracted, and five ML models were developed and validated across two centers using ROC, Kaplan-Meier, time-dependent ROC, and nomogram analyses. RESULT Among the five ML models, the Xgboost (XGB) model demonstrated superior performance in predicting pCR, achieving an AUC of 1.000 (p < 0.001) on the internal data-set and an AUC of 0.950 (p = 0.001) on the external data-set.The XGB model effectively differentiated between high-risk and low-risk prognosis patients across all five centers: internal dataset (DFS, p = 0.002; OS, p = 0.004) and external dataset (DFS, p = 0.074; OS, p = 0.224).Furthermore, the COX regression demonstrated that the tumor length (HR = 1.230, 95%CI: 1.050-1.440, p = 0.010), post-NCRT CEA (HR = 1.716, 95%CI: 1.031- 2.858, p = 0.038), and XGB model score (HR = 0.128, 95%CI: 0.026-0.636, p = 0.012) were independent predictors of DFS after NCRT in the internal data-set.Using COX regression, the nomogram model and time-dependent AUC analysis demonstrated strong predictive discrimination for DFS in LARC patients across two independent institutions. CONCLUSION The ML model based on pathomics demonstrated effective prediction of pCR and prognosis in LARC patients. Further validation in larger cohorts is warranted to confirm the findings of this study.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Huang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meifang Xu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiazheng Zhuang
- Department of Gastrointestinal Surgery, The Quanzhou First Hospital Affiliated of Fujian Medical University, Quanzhou, China
| | - Zhibo Zhou
- Fujian Medical University, Fuzhou, China
| | | | - Bingwang Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guoxian Guan
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Hong Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| | - Xing Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
- National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou City, Fujian, 350001, China.
| |
Collapse
|
2
|
Hoang DT, Dinstag G, Shulman ED, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. A deep-learning framework to predict cancer treatment response from histopathology images through imputed transcriptomics. NATURE CANCER 2024; 5:1305-1317. [PMID: 38961276 DOI: 10.1038/s43018-024-00793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an indirect two-step approach consisting of (1) DeepPT, a deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response to targeted and immune therapies from the inferred expression values. We show that DeepPT successfully predicts transcriptomics in all 16 The Cancer Genome Atlas cohorts tested and generalizes well to two independent datasets. ENLIGHT-DeepPT successfully predicts true responders in five independent patient cohorts involving four different treatments spanning six cancer types, with an overall odds ratio of 2.28 and a 39.5% increased response rate among predicted responders versus the baseline rate. Notably, its prediction accuracy, obtained without any training on the treatment data, is comparable to that achieved by directly predicting the response from the images, which requires specific training on the treatment evaluation cohorts.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | | | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Leandro C Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - James L Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
3
|
Hijazi A, Bifulco C, Baldin P, Galon J. Digital Pathology for Better Clinical Practice. Cancers (Basel) 2024; 16:1686. [PMID: 38730638 PMCID: PMC11083211 DOI: 10.3390/cancers16091686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
(1) Background: Digital pathology (DP) is transforming the landscape of clinical practice, offering a revolutionary approach to traditional pathology analysis and diagnosis. (2) Methods: This innovative technology involves the digitization of traditional glass slides which enables pathologists to access, analyze, and share high-resolution whole-slide images (WSI) of tissue specimens in a digital format. By integrating cutting-edge imaging technology with advanced software, DP promises to enhance clinical practice in numerous ways. DP not only improves quality assurance and standardization but also allows remote collaboration among experts for a more accurate diagnosis. Artificial intelligence (AI) in pathology significantly improves cancer diagnosis, classification, and prognosis by automating various tasks. It also enhances the spatial analysis of tumor microenvironment (TME) and enables the discovery of new biomarkers, advancing their translation for therapeutic applications. (3) Results: The AI-driven immune assays, Immunoscore (IS) and Immunoscore-Immune Checkpoint (IS-IC), have emerged as powerful tools for improving cancer diagnosis, prognosis, and treatment selection by assessing the tumor immune contexture in cancer patients. Digital IS quantitative assessment performed on hematoxylin-eosin (H&E) and CD3+/CD8+ stained slides from colon cancer patients has proven to be more reproducible, concordant, and reliable than expert pathologists' evaluation of immune response. Outperforming traditional staging systems, IS demonstrated robust potential to enhance treatment efficiency in clinical practice, ultimately advancing cancer patient care. Certainly, addressing the challenges DP has encountered is essential to ensure its successful integration into clinical guidelines and its implementation into clinical use. (4) Conclusion: The ongoing progress in DP holds the potential to revolutionize pathology practices, emphasizing the need to incorporate powerful AI technologies, including IS, into clinical settings to enhance personalized cancer therapy.
Collapse
Affiliation(s)
- Assia Hijazi
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
| | - Carlo Bifulco
- Providence Genomics, Portland, OR 02912, USA;
- Earle A Chiles Research Institute, Portland, OR 97213, USA
| | - Pamela Baldin
- Department of Pathology, Cliniques Universitaires Saint Luc, UCLouvain, 1200 Brussels, Belgium;
| | - Jérôme Galon
- The French National Institute of Health & Medical Research (INSERM), Laboratory of Integrative Cancer Immunology, F-75006 Paris, France;
- Equipe Labellisée Ligue Contre le Cancer, F-75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, F-75006 Paris, France
- Veracyte, 13009 Marseille, France
| |
Collapse
|
4
|
Zeng H, Qiu S, Zhuang S, Wei X, Wu J, Zhang R, Chen K, Wu Z, Zhuang Z. Deep learning-based predictive model for pathological complete response to neoadjuvant chemotherapy in breast cancer from biopsy pathological images: a multicenter study. Front Physiol 2024; 15:1279982. [PMID: 38357498 PMCID: PMC10864440 DOI: 10.3389/fphys.2024.1279982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction: Early predictive pathological complete response (pCR) is beneficial for optimizing neoadjuvant chemotherapy (NAC) strategies for breast cancer. The hematoxylin and eosin (HE)-stained slices of biopsy tissues contain a large amount of information on tumor epithelial cells and stromal. The fusion of pathological image features and clinicopathological features is expected to build a model to predict pCR of NAC in breast cancer. Methods: We retrospectively collected a total of 440 breast cancer patients from three hospitals who underwent NAC. HE-stained slices of biopsy tissues were scanned to form whole-slide images (WSIs), and pathological images of representative regions of interest (ROI) of each WSI were selected at different magnifications. Based on several different deep learning models, we propose a novel feature extraction method on pathological images with different magnifications. Further, fused with clinicopathological features, a multimodal breast cancer NAC pCR prediction model based on a support vector machine (SVM) classifier was developed and validated with two additional validation cohorts (VCs). Results: Through experimental validation of several different deep learning models, we found that the breast cancer pCR prediction model based on the SVM classifier, which uses the VGG16 model for feature extraction of pathological images at ×20 magnification, has the best prediction efficacy. The area under the curve (AUC) of deep learning pathological model (DPM) were 0.79, 0.73, and 0.71 for TC, VC1, and VC2, respectively, all of which exceeded 0.70. The AUCs of clinical model (CM), a clinical prediction model established by using clinicopathological features, were 0.79 for TC, 0.73 for VC1, and 0.71 for VC2, respectively. The multimodal deep learning clinicopathological model (DPCM) established by fusing pathological images and clinicopathological features improved the AUC of TC from 0.79 to 0.84. The AUC of VC2 improved from 0.71 to 0.78. Conclusion: Our study reveals that pathological images of HE-stained slices of pre-NAC biopsy tissues can be used to build a pCR prediction model. Combining pathological images and clinicopathological features can further enhance the predictive efficacy of the model.
Collapse
Affiliation(s)
- Huancheng Zeng
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Siqi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| | - Shuxin Zhuang
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, China
| | - Xiaolong Wei
- The Pathology Department, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ranze Zhang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Kai Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Zhiyong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
| | - Zhemin Zhuang
- Engineering College, Shantou University, Shantou, China
| |
Collapse
|
5
|
Hoang DT, Dinstag G, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. Prediction of cancer treatment response from histopathology images through imputed transcriptomics. RESEARCH SQUARE 2023:rs.3.rs-3193270. [PMID: 37790315 PMCID: PMC10543028 DOI: 10.21203/rs.3.rs-3193270/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin (H&E)-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an approach for predicting response to multiple targeted and immunotherapies from H&E-slides. In difference from existing approaches that aim to predict treatment response directly from the slides, ENLIGHT-DeepPT is an indirect two-step approach consisting of (1) DeepPT, a new deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response based on the DeepPT inferred expression values. DeepPT successfully predicts transcriptomics in all 16 TCGA cohorts tested and generalizes well to two independent datasets. Our key contribution is showing that ENLIGHT-DeepPT successfully predicts true responders in five independent patients' cohorts involving four different treatments spanning six cancer types with an overall odds ratio of 2.44, increasing the baseline response rate by 43.47% among predicted responders, without the need for any treatment data for training. Furthermore, its prediction accuracy on these datasets is comparable to a supervised approach predicting the response directly from the images, which needs to be trained and tested on the same cohort. ENLIGHT-DeepPT future application could provide clinicians with rapid treatment recommendations to an array of different therapies and importantly, may contribute to advancing precision oncology in developing countries.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | | | - Leandro C. Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H. Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R. Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - James L. Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A. Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
6
|
Martos O, Hoque MZ, Keskinarkaus A, Kemi N, Näpänkangas J, Eskuri M, Pohjanen VM, Kauppila JH, Seppänen T. Optimized detection and segmentation of nuclei in gastric cancer images using stain normalization and blurred artifact removal. Pathol Res Pract 2023; 248:154694. [PMID: 37494804 DOI: 10.1016/j.prp.2023.154694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Histological analysis with microscopy is the gold standard to diagnose and stage cancer, where slides or whole slide images are analyzed for cell morphological and spatial features by pathologists. The nuclei of cancerous cells are characterized by nonuniform chromatin distribution, irregular shapes, and varying size. As nucleus area and shape alone carry prognostic value, detection and segmentation of nuclei are among the most important steps in disease grading. However, evaluation of nuclei is a laborious, time-consuming, and subjective process with large variation among pathologists. Recent advances in digital pathology have allowed significant applications in nuclei detection, segmentation, and classification, but automated image analysis is greatly affected by staining factors, scanner variability, and imaging artifacts, requiring robust image preprocessing, normalization, and segmentation methods for clinically satisfactory results. In this paper, we aimed to evaluate and compare the digital image analysis techniques used in clinical pathology and research in the setting of gastric cancer. A literature review was conducted to evaluate potential methods of improving nuclei detection. Digitized images of 35 patients from a retrospective cohort of gastric adenocarcinoma at Oulu University Hospital in 1987-2016 were annotated for nuclei (n = 9085) by expert pathologists and 14 images of different cancer types from public TCGA dataset with annotated nuclei (n = 7000) were used as a comparison to evaluate applicability in other cancer types. The detection and segmentation accuracy with the selected color normalization and stain separation techniques were compared between the methods. The extracted information can be supplemented by patient's medical data and fed to the existing statistical clinical tools or subjected to subsequent AI-assisted classification and prediction models. The performance of each method is evaluated by several metrics against the annotations done by expert pathologists. The F1-measure of 0.854 ± 0.068 is achieved with color normalization for the gastric cancer dataset, and 0.907 ± 0.044 with color deconvolution for the public dataset, showing comparable results to the earlier state-of-the-art works. The developed techniques serve as a basis for further research on application and interpretability of AI-assisted tools for gastric cancer diagnosis.
Collapse
Affiliation(s)
- Oleg Martos
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| | - Md Ziaul Hoque
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland.
| | - Anja Keskinarkaus
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| | - Niko Kemi
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Juha Näpänkangas
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Maarit Eskuri
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Vesa-Matti Pohjanen
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Joonas H Kauppila
- Department of Surgery, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Tapio Seppänen
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| |
Collapse
|
7
|
Wu Y, Li Y, Xiong X, Liu X, Lin B, Xu B. Recent advances of pathomics in colorectal cancer diagnosis and prognosis. Front Oncol 2023; 13:1094869. [PMID: 37538112 PMCID: PMC10396402 DOI: 10.3389/fonc.2023.1094869] [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: 11/10/2022] [Accepted: 06/13/2023] [Indexed: 08/05/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies, with the third highest incidence and the second highest mortality in the world. To improve the therapeutic outcome, the risk stratification and prognosis predictions would help guide clinical treatment decisions. Achieving these goals have been facilitated by the fast development of artificial intelligence (AI) -based algorithms using radiological and pathological data, in combination with genomic information. Among them, features extracted from pathological images, termed pathomics, are able to reflect sub-visual characteristics linking to better stratification and prediction of therapeutic responses. In this paper, we review recent advances in pathological image-based algorithms in CRC, focusing on diagnosis of benign and malignant lesions, micro-satellite instability, as well as prediction of neoadjuvant chemoradiotherapy and the prognosis of CRC patients.
Collapse
Affiliation(s)
- Yihan Wu
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Li
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
- Bioengineering College, Chongqing University, Chongqing, China
| | - Xiaomin Xiong
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
- Bioengineering College, Chongqing University, Chongqing, China
| | - Xiaohua Liu
- Bioengineering College, Chongqing University, Chongqing, China
| | - Bo Lin
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Bo Xu
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
8
|
Wu Y, Liu X, Liu F, Li Y, Xiong X, Sun H, Lin B, Li Y, Xu B. A multi-class classification algorithm based on hematoxylin-eosin staining for neoadjuvant therapy in rectal cancer: a retrospective study. PeerJ 2023; 11:e15408. [PMID: 37334122 PMCID: PMC10269576 DOI: 10.7717/peerj.15408] [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: 12/27/2022] [Accepted: 04/21/2023] [Indexed: 06/20/2023] Open
Abstract
Neoadjuvant therapy (NAT) is a major treatment option for locally advanced rectal cancer. With recent advancement of machine/deep learning algorithms, predicting the treatment response of NAT has become possible using radiological and/or pathological images. However, programs reported thus far are limited to binary classifications, and they can only distinguish the pathological complete response (pCR). In the clinical setting, the pathological NAT responses are classified as four classes: (TRG0-3), with 0 as pCR, 1 as moderate response, 2 as minimal response and 3 as poor response. Therefore, the actual clinical need for risk stratification remains unmet. By using ResNet (Residual Neural Network), we developed a multi-class classifier based on Hematoxylin-Eosin (HE) images to divide the response to three groups (TRG0, TRG1/2, and TRG3). Overall, the model achieved the AUC 0.97 at 40× magnification and AUC 0.89 at 10× magnification. For TRG0, the model under 40× magnification achieved a precision of 0.67, a sensitivity of 0.67, and a specificity of 0.95. For TRG1/2, a precision of 0.92, a sensitivity of 0.86, and a specificity of 0.89 were achieved. For TRG3, the model obtained a precision of 0.71, a sensitivity of 0.83, and a specificity of 0.88. To find the relationship between the treatment response and pathological images, we constructed a visual heat map of tiles using Class Activation Mapping (CAM). Notably, we found that tumor nuclei and tumor-infiltrating lymphocytes appeared to be potential features of the algorithm. Taken together, this multi-class classifier represents the first of its kind to predict different NAT responses in rectal cancer.
Collapse
Affiliation(s)
- Yihan Wu
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaohua Liu
- Bioengineering College of Chongqing University, Chongqing, China
| | - Fang Liu
- Department of Pathology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yi Li
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
- Bioengineering College of Chongqing University, Chongqing, China
| | - Xiaomin Xiong
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
- Bioengineering College of Chongqing University, Chongqing, China
| | - Hao Sun
- Department of Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Bo Lin
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| | - Yu Li
- Department of Pathology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Bo Xu
- School of Medicine, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
9
|
Wang L, Wu X, Tian R, Ma H, Jiang Z, Zhao W, Cui G, Li M, Hu Q, Yu X, Xu W. MRI-based pre-Radiomics and delta-Radiomics models accurately predict the post-treatment response of rectal adenocarcinoma to neoadjuvant chemoradiotherapy. Front Oncol 2023; 13:1133008. [PMID: 36925913 PMCID: PMC10013156 DOI: 10.3389/fonc.2023.1133008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Objectives To develop and validate magnetic resonance imaging (MRI)-based pre-Radiomics and delta-Radiomics models for predicting the treatment response of local advanced rectal cancer (LARC) to neoadjuvant chemoradiotherapy (NCRT). Methods Between October 2017 and August 2022, 105 LARC NCRT-naïve patients were enrolled in this study. After careful evaluation, data for 84 patients that met the inclusion criteria were used to develop and validate the NCRT response models. All patients received NCRT, and the post-treatment response was evaluated by pathological assessment. We manual segmented the volume of tumors and 105 radiomics features were extracted from three-dimensional MRIs. Then, the eXtreme Gradient Boosting algorithm was implemented for evaluating and incorporating important tumor features. The predictive performance of MRI sequences and Synthetic Minority Oversampling Technique (SMOTE) for NCRT response were compared. Finally, the optimal pre-Radiomics and delta-Radiomics models were established respectively. The predictive performance of the radionics model was confirmed using 5-fold cross-validation, 10-fold cross-validation, leave-one-out validation, and independent validation. The predictive accuracy of the model was based on the area under the receiver operator characteristic (ROC) curve (AUC). Results There was no significant difference in clinical factors between patients with good and poor reactions. Integrating different MRI modes and the SMOTE method improved the performance of the radiomics model. The pre-Radiomics model (train AUC: 0.93 ± 0.06; test AUC: 0.79) and delta-Radiomcis model (train AUC: 0.96 ± 0.03; test AUC: 0.83) all have high NCRT response prediction performance by LARC. Overall, the delta-Radiomics model was superior to the pre-Radiomics model. Conclusion MRI-based pre-Radiomics model and delta-Radiomics model all have good potential to predict the post-treatment response of LARC to NCRT. Delta-Radiomics analysis has a huge potential for clinical application in facilitating the provision of personalized therapy.
Collapse
Affiliation(s)
- Likun Wang
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Molecular Imaging and Nuclear Medicine, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Department of Ultrasound Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xueliang Wu
- Graduate School, Tianjin Medical University, Tianjin, China.,Department of Gastrointestinal Surgery, Tianjin Medical University Nankai Hospital, Tianjin, China
| | - Ruoxi Tian
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongqing Ma
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zekun Jiang
- College of Computer Science, Sichuan University, Chengdu, China
| | - Weixin Zhao
- College of Computer Science, Sichuan University, Chengdu, China
| | - Guoqing Cui
- Medical Image Center, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Meng Li
- Graduate School, Hebei North University, Zhangjiakou, China
| | - Qinsheng Hu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangyang Yu
- Department of Gastrointestinal Surgery, Tianjin Medical University Nankai Hospital, Tianjin, China
| | - Wengui Xu
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
10
|
Predicting neoadjuvant chemotherapy benefit using deep learning from stromal histology in breast cancer. NPJ Breast Cancer 2022; 8:124. [PMID: 36418332 PMCID: PMC9684483 DOI: 10.1038/s41523-022-00491-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) is a standard treatment option for locally advanced breast cancer. However, not all patients benefit from NAC; some even obtain worse outcomes after therapy. Hence, predictors of treatment benefit are crucial for guiding clinical decision-making. Here, we investigated the predictive potential of breast cancer stromal histology via a deep learning (DL)-based approach and proposed the tumor-associated stroma score (TS-score) for predicting pathological complete response (pCR) to NAC with a multicenter dataset. The TS-score was demonstrated to be an independent predictor of pCR, and it not only outperformed the baseline variables and stromal tumor-infiltrating lymphocytes (sTILs) but also significantly improved the prediction performance of the baseline variable-based model. Furthermore, we discovered that unlike lymphocytes, collagen and fibroblasts in the stroma were likely associated with a poor response to NAC. The TS-score has the potential to better stratify breast cancer patients in NAC settings.
Collapse
|
11
|
Deep learning with biopsy whole slide images for pretreatment prediction of pathological complete response to neoadjuvant chemotherapy in breast cancer:A multicenter study. Breast 2022; 66:183-190. [PMID: 36308926 PMCID: PMC9619175 DOI: 10.1016/j.breast.2022.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION Predicting pathological complete response (pCR) for patients receiving neoadjuvant chemotherapy (NAC) is crucial in establishing individualized treatment. Whole-slide images (WSIs) of tumor tissues reflect the histopathologic information of the tumor, which is important for therapeutic response effectiveness. In this study, we aimed to investigate whether predictive information for pCR could be detected from WSIs. MATERIALS AND METHODS We retrospectively collected data from four cohorts of 874 patients diagnosed with biopsy-proven breast cancer. A deep learning pathological model (DLPM) was constructed to predict pCR using biopsy WSIs in the primary cohort, and it was then validated in three external cohorts. The DLPM could generate a deep learning pathological score (DLPs) for each patient; stromal tumor-infiltrating lymphocytes (TILs) were selected for comparison with DLPs. RESULTS The WSI feature-based DLPM showed good predictive performance with the highest area under the curve (AUC) of 0.72 among the cohorts. Alternatively, the combination of the DLPM and clinical characteristics offered a better prediction performance (AUC >0.70) in all cohorts. We also evaluated the performance of DLPM in three different breast subtypes with the best prediction for the triple-negative breast cancer (TNBC) subtype (AUC: 0.73). Moreover, DLPM combined with clinical characteristics and stromal TILs achieved the highest AUC in the primary cohort (AUC: 0.82) and validation cohort 1 (AUC: 0.80). CONCLUSION Our study suggested that WSIs integrated with deep learning could potentially predict pCR to NAC in breast cancer. The predictive performance will be improved by combining clinical characteristics. DLPs from DLPM can provide more information compared to stromal TILs for pCR prediction.
Collapse
|
12
|
Viswanath SE. Editorial for "Selecting Candidates for Organ-Preserving Strategies After Neoadjuvant Chemoradiotherapy for Rectal Cancer: Development and Validation of a Model Integrating MRI Radiomics and Pathomics". J Magn Reson Imaging 2022; 56:1143-1144. [PMID: 35244965 PMCID: PMC9440947 DOI: 10.1002/jmri.28139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Satish E Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
13
|
Jiang W, Wang S, Wan J, Zheng J, Dong X, Liu Z, Wang G, Xu S, Xiao W, Gao Y, Zhuo S, Yan J. Association of the Collagen Signature with Pathological Complete Response in Rectal Cancer Patients. Cancer Sci 2022; 113:2409-2424. [PMID: 35485874 PMCID: PMC9277261 DOI: 10.1111/cas.15385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 11/28/2022] Open
Abstract
Collagen in the tumor microenvironment is recognized as a potential biomarker for predicting treatment response. This study investigated whether the collagen features are associated with pathological complete response (pCR) in locally advanced rectal cancer (LARC) patients receiving neoadjuvant chemoradiotherapy (nCRT) and develop and validate a prediction model for individualized prediction of pCR. The prediction model was developed in a primary cohort (353 consecutive patients). In total, 142 collagen features were extracted from the multiphoton image of pretreatment biopsy, and the least absolute shrinkage and selection operator (Lasso) regression was applied for feature selection and collagen signature building. A nomogram was developed using multivariable analysis. The performance of the nomogram was assessed with respect to its discrimination, calibration, and clinical utility. An independent cohort (163 consecutive patients) was used to validate the model. The collagen signature comprised four collagen features significantly associated with pCR both in the primary and validation cohorts (p < 0.001). Predictors in the individualized prediction nomogram included the collagen signature and clinicopathological predictors. The nomogram showed good discrimination with area under the ROC curve (AUC) of 0.891 in the primary cohort and good calibration. Application of the nomogram in the validation cohort still gave good discrimination (AUC = 0.908) and good calibration. Decision curve analysis demonstrated that the nomogram was clinically useful. In conclusion, the collagen signature in the tumor microenvironment of pretreatment biopsy is significantly associated with pCR. The nomogram based on the collagen signature and clinicopathological predictors could be used for individualized prediction of pCR in LARC patients before nCRT.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.,School of Science, Jimei University, Xiamen, Fujian, 361021, China
| | - Shijie Wang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jinliang Wan
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jixiang Zheng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoyu Dong
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhangyuanzhu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Guangxing Wang
- School of Science, Jimei University, Xiamen, Fujian, 361021, China
| | - Shuoyu Xu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.,Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Weiwei Xiao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, China
| | - Yuanhong Gao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, Fujian, 361021, China
| | - Jun Yan
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
14
|
He L, Huang Y, Chen X, Huang X, Wang H, Zhang Y, Liang C, Li Z, Yan L, Liu Z. Development and Validation of an Immune-Based Prognostic Risk Score for Patients With Resected Non-Small Cell Lung Cancer. Front Immunol 2022; 13:835630. [PMID: 35401554 PMCID: PMC8983932 DOI: 10.3389/fimmu.2022.835630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDespite the well-known role of immunoscore, as a prognostic tool, that appeared to be superior to tumor–node–metastasis (TNM) staging system, no prognostic scoring system based on immunohistochemistry (IHC) staining digital image analysis has been established in non-small cell lung cancer (NSCLC). Hence, we aimed to develop and validate an immune-based prognostic risk score (IMPRS) that could markedly improve individualized prediction of postsurgical survival in patients with resected NSCLC.MethodsIn this retrospective study, complete resection of NSCLC (stage I–IIIA) was performed for two independent patient cohorts (discovery cohort, n=168; validation cohort, n=115). Initially, paraffin-embedded resected specimens were stained by immunohistochemistry (IHC) of three immune cell types (CD3+, CD4+, and CD8+ T cells), and a total of 5,580 IHC-immune features were extracted from IHC digital images for each patient by using fully automated pipeline. Then, an IHC-immune signature was constructed with selected features using the LASSO Cox analysis, and the association of signature with patients’ overall survival (OS) was analyzed by Kaplan–Meier method. Finally, IMPRS was established by incorporating IHC-immune signature and independent clinicopathological variables in multivariable Cox regression analysis. Furthermore, an external validation cohort was included to validate this prognostic risk score.ResultsEight key IHC-immune features were selected for the construction of IHC-immune signature, which showed significant associations with OS in all cohorts [discovery: hazard ratio (HR)=11.518, 95%CI, 5.444–24.368; validation: HR=2.664, 95%CI, 1.029–6.896]. Multivariate analyses revealed IHC-immune signature as an independent prognostic factor, and age, T stage, and N stage were also identified and entered into IMPRS (all p<0.001). IMPRS had good discrimination ability for predicting OS (C-index, 0.869; 95%CI, 0.861–0.877), confirmed using external validation cohort (0.731, 0.717–0.745). Interestingly, IMPRS had better prognostic value than clinicopathological-based model and TNM staging system termed as C-index (clinicopathological-based model: 0.674; TNM staging: 0.646, all p<0.05). More importantly, decision curve analysis showed that IMPRS had adequate performance for predicting OS in resected NSCLC patients.ConclusionsOur findings indicate that the IMPRS that we constructed can provide more accurate prognosis for individual prediction of OS for patients with resected NSCLC, which can help in guiding personalized therapy and improving outcomes for patients.
Collapse
Affiliation(s)
- Lan He
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaomei Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huihui Wang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuan Zhang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhenhui Li
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
- *Correspondence: Zaiyi Liu, ; Lixu Yan, ; Zhenhui Li,
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Zaiyi Liu, ; Lixu Yan, ; Zhenhui Li,
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Zaiyi Liu, ; Lixu Yan, ; Zhenhui Li,
| |
Collapse
|
15
|
Wan L, Sun Z, Peng W, Wang S, Li J, Zhao Q, Wang S, Ouyang H, Zhao X, Zou S, Zhang H. Selecting Candidates for Organ-Preserving Strategies After Neoadjuvant Chemoradiotherapy for Rectal Cancer: Development and Validation of a Model Integrating MRI Radiomics and Pathomics. J Magn Reson Imaging 2022; 56:1130-1142. [PMID: 35142001 DOI: 10.1002/jmri.28108] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Histopathologic evaluation after surgery is the gold standard to evaluate treatment response to neoadjuvant chemoradiotherapy (nCRT) in locally advanced rectal cancer (LARC). However, it cannot be used to guide organ-preserving strategies due to poor timeliness. PURPOSE To develop and validate a multiscale model incorporating radiomics and pathomics features for predicting pathological good response (pGR) of down-staging to stage ypT0-1N0 after nCRT. STUDY TYPE Retrospective. POPULATION A total of 153 patients (median age, 55 years; 109 men; 107 training group; 46 validation group) with clinicopathologically confirmed LARC. FIELD STRENGTH/SEQUENCE A 3.0-T; fast spin echo T2 -weighted and single-shot EPI diffusion-weighted images. ASSESSMENT The differences in clinicoradiological variables between pGR and non-pGR groups were assessed. Pretreatment and posttreatment radiomics signatures, and pathomics signature were constructed. A multiscale pGR prediction model was established. The predictive performance of the model was evaluated and compared to that of the clinicoradiological model. STATISTICAL TESTS The χ2 test, Fisher's exact test, t-test, the minimum redundancy maximum relevance algorithm, the least absolute shrinkage and selection operator logistic regression algorithm, regression analysis, receiver operating characteristic curve (ROC) analysis, Delong method. P < 0.05 indicated a significant difference. RESULTS Pretreatment radiomics signature (odds ratio [OR] = 2.53; 95% CI: 1.58-4.66), posttreatment radiomics signature (OR = 9.59; 95% CI: 3.04-41.46), and pathomics signature (OR = 3.14; 95% CI: 1.40-8.31) were independent factors for predicting pGR. The multiscale model presented good predictive performance with areas under the curve (AUC) of 0.93 (95% CI: 0.88-0.98) and 0.90 (95% CI: 0.78-1.00) in the training and validation groups, those were significantly higher than that of the clinicoradiological model with AUCs of 0.69 (95% CI: 0.55-0.82) and 0.68 (95% CI: 0.46-0.91) in both groups. DATA CONCLUSION A model incorporating radiomics and pathomics features effectively predicted pGR after nCRT in patients with LARC. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Lijuan Wan
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Zhuo Sun
- Thorough Images, Chaoyang District, Beijing, China
| | - Wenjing Peng
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Sicong Wang
- Department of Pharmaceutical Diagnosis, GE Healthcare, Life Sciences, Beijing, China
| | - Jiangtao Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Qing Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Shuhao Wang
- Thorough Images, Chaoyang District, Beijing, China
| | - Han Ouyang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Xinming Zhao
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| | - Hongmei Zhang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing, China
| |
Collapse
|
16
|
Digital pathology and artificial intelligence in translational medicine and clinical practice. Mod Pathol 2022; 35:23-32. [PMID: 34611303 PMCID: PMC8491759 DOI: 10.1038/s41379-021-00919-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023]
Abstract
Traditional pathology approaches have played an integral role in the delivery of diagnosis, semi-quantitative or qualitative assessment of protein expression, and classification of disease. Technological advances and the increased focus on precision medicine have recently paved the way for the development of digital pathology-based approaches for quantitative pathologic assessments, namely whole slide imaging and artificial intelligence (AI)-based solutions, allowing us to explore and extract information beyond human visual perception. Within the field of immuno-oncology, the application of such methodologies in drug development and translational research have created invaluable opportunities for deciphering complex pathophysiology and the discovery of novel biomarkers and drug targets. With an increasing number of treatment options available for any given disease, practitioners face the growing challenge of selecting the most appropriate treatment for each patient. The ever-increasing utilization of AI-based approaches substantially expands our understanding of the tumor microenvironment, with digital approaches to patient stratification and selection for diagnostic assays supporting the identification of the optimal treatment regimen based on patient profiles. This review provides an overview of the opportunities and limitations around implementing AI-based methods in biomarker discovery and patient selection and discusses how advances in digital pathology and AI should be considered in the current landscape of translational medicine, touching on challenges this technology may face if adopted in clinical settings. The traditional role of pathologists in delivering accurate diagnoses or assessing biomarkers for companion diagnostics may be enhanced in precision, reproducibility, and scale by AI-powered analysis tools.
Collapse
|
17
|
Zhang D, Duan Y, Guo J, Wang Y, Yang Y, Li Z, Wang K, Wu L, Yu M. Using Multi-Scale Convolutional Neural Network Based on Multi-Instance Learning to Predict the Efficacy of Neoadjuvant Chemoradiotherapy for Rectal Cancer. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2022; 10:4300108. [PMID: 35317416 PMCID: PMC8932521 DOI: 10.1109/jtehm.2022.3156851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/06/2021] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
Abstract
Background: At present, radical total mesorectal excision after neoadjuvant chemoradiotherapy is crucial for locally advanced rectal cancer. Therefore, the use of histopathological images analysis technology to predict the efficacy of neoadjuvant chemoradiotherapy for rectal cancer is of great significance for the subsequent treatment of patients. Methods: In this study, we propose a new pathological images analysis method based on multi-instance learning to predict the efficacy of neoadjuvant chemoradiotherapy for rectal cancer. Specifically, we proposed a gated attention normalization mechanism based on the multilayer perceptron, which accelerates the convergence of stochastic gradient descent optimization and can speed up the training process. We also proposed a bilinear attention multi-scale feature fusion mechanism, which organically fuses the global features of the larger receptive fields and the detailed features of the smaller receptive fields and alleviates the problem of pathological images context information loss caused by block sampling. At the same time, we also designed a weighted loss function to alleviate the problem of imbalance between cancerous instances and normal instances. Results: We evaluated our method on a locally advanced rectal cancer dataset containing 150 whole slide images. In addition, to verify our method’s generalization performance, we also tested on two publicly available datasets, Camelyon16 and MSKCC. The results show that the AUC values of our method on the Camelyon16 and MSKCC datasets reach 0.9337 and 0.9091, respectively. Conclusion: Our method has outstanding performance and advantages in predicting the efficacy of neoadjuvant chemoradiotherapy for rectal cancer. Clinical and Translational Impact Statement—This study aims to predict the efficacy of neoadjuvant chemoradiotherapy for rectal cancer to assist clinicians quickly diagnose and formulate personalized treatment plans for patients.
Collapse
Affiliation(s)
- Dehai Zhang
- School of SoftwareYunnan University Kunming 650106 China
| | - Yongchun Duan
- School of SoftwareYunnan University Kunming 650106 China
| | - Jing Guo
- School of Information Science and EngineeringYunnan University Kunming 650106 China
| | - Yaowei Wang
- School of SoftwareYunnan University Kunming 650106 China
| | - Yun Yang
- Key Laboratory in Software Engineering of Yunnan Province, School of SoftwareYunnan University Kunming 650504 China
| | - Zhenhui Li
- Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center Kunming 650118 China
| | - Kelong Wang
- School of SoftwareYunnan University Kunming 650106 China
| | - Lin Wu
- Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center Kunming 650118 China
| | - Minghao Yu
- School of SoftwareYunnan University Kunming 650106 China
| |
Collapse
|
18
|
Li F, Yang Y, Wei Y, He P, Chen J, Zheng Z, Bu H. Deep learning-based predictive biomarker of pathological complete response to neoadjuvant chemotherapy from histological images in breast cancer. J Transl Med 2021; 19:348. [PMID: 34399795 PMCID: PMC8365907 DOI: 10.1186/s12967-021-03020-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/02/2021] [Indexed: 02/08/2023] Open
Abstract
Background Pathological complete response (pCR) is considered a surrogate endpoint for favorable survival in breast cancer patients treated with neoadjuvant chemotherapy (NAC). Predictive biomarkers of treatment response are crucial for guiding treatment decisions. With the hypothesis that histological information on tumor biopsy images could predict NAC response in breast cancer, we proposed a novel deep learning (DL)-based biomarker that predicts pCR from images of hematoxylin and eosin (H&E)-stained tissue and evaluated its predictive performance. Methods In total, 540 breast cancer patients receiving standard NAC were enrolled. Based on H&E-stained images, DL methods were employed to automatically identify tumor epithelium and predict pCR by scoring the identified tumor epithelium to produce a histopathological biomarker, the pCR-score. The predictive performance of the pCR-score was assessed and compared with that of conventional biomarkers including stromal tumor-infiltrating lymphocytes (sTILs) and subtype. Results The pCR-score derived from H&E staining achieved an area under the curve (AUC) of 0.847 in predicting pCR directly, and achieved accuracy, F1 score, and AUC of 0.853, 0.503, and 0.822 processed by the logistic regression method, respectively, higher than either sTILs or subtype; a prediction model of pCR constructed by integrating sTILs, subtype and pCR-score yielded a mean AUC of 0.890, outperforming the baseline sTIL-subtype model by 0.051 (0.839, P = 0.001). Conclusion The DL-based pCR-score from histological images is predictive of pCR better than sTILs and subtype, and holds the great potentials for a more accurate stratification of patients for NAC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03020-z.
Collapse
Affiliation(s)
- Fengling Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.,Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongquan Yang
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yani Wei
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.,Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping He
- Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Chen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongxi Zheng
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Liang Y, Zhu Y, Lin H, Zhang S, Li S, Huang Y, Liu C, Qu J, Liang C, Zhao K, Li Z, Liu Z. The value of the tumour-stroma ratio for predicting neoadjuvant chemoradiotherapy response in locally advanced rectal cancer: a case control study. BMC Cancer 2021; 21:729. [PMID: 34172021 PMCID: PMC8235870 DOI: 10.1186/s12885-021-08516-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/16/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The tumour-stroma ratio (TSR) is recognized as a practical prognostic factor in colorectal cancer. However, TSR assessment generally utilizes surgical specimens. This study aims to investigate whether the TSR evaluated from preoperative biopsy specimens by a semi-automatic quantification method can predict the response after neoadjuvant chemoradiotherapy (nCRT) of patients with locally advanced rectal cancer (LARC). METHODS A total of 248 consecutive patients diagnosed with LARC and treated with nCRT followed by resection were included. Haematoxylin and eosin (HE)-stained sections of biopsy specimens were collected, and the TSR was evaluated by a semi-automatic quantification method and was divided into three categories, using the cut-offs determined in the whole cohort to balance the proportion of patients in each category. The response to nCRT was evaluated on the primary tumour resection specimen by an expert pathologist using the four-tier tumour regression grade (TRG) system. RESULTS The TSR can discriminate patients that are major-responders (TRG 0-1) from patients that are non-responders (TRG 2-3). Patients were divided into stroma-low (33.5%), stroma-intermediate (33.9%), and stroma-high (32.7%) groups using 56.3 and 72.8% as the cutoffs. In the stroma-low group, 58 (69.9%) patients were major-responders, and only 39 (48.1%) patients were considered major-responders in the stroma-high group (P = 0.018). Multivariate analysis showed that the TSR was the only pre-treatment predictor of response to nCRT (adjusted odds ratio 0.40, 95% confidence interval 0.21-0.76, P = 0.002). CONCLUSION An elevated TSR in preoperative biopsy specimens is an independent predictor of nCRT response in LARC. This semi-automatic quantified TSR could be easily translated into routine pathologic assessment due to its reproducibility and reliability.
Collapse
Affiliation(s)
- Yanting Liang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huan Lin
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Shenyan Zhang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Suyun Li
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yanqi 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, China
| | - Chen Liu
- 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, China
| | - Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Ke Zhao
- School of Medicine, South China University of Technology, Guangzhou, China.
- 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, Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
20
|
Li T, Yu Z, Yang Y, Fu Z, Chen Z, Li Q, Zhang K, Luo Z, Qiu Z, Huang C. Rapid multi-dynamic algorithm for gray image analysis of the stroma percentage on colorectal cancer. J Cancer 2021; 12:4561-4573. [PMID: 34149920 PMCID: PMC8210572 DOI: 10.7150/jca.58887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Tumor stroma percentage (TSP), as an independent, low-cost prognostic factor, could complement current pathology and act as a more feasible risk factor for prognosis. However, TSP hadn't been applied into TNM staging. Here, the objective of our study was to investigate the prognostic significance of TSP in a robust rapid multi-dynamic approach with the application of MATLAB and threshold Algorithm for Gray Image analysis. Methods: Using a retrospective collection of 1539 CRC patients comprising three independent cohorts; one SGH cohort (N=996) and two validation cohorts (N =106, N= 437) from 2 institutions. We investigated 996 CRC of no special type. According to our established thresholds, 357 cases (35.84%) were classified as TSP-high and 639 cases (64.16%) as TSP-low. We determined the gray image area as the stromal part of the WSI and calculated the stroma percentage with our proposed method on MATLAB software. Results: In both TSP-cad(50%) and TSP-cad(median), multivariate analysis showed the TSP-cad was an independent prognostic factor for the vessel invasion and tumor location. For OS, TSP-manual HR=1.512 (95% CI 1.045-2.187); TSP-cad HR=1.443 (95% CI 0.993-2.097) and TSP-cad(median) HR=1.632 (95% CI 1.105-2.410). Fortunately, TSP-manual and TSP-cad were also found independent prognostic factor in all the cohorts. It was found that TSP-cad had slightly higher HR and wider CI than TSP-manual. Conclusions: Our research showed that TSP was an independent prognostic factor in CRC. Moreover, threshold algorithm for the quantitation of TSP could be established. In conclusion, with this Rapid multi-dynamic threshold Algorithm for Gray Image counting of TSP, which showed a higher accuracy than manual evaluation by pathologists and could be a practical method for CRC to guide clinical decision making.
Collapse
Affiliation(s)
- Tengfei Li
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China.,Graduate School of Bengbu Medical College, Bengbu 233000, China
| | - Zekuan Yu
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China.,Key Laboratory of Industrial Dust Prevention and Control & Occupational Health and Safety, Ministry of Education
| | - Yan Yang
- Graduate School of Bengbu Medical College, Bengbu 233000, China
| | - Zhongmao Fu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China
| | - Ziang Chen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Kundong Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China
| | - Zai Luo
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China
| | - Zhengjun Qiu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China
| | - Chen Huang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 201600, China
| |
Collapse
|
21
|
Wei FZ, Mei SW, Chen JN, Wang ZJ, Shen HY, Li J, Zhao FQ, Liu Z, Liu Q. Nomograms and risk score models for predicting survival in rectal cancer patients with neoadjuvant therapy. World J Gastroenterol 2020; 26:6638-6657. [PMID: 33268952 PMCID: PMC7673964 DOI: 10.3748/wjg.v26.i42.6638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer is a common digestive cancer worldwide. As a comprehensive treatment for locally advanced rectal cancer (LARC), neoadjuvant therapy (NT) has been increasingly used as the standard treatment for clinical stage II/III rectal cancer. However, few patients achieve a complete pathological response, and most patients require surgical resection and adjuvant therapy. Therefore, identifying risk factors and developing accurate models to predict the prognosis of LARC patients are of great clinical significance. AIM To establish effective prognostic nomograms and risk score prediction models to predict overall survival (OS) and disease-free survival (DFS) for LARC treated with NT. METHODS Nomograms and risk factor score prediction models were based on patients who received NT at the Cancer Hospital from 2015 to 2017. The least absolute shrinkage and selection operator regression model were utilized to screen for prognostic risk factors, which were validated by the Cox regression method. Assessment of the performance of the two prediction models was conducted using receiver operating characteristic curves, and that of the two nomograms was conducted by calculating the concordance index (C-index) and calibration curves. The results were validated in a cohort of 65 patients from 2015 to 2017. RESULTS Seven features were significantly associated with OS and were included in the OS prediction nomogram and prediction model: Vascular_tumors_bolt, cancer nodules, yN, body mass index, matchmouth distance from the edge, nerve aggression and postoperative carcinoembryonic antigen. The nomogram showed good predictive value for OS, with a C-index of 0.91 (95%CI: 0.85, 0.97) and good calibration. In the validation cohort, the C-index was 0.69 (95%CI: 0.53, 0.84). The risk factor prediction model showed good predictive value. The areas under the curve for 3- and 5-year survival were 0.811 and 0.782. The nomogram for predicting DFS included ypTNM and nerve aggression and showed good calibration and a C-index of 0.77 (95%CI: 0.69, 0.85). In the validation cohort, the C-index was 0.71 (95%CI: 0.61, 0.81). The prediction model for DFS also had good predictive value, with an AUC for 3-year survival of 0.784 and an AUC for 5-year survival of 0.754. CONCLUSION We established accurate nomograms and prediction models for predicting OS and DFS in patients with LARC after undergoing NT.
Collapse
Affiliation(s)
- Fang-Ze Wei
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Shi-Wen Mei
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Jia-Nan Chen
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Zhi-Jie Wang
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Hai-Yu Shen
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Juan Li
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Fu-Qiang Zhao
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union College, Beijing 100021, China
| |
Collapse
|
22
|
Zhao K, Li Z, Li Y, Yao S, Huang Y, Wang Y, Zhang F, Wu L, Chen X, Liang C, Liu Z. Hist-Immune signature: a prognostic factor in colorectal cancer using immunohistochemical slide image analysis. Oncoimmunology 2020; 9:1841935. [PMID: 33194320 PMCID: PMC7605350 DOI: 10.1080/2162402x.2020.1841935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Computerized image analysis for whole-slide images has been shown to improve efficiency, accuracy, and consistency in histopathology evaluations. We aimed to assess whether immunohistochemistry (IHC) image quantitative features can reflect the immune status and provide prognostic information for colorectal cancer patients. A fully automated pipeline was designed to extract histogram features from IHC digital images in a training set (N = 243). A Hist-Immune signature was generated with selected features using the LASSO Cox model. The results were validated using internal (N = 147) and external (N = 76) validation sets. The five-feature-based Hist-Immune signature was significantly associated with overall survival in training (HR 2.72, 95% CI 1.68-4.41, P < .001), internal (2.86, 1.28-6.39, 0.010), and external (2.30, 1.02-6.16, 0.044) validation sets. The full model constructed by integrating the Hist-Immune signature and clinicopathological factors had good discrimination ability (C-index 0.727, 95% CI 0.678-0.776), confirmed using internal (0.703, 0.621-0.784) and external (0.756, 0.653-0.859) validation sets. Our findings indicate that the Hist-Immune signature constructed based on the quantitative features could reflect the immune status of patients with colorectal cancer, which might advocate change in risk stratification and consequent precision medicine.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yingyi Wang
- Department of Radiology, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Fang Zhang
- School of Computer Science Engineering, South China University of Technology, Guangzhou, China
| | - Lin Wu
- Department of Pathology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
23
|
Zhang Y, Wu Q, Zhou M, Luo Z, Lv L, Pei J, Wang C, Chai B, Sui B, Huang F, Fu ZF, Zhao L. Composition of the murine gut microbiome impacts humoral immunity induced by rabies vaccines. Clin Transl Med 2020; 10:e161. [PMID: 32898335 PMCID: PMC7443138 DOI: 10.1002/ctm2.161] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gut microbiome plays a crucial role in modulating human and animal immune responses. Rabies is a fatal zoonosis causing encephalitis in mammals and vaccination is the most effective method to control and eliminate rabies. The relationship between the gut microbiome and humoral immunity post rabies vaccination has not been investigated yet. METHODS Mice orally administrated with a cocktail of broad-spectrum antibiotics were inoculated with rabies vaccines, and humoral immune response was analyzed at indicated time points. The 16S ribosomal RNA (16S rRNA) gene sequencing was performed on fecal samples from groups in vancomycin-treated and untreated mice. Mice were immunized with rabies vaccines and virus-neutralizing antibody (VNA) levels were measured, resulting in VNA high (H) and low (L) groups. Then 16S rRNA gene sequencing was performed on fecal samples from H and L group mice. RESULTS After antibiotic (Abx) treatment, mice had decreased levels of rabies virus (RABV)-specific IgM, IgG, and virus-neutralizing antibody compared with untreated mice. Abx-treated mice had fewer T follicular helper cells, germinal center B cells, and antibody secreting cells (ASCs) in lymph nodes than did untreated mice. Gut microbiome facilitated secondary immune responses by increasing the generation of ASCs. Treatment with vancomycin alone had a similarly impaired effect on the humoral immune responses compared with Abx-treated mice. From the natural population group of mice received rabies vaccines, VNA titers vary significantly and the abundance of Clostridiales and Lachnospiraceae was positively associated with the antibody titers in mice. CONCLUSIONS Our results provide the evidence that the gut microbiome impacts humoral immunity post rabies vaccination, and further investigation of the mechanism will help the development of novel adjuvants and vaccines.
Collapse
Affiliation(s)
- Yachun Zhang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Qiong Wu
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Ming Zhou
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Zhaochen Luo
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Lei Lv
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Jie Pei
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Caiqian Wang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Benjie Chai
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Baokun Sui
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Fei Huang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Zhen F. Fu
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Ling Zhao
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
24
|
Zhang F, Yao S, Li Z, Liang C, Zhao K, Huang Y, Gao Y, Qu J, Li Z, Liu Z. Predicting treatment response to neoadjuvant chemoradiotherapy in local advanced rectal cancer by biopsy digital pathology image features. Clin Transl Med 2020; 10:e110. [PMID: 32594660 PMCID: PMC7403709 DOI: 10.1002/ctm2.110] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Fang Zhang
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China.,Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhi Li
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.,Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Gao
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jinrong Qu
- Department of Radiology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospita, Yunnan Cancer Center, Kunming, Yunnan, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|