1
|
Ma H, Srivastava S, Ho SWT, Xu C, Lian BSX, Ong X, Tay ST, Sheng T, Lum HYJ, Abdul Ghani SAB, Chu Y, Huang KK, Goh YT, Lee M, Hagihara T, Ng CSY, Tan ALK, Zhang Y, Ding Z, Zhu F, Ng MSW, Joseph CRC, Chen H, Li Z, Zhao JJ, Rha SY, Teh M, Yeong J, Yong WP, So JBY, Sundar R, Tan P. Spatially Resolved Tumor Ecosystems and Cell States in Gastric Adenocarcinoma Progression and Evolution. Cancer Discov 2025; 15:767-792. [PMID: 39774838 PMCID: PMC11962405 DOI: 10.1158/2159-8290.cd-24-0605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025]
Abstract
SIGNIFICANCE Integration of spatial transcriptomic (GeoMx Digital Spatial Profiler) and single-cell RNA sequencing data from multiple gastric cancers identifies spatially resolved expression-based intratumoral heterogeneity, associated with distinct immune microenvironments. We uncovered two separate evolutionary trajectories associated with specific molecular subtypes, clinical prognoses, stromal neighborhoods, and genetic drivers. Tumor-stroma interfaces emerged as a unique state of tumor ecology.
Collapse
Affiliation(s)
- Haoran Ma
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Supriya Srivastava
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shamaine Wei Ting Ho
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chang Xu
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xuewen Ong
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Taotao Sheng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | | | | | - Yunqiang Chu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Minghui Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Takeshi Hagihara
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Clara Shi Ya Ng
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Angie Lay Keng Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yanrong Zhang
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore, Singapore
| | - Zichen Ding
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Zhu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Shu Wen Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Craig Ryan Cecil Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui Chen
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Zhen Li
- MGI Tech Singapore Pte. Ltd., Singapore, Singapore
| | - Joseph J. Zhao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, Seoul, Republic of Korea
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ming Teh
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joe Yeong
- Department of Pathology, National University Hospital, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wei Peng Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
| | - Jimmy Bok-Yan So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore, Singapore
- Division of Surgical Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Gastric Cancer Consortium, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
- Singhealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Jia G, He P, Dai T, Goh D, Wang J, Sun M, Wee F, Li F, Lim JCT, Hao S, Liu Y, Lim TKH, Ngo NT, Tao Q, Wang W, Umar A, Nashan B, Zhang Y, Ding C, Yeong J, Liu L, Sun C. Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature 2025; 640:1031-1041. [PMID: 40074893 DOI: 10.1038/s41586-025-08668-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 01/17/2025] [Indexed: 03/14/2025]
Abstract
Given the high recurrence rates of hepatocellular carcinoma (HCC) post-resection1-3, improved early identification of patients at high risk for post-resection recurrence would help to improve patient outcomes and prioritize healthcare resources4-6. Here we observed a spatial and HCC recurrence-associated distribution of natural killer (NK) cells in the invasive front and tumour centre from 61 patients. Using extreme gradient boosting and inverse-variance weighting, we developed the tumour immune microenvironment spatial (TIMES) score based on the spatial expression patterns of five biomarkers (SPON2, ZFP36L2, ZFP36, VIM and HLA-DRB1) to predict HCC recurrence risk. The TIMES score (hazard ratio = 88.2, P < 0.001) outperformed current standard tools for patient risk stratification including the TNM and BCLC systems. We validated the model in 231 patients from five multicentred cohorts, achieving a real-world accuracy of 82.2% and specificity of 85.7%. The predictive power of these biomarkers emerged through the integration of their spatial distributions, rather than individual marker expression levels alone. In vivo models, including NK cell-specific Spon2-knockout mice, revealed that SPON2 enhances IFNγ secretion and NK cell infiltration at the invasive front. Our study introduces TIMES, a publicly accessible tool for predicting HCC recurrence risk, offering insights into its potential to inform treatment decisions for early-stage HCC.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Cohort Studies
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/metabolism
- Interferon-gamma/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/cytology
- Liver Neoplasms/diagnosis
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- Mice, Knockout
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/pathology
- Reproducibility of Results
- Tumor Microenvironment
Collapse
Affiliation(s)
- Gengjie Jia
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Peiqi He
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tianli Dai
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Mengyuan Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Fuling Li
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Shuxia Hao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tony Kiat Hon Lim
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Qingping Tao
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Ahitsham Umar
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Björn Nashan
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
| | - Yongchang Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Central South University, Changsha, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Cancer Science Institute, National University of Singapore, Singapore, Singapore.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
| | - Cheng Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
3
|
Saw SPL, Takano A, Zhou S, Hlaing NO, James A, Joseph C, Lai GGY, Lim DWT, Kanesvaran R, Ang MK, Ng QS, Jain A, Tan WL, Teh YL, Tan AC, Ong BH, Lim TKH, Yeong JPS, Tan SH, Tan DSW. EGFR mutation status affects intra-tumoural heterogeneity of PD-L1 expression but not agreement between assays in resectable non-small cell lung cancer. Lung Cancer 2025; 202:108463. [PMID: 40023889 DOI: 10.1016/j.lungcan.2025.108463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The predictive value of PD-L1 to select patients for immunotherapy in resectable NSCLC remains imprecise, confounded by different assays used across trials and intra-tumoural heterogeneity (ITH). We sought to compare the concordance between 3 PD-L1 antibodies stratified by EGFR mutation status, evaluate ITH and implications on survival outcomes. METHODS Tissue microarrays were constructed from stage IA-IIIA NSCLC with 3 tumour cores per patient. Tumour proportion score (TPS) was evaluated by 3 pathologists for SP263, SP142, 22C3 and analysed in tertiles of < 1 %, 1-49 % and ≥ 50 %. ITH was defined as discordant TPS in ≥ 2/3 tumour cores. Cohen's kappa test was used to assess agreement. Survival outcomes were estimated using Kaplan-Meier. RESULTS A total of 561 patients were included, 59.5% (334/561) were EGFR-mutant. Stage IA comprised 45.5%(255/561), IB 24.1%(135/561), IIA 12.7%(71/561), IIB 4.5%(25/561) and IIIA 13.4%(75/561). Across 1683 tumour cores, SP263 and 22C3 had the highest concordance (Kappa = 0.689), followed by 22C3 and SP142 (Kappa = 0.354), then SP263 and SP142 (Kappa = 0.284), similar between EGFR-mutant and EGFR-wildtype. Agreement between pathologists was almost perfect. ITH by SP263 was observed in 14.1 % of EGFR-mutant versus 24.2 % in EGFR-wildtype(p = 0.002). Discordance was highest among TPS 1-49 % at 92.6 % (88/95) followed by ≥ 50 % at 37.8 % (14/37) and least among < 1 % at 0 % (0/429) (p < 0.001). For tumour cores scored 1-49 %, 63 %/70 % of adjacent cores were scored < 1 % for EGFR-wildtype/mutant respectively. Histological grade was the only independent predictor of PD-L1 ITH on multivariable analysis. PD-L1 ITH was not associated with survival on multivariable analysis. CONCLUSION PD-L1 scoring by SP263 and 22C3 are interchangeable but not SP142 regardless of EGFR status. PD-L1 ITH was more common in EGFR-wildtype versus EGFR-mutant tumours. Extra care should be taken to select the most representative tumour core for tumours with high histological grade or TPS 1-49% as this may influence peri-operative treatment decisions.
Collapse
Affiliation(s)
- Stephanie P L Saw
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore.
| | - Angela Takano
- Department of Pathology, Singapore General Hospital, Singapore
| | - Siqin Zhou
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | - Nwe Oo Hlaing
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Anne James
- Department of Pathology, Singapore General Hospital, Singapore
| | - Craig Joseph
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Gillianne G Y Lai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Darren W T Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Mei-Kim Ang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Quan Sing Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Amit Jain
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Wan Ling Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yi Lin Teh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | - Boon-Hean Ong
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore
| | - Tony K H Lim
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore
| | - Joe P S Yeong
- Duke-NUS Medical School, National University of Singapore, Singapore; Department of Pathology, Singapore General Hospital, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Sze Huey Tan
- Duke-NUS Medical School, National University of Singapore, Singapore; Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore; Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| |
Collapse
|
4
|
Khairi S, Wang CY, Anuraga G, Prayugo FB, Ansar M, Lesmana MHS, Irham LM, Shen CY, Chung MH. Integrative Analysis of DNA Methylation and microRNA Reveals GNPDA1 and SLC25A16 Related to Biopsychosocial Factors Among Taiwanese Women with a Family History of Breast Cancer. J Pers Med 2025; 15:134. [PMID: 40278313 PMCID: PMC12028518 DOI: 10.3390/jpm15040134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Biopsychosocial factors, including family history, influence the development of breast cancer. Malignancies in women with a family history of breast cancer may be detectable based on DNA methylation and microRNA. Objectives: The present study extended an integrative analysis of DNA methylation and microRNA to identify genes associated with biopsychosocial factors. Methods: We identified 3060 healthy women from the Taiwan Biobank and included 32 blood plasma samples for analysis of biopsychosocial factors and epigenetic changes. GEO databases and bioinformatics approaches were used for the identification and validation of potential genes. Results: Our integrative analysis revealed GNPDA1 and SLC25A16 as potential genes. Age, a family history of cancer, and alcohol consumption were associated with GNPDA1 and SLC25A16 based on the current data set and the GEO data set. GNPDA1 and SLC25A16 exhibited significant expression in breast cancer tissues based on UALCAN analysis, where they were overexpressed and underexpressed, respectively. Through a MethSurv analysis, GNPDA1 hypomethylation and SLC25A16 hypermethylation were associated with poor prognoses in terms of overall survival in breast cancer. Moreover, through a MetaCore functional enrichment analysis, GNPDA1 and SLC25A16 were associated with the BRCA1, BRCA2, and pro-oncogenic actions of the androgen receptor in breast cancer. Further, GNPDA1 and SLC25A16 were enriched in known targets of approved cancer drugs as potential genes associated with breast cancer. Conclusions: These two genes might serve as biomarkers for the early detection of breast cancer, especially for women with a family history of breast cancer.
Collapse
Affiliation(s)
- Sabiah Khairi
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan
| | - Gangga Anuraga
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Fidelia Berenice Prayugo
- Chang Gung Medical Education Research Centre (CG-MERC), Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan;
- School of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Muhamad Ansar
- Ph.D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei City 110301, Taiwan;
| | - Mohammad Hendra Setia Lesmana
- Department of Mental Health and Community, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | | | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei City 11031, Taiwan
- College of Public Health, China Medical University, Taichung City 406040, Taiwan
| | - Min-Huey Chung
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
- Department of Nursing, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
5
|
Theodorou SDP, Ntostoglou K, Nikas IP, Goutas D, Georgoulias V, Kittas C, Pateras IS. Double-Multiplex Immunostainings for Immune Profiling of Invasive Breast Carcinoma: Emerging Novel Immune-Based Biomarkers. Int J Mol Sci 2025; 26:2838. [PMID: 40243442 PMCID: PMC11988469 DOI: 10.3390/ijms26072838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
The role of tumor microenvironment in invasive breast cancer prognosis and treatment is highly appreciated. With the advent of immunotherapy, immunophenotypic characterization in primary tumors is gaining attention as it can improve patient stratification. Here, we discuss the benefits of spatial analysis employing double and multiplex immunostaining, allowing the simultaneous detection of more than one protein on the same tissue section, which in turn helps us provide functional insight into infiltrating immune cells within tumors. We focus on studies demonstrating the prognostic and predictive impact of distinct tumor-infiltrating lymphocyte subpopulations including different CD8(+) T subsets as well as CD4(+) T cells and tumor-associated macrophages in invasive breast carcinoma. The clinical value of immune cell topography is also appreciated. We further refer to how the integration of digital pathology and artificial intelligence in routine practice could enhance the accuracy of multiplex immunostainings evaluation within the tumor microenvironment, maximizing our perception of host immune response, improving in turn decision-making towards more precise immune-associated therapies.
Collapse
Affiliation(s)
- Sofia D. P. Theodorou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.D.P.T.); (K.N.); (C.K.)
| | - Konstantinos Ntostoglou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.D.P.T.); (K.N.); (C.K.)
| | - Ilias P. Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus;
| | - Dimitrios Goutas
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | | | - Christos Kittas
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.D.P.T.); (K.N.); (C.K.)
| | - Ioannis S. Pateras
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| |
Collapse
|
6
|
Andour L, Hagenaars SC, Gregus B, Tőkes AM, Karancsi Z, Tollenaar RAEM, Kroep JR, Kulka J, Mesker WE. The prognostic value of the tumor-stroma ratio compared to tumor-infiltrating lymphocytes in triple-negative breast cancer: a review. Virchows Arch 2025; 486:427-444. [PMID: 39904885 PMCID: PMC11950021 DOI: 10.1007/s00428-025-04039-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/17/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Previous literature extensively explored biomarkers to personalize treatment for breast cancer patients. The clinical need is especially high in patients with triple-negative breast cancer (TNBC) due to its aggressive nature and limited treatment modalities. This review aims to evaluate the value of tumor-infiltrating lymphocytes (TILs) and tumor-stroma ratio (TSR) as prognostic biomarkers in TNBC patients and assess their clinical potential. A literature search was conducted in PubMed, Embase, Emcare, Web of Science, and Cochrane Library. Papers comparing survival outcomes of TNBC patients with low/high or negative/positive TSR and immune cells were included. The most frequently mentioned subgroups of TILs were selected and reported in this review. Data from 43 articles on TILs and eight articles on TSR were included. Among TNBC patients, high CD8 expression was generally associated with better survival. Notable, the poor survival outcomes were related to high intra-tumoral PD-L1 expression, whereas high stromal PD-L1 expression more often was correlated with favorable outcomes. For the TSR, a high amount of stroma in the primary tumor of TNBC patients was consistently associated with worse survival. This review highlights that a high number of CD8-positive T-cells is a promising prognostic factor for TNBC patients. PD-L1 expression analyzed for intra-tumoral and stromal expression separately reports strong but contrasting information. Finally, the TSR shows potential to be an important prognostic marker, especially for TNBC patients. Utilizing both biomarkers, either on itself or combined, could enhance clinical decision-making and personalization of treatment.
Collapse
Affiliation(s)
- Layla Andour
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sophie C Hagenaars
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Barbara Gregus
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Mária Tőkes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Zsófia Karancsi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
7
|
Novakova A, Morris SA, Vaiarelli L, Frank S. Manufacturing and Financial Evaluation of Peptide-Based Neoantigen Cancer Vaccines for Triple-Negative Breast Cancer in the United Kingdom: Opportunities and Challenges. Vaccines (Basel) 2025; 13:144. [PMID: 40006691 PMCID: PMC11860436 DOI: 10.3390/vaccines13020144] [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: 12/21/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review evaluates the financial burden of current treatments for triple-negative breast cancer (TNBC) and projects potential financial scenarios to assess the feasibility of introducing a peptide-based neoantigen cancer vaccine (NCV) targeting the disease, using the UK as a healthcare system model. TNBC, the most aggressive breast cancer subtype, is associated with poor prognosis, worsened by the lack of personalised treatment options. Neoantigen cancer vaccine therapies present a personalised alternative with the potential to enhance T-cell responses independently of genetic factors, unlike approved immunotherapies for TNBC. Through a systematic literature review, the underlying science and manufacturing processes of NCVs are explored, the direct medical costs of existing TNBC treatments are enumerated, and two contrasting pricing scenarios for NCV clinical adoption are evaluated. The findings indicate that limited immunogenicity is the main scientific barrier to NCV clinical advancement, alongside production inefficiencies. Financial analysis shows that the UK spends approximately GBP 230 million annually on TNBC treatments, ranging from GBP 2200 to GBP 54,000 per patient. A best-case pricing model involving government-sponsored NCV therapy appears financially viable, while a worst-case, privately funded model exceeds the National Institute for Health and Care Excellence (NICE) cost thresholds. This study concludes that while NCVs show potential clinical benefits for TNBC, uncertainties about their standalone efficacy make their widespread adoption in the UK unlikely without further clinical research.
Collapse
Affiliation(s)
| | | | - Ludovica Vaiarelli
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 6BT, UK; (A.N.); (S.A.M.)
| |
Collapse
|
8
|
Yu X, Huang C, Song Y, Zhang C, You D, Dong X, Wu D, Meeker AK, Feng H, Wang Y. Research progress and perspectives on the application of tyramide signal amplification-based multiplex immunohistochemistry/immunofluorescence: a bibliometrics analysis. Front Oncol 2025; 14:1473414. [PMID: 39927119 PMCID: PMC11804208 DOI: 10.3389/fonc.2024.1473414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
Background and aims Multiplex immunohistochemistry/immunofluorescence (mIHC/IF), which uses the tyramide signal amplification (TSA) technique, enables sequential staining of multiple targets in formalin-fixed paraffin-embedded (FFPE) samples without worrying about cross-reactivity. This approach has received considerable attention from researchers over the past decades. This article aims to provide a bibliometric analysis of the research progress and perspectives on the application of TSA-based mIHC/IF. Methods We collected all the TSA-based mIHC/IF documents published between 2007 and 2023 from the Web of Science Core Collection (WoSCC) database. CiteSpace, VOSviewer and Bibliometrix R Package were used to perform the bibliometrics analysis, including details about annual publications, countries, institutions, authors, journals, and research topics and hotspots. Results A total of 873 relevant publications (811 articles and 62 reviews) with a time span of 17 years (2007-2023) were obtained. The number of annual publications started to increase rapidly since 2016. The United States (307, 35.17%) and the People's Republic of China (297, 34.02%) are the top two listed countries for both the number of articles produced and the citations. The University of Texas System (53, 6.07%) was the most productive institution. Integrating these results of hotspot and frontier analysis, TSA-based mIHC/IF provides significant benefits, particularly in neurology, cancer and immunology. Conclusion This study conducted a comprehensive bibliometric analysis for the use of TSA-based mIHC/IF. As TSA-based mIHC/IF and its associated imaging systems and analytic software progress, it will become the most promising tool for describing the variety of the whole tissue for a better understanding of pathological or physiological behavior.
Collapse
Affiliation(s)
- Xiaotong Yu
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Chen Huang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Yan Song
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Debo You
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - XuRan Dong
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - DeFu Wu
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Alan Keith Meeker
- Oncology Tissue and Imaging Services, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Hao Feng
- Oncology Tissue and Imaging Services, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Yuqing Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Wan Z, Cui M, Yang J, Liao D, Chen J, Li F, Xiang Y, Cui Z, Yang Y. Prognostic significance of programmed cell death 1 expression on CD8+T cells in various cancers: a systematic review and meta-analysis. Front Oncol 2025; 14:1531219. [PMID: 39876901 PMCID: PMC11772205 DOI: 10.3389/fonc.2024.1531219] [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: 11/20/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Background Increased PD-1 expression on CD8+ T cells is considered as a hallmark for T-cell exhaustion, and is thought to be related to the prognosis of cancer patients. However, discrepant results have made it difficult to apply PD-1+CD8+T cells and tumor prognosis to clinical practice. Therefore, we conducted a meta-analysis to evaluate its prognostic value in human cancers. Methods PRISMA reporting guidelines were strictly followed for conducting the current meta-analysis. The PubMed, Web of Science, Embase databases were searched from inception to November 2024. The pooled Hazard Ratio (HR) along with 95% confidence intervals (CIs) of each article were combined for the associations of PD-1+CD8+ T cells with overall survival (OS), progression- free survival (PFS) and disease-free survival(DFS). Subgroup analyses were performed for area, specimen type, cancer type, treatment, detected method and cancer stage. Results A total of 20 studies (23 cohorts, 3086 cancer patients) were included in our study. The expression PD-1+CD8+ T cells in cancer patients tended to predict poor overall survival (OS) (HR: 1.379, 95%CI: 1.084-1.753, p= 0.009), and unfavorable disease-free survival(DFS) (HR: 1.468, 95%CI: 0.931-2.316, p=0.099), though it did not reach statistical significance. Begg's and Egger's test demonstrated that no obvious publication bias was exist. Conclusions High PD-1 expression on CD8+ T cells is associated with worse survival outcomes, which can be potentially used as a prognostic marker of malignant tumor.
Collapse
Affiliation(s)
- Zhiyong Wan
- Department of General Practice, People’s Hospital of Leshan, Leshan, China
| | - Meng Cui
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Jia Yang
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Dan Liao
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Junliang Chen
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Fanmin Li
- Department of General Practice, People’s Hospital of Leshan, Leshan, China
| | - Yin Xiang
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Zhiwei Cui
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| | - Yang Yang
- Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, China
| |
Collapse
|
10
|
Yan L, Luo G, Han C, Meng J, Liang C. Exploring the oncogenic role of RGS19 in bladder cancer progression and prognosis. Acta Histochem 2024; 126:152212. [PMID: 39481225 DOI: 10.1016/j.acthis.2024.152212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024]
Abstract
This study investigates the role of autophagy-related genes (ARGs) in bladder cancer (BLCA), focusing on the regulator of G protein signaling 19 (RGS19). Using data from The Cancer Genome Atlas (TCGA) and the Human Autophagy Database (HADb), we identified RGS19 as significantly upregulated and linked to poor prognosis in BLCA. Kaplan-Meier survival analysis confirmed its association with increased mortality and. In vitro, RGS19 knockdown in BLCA cell lines inhibited proliferation, migration, and invasion, while inducing apoptosis and autophagy. Transmission electron microscopy showed autophagic structures in RGS19-silenced cells. In vivo, a xenograft mouse model demonstrated reduced tumor growth with RGS19 knockdown. Immunohistochemical (IHC) analysis revealed decreased Ki67 and increased autophagy markers in tumors with reduced RGS19. Pathway analysis suggested RGS19 acts through the cGMP-PKG signaling pathway, validated by altered expression of soluble guanylate cyclase (sGC), protein kinase G (PKG1), phosphodiesterase 5 A (PDE5A), vasodilator-stimulated phosphoprotein (VASP), and phosphorylated VASP (p-VASP) upon RGS19 knockdown. These results highlight RGS19 as a potential biomarker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Lei Yan
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Guangyue Luo
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Chengxiang Han
- Department of Urology, People's Hospital of Hanshan County, Anhui, China
| | - Jialin Meng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China.
| |
Collapse
|
11
|
Luo Y, Gao H, Zhao J, Chen L, Shao J, Ju L. The mechanism of PDE7B inhibiting the development of hepatocellular carcinoma through oxidative stress. Front Immunol 2024; 15:1469740. [PMID: 39640266 PMCID: PMC11617559 DOI: 10.3389/fimmu.2024.1469740] [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: 07/24/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
Background Liver cancer presents a significant challenge to global health and is currently ranked as the sixth most common form of cancer worldwide. Recent research indicates that phosphodiesterases play a role in various physiological and pathological processes, with a specific focus on their impact on cancer advancement. There is a scarcity of studies investigating the function and mechanisms of phosphodiesterases in the development and progression of hepatocellular carcinoma (HCC). Methods Real-time fluorescence quantitative polymerase chain reaction (qRT-PCR) and Western blotting were employed to analyze the expression of PDE7B in hepatocellular carcinoma tissues and cells. The biological role of PDE7B in HCC was investigated by both overexpressing and knocking down PDE7B in liver cancer cell lines. Furthermore, potential target proteins of PDE7B were identified through transcriptome sequencing. Results PDE7B is conspicuously reduced in tissues and cells of hepatocellular carcinoma, showing a connection with an unfavorable prognosis. Inhibiting PDE7B boosts the growth, movement, and infiltration of liver cancer cells, while its increased expression has the reverse impact. According to our trials relating to oxidative stress, PDE7B appears to control cell death in liver cancer cells by impacting the production of reactive oxygen species. Therefore, we propose that PDE7B could hinder the initiation and advancement of HCC through an oxidative stress pathway. Conclusion The research we conducted reveals that PDE7B, a gene with minimal levels of activity in hepatocellular carcinoma, possesses the capacity to inhibit the proliferation, invasion, and migration of HCC cells. PDE7B can impact the development of hepatocellular carcinoma by adjusting mechanisms related to oxidative stress.
Collapse
Affiliation(s)
- Yunfeng Luo
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People’s Hospital, Nantong, Jiangsu, China
- Medical College of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Huaide Gao
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People’s Hospital, Nantong, Jiangsu, China
- Medical College of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Jianghua Zhao
- Medical College of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Lin Chen
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People’s Hospital, Nantong, Jiangsu, China
| | - Jianguo Shao
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People’s Hospital, Nantong, Jiangsu, China
- School of Health Medicine, Nantong Institute of Technology, Nantong, Jiangsu, China
| | - Linling Ju
- Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People’s Hospital, Nantong, Jiangsu, China
- Medical College of Nantong University, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
12
|
Chen VCW, Joseph CR, Chan WOY, Sia WR, Su Q, Sam XX, Tamilarasan H, Mah YY, Ng WL, Yeong J, Wang LF, Krishnamoorthy TL, Leow WQ, Ahn M, Chow WC. Inflammasome-Driven Fatal Acute-on-Chronic Liver Failure Triggered by Mild COVID-19. Viruses 2024; 16:1646. [PMID: 39459978 PMCID: PMC11512379 DOI: 10.3390/v16101646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Inflammasome is linked to many inflammatory diseases, including COVID-19 and autoimmune liver diseases. While severe COVID-19 was reported to exacerbate liver failure, we report a fatal acute-on-chronic liver failure (ACLF) in a stable primary biliary cholangitis-autoimmune hepatitis overlap syndrome patient triggered by a mild COVID-19 infection. Postmortem liver biopsy showed sparse SARS-CoV-2-infected macrophages with extensive ASC (apoptosis-associated speck-like protein containing a CARD) speck-positive hepatocytes, correlating with elevated circulating ASC specks and inflammatory cytokines, and depleted blood monocyte subsets, indicating widespread liver inflammasome activation. This first report of a fatal inflammatory cascade in an autoimmune liver disease triggered by a mild remote viral infection hopes to elucidate a less-described pathophysiology of ACLF that could prompt consideration of new diagnostic and therapeutic options.
Collapse
Affiliation(s)
- Vivian Chih-Wei Chen
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Wharton O. Y. Chan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Wan Rong Sia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Qi Su
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Xin Xiu Sam
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Hemavathi Tamilarasan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
| | - Yun Yan Mah
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Wei Lun Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore
- Immunology & Serology Section, Department of Microbiology, Division of Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
- SingHealth Duke-NUS Global Health Institute, Singapore 169857, Singapore
| | - Thinesh L. Krishnamoorthy
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore 169608, Singapore
| | - Wei-Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Matae Ahn
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore (L.-F.W.)
- SingHealth Duke-NUS Medicine Academic Clinical Program, Singapore 168753, Singapore
- SingHealth Internal Medicine Residency Program, Singapore 169608, Singapore
| | - Wan Cheng Chow
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Medicine Academic Clinical Program, Singapore 168753, Singapore
| |
Collapse
|
13
|
Dong Y, Chen Z, Yang F, Wei J, Huang J, Long X. Prediction of immunotherapy responsiveness in melanoma through single-cell sequencing-based characterization of the tumor immune microenvironment. Transl Oncol 2024; 43:101910. [PMID: 38417293 PMCID: PMC10907870 DOI: 10.1016/j.tranon.2024.101910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/13/2024] [Accepted: 02/08/2024] [Indexed: 03/01/2024] Open
Abstract
Immune checkpoint inhibitors (ICB) therapy have emerged as effective treatments for melanomas. However, the response of melanoma patients to ICB has been highly heterogenous. Here, by analyzing integrated scRNA-seq datasets from melanoma patients, we revealed significant differences in the TiME composition between ICB-resistant and responsive tissues, with resistant or responsive tissues characterized by an abundance of myeloid cells and CD8+ T cells or CD4+ T cell predominance, respectively. Among CD4+ T cells, CD4+ CXCL13+ Tfh-like cells were associated with an immunosuppressive phenotype linked to immune escape-related genes and negative regulation of T cell activation. We also develop an immunotherapy response prediction model based on the composition of the immune compartment. Our predictive model was validated using CIBERSORTx on bulk RNA-seq datasets from melanoma patients pre- and post-ICB treatment and showed a better performance than other existing models. Our study presents an effective immunotherapy response prediction model with potential for further translation, as well as underscores the critical role of the TiME in influencing the response of melanomas to immunotherapy.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhizhuo Chen
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - Xiao Long
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
14
|
Meng J, Tan JYT, Joseph CR, Ye J, Lim JCT, Goh D, Xue Y, Lim X, Koh VCY, Wee F, Tay TKY, Chan JY, Ng CCY, Iqbal J, Lau MC, Lim HE, Toh HC, Teh BT, Dent RA, Tan PH, Yeong JPS. The Prognostic Value of CD39 as a Marker of Tumor-Specific T Cells in Triple-Negative Breast Cancer in Asian Women. J Transl Med 2024; 104:100303. [PMID: 38103870 DOI: 10.1016/j.labinv.2023.100303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a poor prognosis with limited therapeutic options available for affected patients. Efforts are ongoing to identify surrogate markers for tumor-specific CD8+ T cells that can predict the response to immune checkpoint inhibitor (ICI) therapies, such as programmed cell death protein 1 or programmed cell death ligand-1 blockade. We have previously identified tumor-specific CD39+CD8+ T cells in non-small cell lung cancer that might help predict patient responses to programmed cell death protein 1 or programmed cell death ligand-1 blockade. Based on this finding, we conducted a comparative interrogation of TNBC in an Asian cohort to evaluate the potential of CD39 as a surrogate marker of tumor-specific CD8+ T cells. Using ICI-treated TNBC mouse models (n = 24), flow cytometric analyses of peripheral blood mononuclear cells and tumor-infiltrating lymphocytes revealed that >99% of tumor-specific CD8+ T cells also expressed CD39. To investigate the relationship between CD39+CD8+ T-cell density and CD39 expression with disease prognosis, we performed multiplex immunohistochemistry staining on treatment-naive human TNBC tissues (n = 315). We saw that the proportion of CD39+CD8+ T cells in human TNBC tumors correlated with improved overall survival, as did the densities of other CD39+ immune cell infiltrates, such as CD39+CD68+ macrophages. Finally, increased CD39 expression on CD8+ T cells was also found to predict the response to ICI therapy (pembrolizumab) in a separate cohort of 11 TNBC patients. These findings support the potential of CD39+CD8+ T-cell density as a prognostic factor in Asian TNBC patients.
Collapse
Affiliation(s)
- Jia Meng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jing Ying Tira Tan
- Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Yuezhen Xue
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Xinru Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Valerie Cui Yun Koh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Timothy Kwang Yong Tay
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | | | | | - Jabed Iqbal
- Duke-NUS Medical School, Singapore, Republic of Singapore; Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Mai Chan Lau
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Hsuen Elaine Lim
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Han Chong Toh
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Bin Tean Teh
- National Cancer Centre Singapore, Singapore, Republic of Singapore
| | - Rebecca Alexandra Dent
- Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore.
| | - Puay Hoon Tan
- KK Women's and Children's Hospital, Singapore, Republic of Singapore; Luma Women's Imaging Centre/Medical Centre, Singapore, Republic of Singapore.
| | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Duke-NUS Medical School, Singapore, Republic of Singapore; National Cancer Centre Singapore, Singapore, Republic of Singapore; Department of Anatomical Pathology, Singapore General Hospital, Singapore, Republic of Singapore.
| |
Collapse
|
15
|
Ntostoglou K, Theodorou SDP, Proctor T, Nikas IP, Awounvo S, Sepsa A, Georgoulias V, Ryu HS, Pateras IS, Kittas C. Distinct profiles of proliferating CD8+/TCF1+ T cells and CD163+/PD-L1+ macrophages predict risk of relapse differently among treatment-naïve breast cancer subtypes. Cancer Immunol Immunother 2024; 73:46. [PMID: 38349444 PMCID: PMC10864422 DOI: 10.1007/s00262-024-03630-8] [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: 08/10/2023] [Accepted: 01/07/2024] [Indexed: 02/15/2024]
Abstract
Immunophenotypic analysis of breast cancer microenvironment is gaining attraction as a clinical tool improving breast cancer patient stratification. The aim of this study is to evaluate proliferating CD8 + including CD8 + TCF1 + Τ cells along with PD-L1 expressing tissue-associated macrophages among different breast cancer subtypes. A well-characterized cohort of 791 treatment-naïve breast cancer patients was included. The analysis demonstrated a distinct expression pattern among breast cancer subtypes characterized by increased CD8 + , CD163 + and CD163 + PD-L1 + cells along with high PD-L1 status and decreased fraction of CD8 + Ki67 + T cells in triple negative (TNBC) and HER2 + compared to luminal tumors. Kaplan-Meier and Cox univariate survival analysis revealed that breast cancer patients with high CD8 + , CD8 + Ki67 + , CD8 + TCF1 + cells, PD-L1 score and CD163 + PD-L1 + cells are likely to have a prolonged relapse free survival, while patients with high CD163 + cells have a worse prognosis. A differential impact of high CD8 + , CD8 + Ki67 + , CD8 + TCF1 + T cells, CD163 + PD-L1 + macrophages and PD-L1 status on prognosis was identified among the various breast cancer subtypes since only TNBC patients experience an improved prognosis compared to patients with luminal A tumors. Conversely, high infiltration by CD163 + cells is associated with worse prognosis only in patients with luminal A but not in TNBC tumors. Multivariate Cox regression analysis in TNBC patients revealed that increased CD8 + [hazard ratio (HR) = 0.542; 95% confidence interval (CI) 0.309-0.950; p = 0.032), CD8 + TCF1 + (HR = 0.280; 95% CI 0.101-0.779; p = 0.015), CD163 + PD-L1 + (HR: 0.312; 95% CI 0.112-0.870; p = 0.026) cells along with PD-L1 status employing two different scoring methods (HR: 0.362; 95% CI 0.162-0.812; p = 0.014 and HR: 0.395; 95% CI 0.176-0.884; p = 0.024) were independently linked with a lower relapse rate. Multivariate analysis in Luminal type A patients revealed that increased CD163 + was independently associated with a higher relapse rate (HR = 2.360; 95% CI 1.077-5.170; p = 0.032). This study demonstrates that the evaluation of the functional status of CD8 + T cells in combination with the analysis of immunosuppressive elements could provide clinically relevant information in different breast cancer subtypes.
Collapse
Affiliation(s)
- Konstantinos Ntostoglou
- Department of Histopathology, Biomedicine Group of Health Company, 15626, Athens, Greece
- Medical School, National and Kapodistrian University of Athens, 11527, Goudi, Athens, Greece
| | - Sofia D P Theodorou
- Medical School, National and Kapodistrian University of Athens, 11527, Goudi, Athens, Greece
| | - Tanja Proctor
- Institute of Medical Biometry, University of Heidelberg, 69120, Heidelberg, Germany
| | - Ilias P Nikas
- Medical School, University of Cyprus, 2029, Nicosia, Cyprus
| | - Sinclair Awounvo
- Institute of Medical Biometry, University of Heidelberg, 69120, Heidelberg, Germany
| | - Athanasia Sepsa
- Department of Anatomic Pathology, Metropolitan Hospital, 9 Ethnarchou Makariou & 1 E. Venizelou Street, Neo Faliro, 18547, Piraeus, Greece
| | | | - Han Suk Ryu
- Department of Pathology, College of Medicine, Seoul National University Hospital, 03080, Seoul, Republic of Korea
| | - Ioannis S Pateras
- 2nd Department of Pathology, Medical School, "Attikon" University Hospital, National and Kapodistrian University of Athens, 124 62, Athens, Greece.
| | - Christos Kittas
- Department of Histopathology, Biomedicine Group of Health Company, 15626, Athens, Greece
- Medical School, National and Kapodistrian University of Athens, 11527, Goudi, Athens, Greece
| |
Collapse
|
16
|
Rodríguez-Bejarano OH, Roa L, Vargas-Hernández G, Botero-Espinosa L, Parra-López C, Patarroyo MA. Strategies for studying immune and non-immune human and canine mammary gland cancer tumour infiltrate. Biochim Biophys Acta Rev Cancer 2024; 1879:189064. [PMID: 38158026 DOI: 10.1016/j.bbcan.2023.189064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
The tumour microenvironment (TME) is usually defined as a cell environment associated with tumours or cancerous stem cells where conditions are established affecting tumour development and progression through malignant cell interaction with non-malignant cells. The TME is made up of endothelial, immune and non-immune cells, extracellular matrix (ECM) components and signalling molecules acting specifically on tumour and non-tumour cells. Breast cancer (BC) is the commonest malignant neoplasm worldwide and the main cause of mortality in women globally; advances regarding BC study and understanding it are relevant for acquiring novel, personalised therapeutic tools. Studying canine mammary gland tumours (CMGT) is one of the most relevant options for understanding BC using animal models as they share common epidemiological, clinical, pathological, biological, environmental, genetic and molecular characteristics with human BC. In-depth, detailed investigation regarding knowledge of human BC-related TME and in its canine model is considered extremely relevant for understanding changes in TME composition during tumour development. This review addresses important aspects concerned with different methods used for studying BC- and CMGT-related TME that are important for developing new and more effective therapeutic strategies for attacking a tumour during specific evolutionary stages.
Collapse
Affiliation(s)
- Oscar Hernán Rodríguez-Bejarano
- Health Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia; Molecular Biology and Immunology Department, Fundacion Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia; PhD Programme in Biotechnology, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| | - Leonardo Roa
- Veterinary Clinic, Faculty of Agricultural Sciences, Universidad de La Salle, Carrera 7 #179-03, Bogotá 110141, Colombia
| | - Giovanni Vargas-Hernández
- Animal Health Department, Faculty of Veterinary Medicine and Zootechnics, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| | - Lucía Botero-Espinosa
- Animal Health Department, Faculty of Veterinary Medicine and Zootechnics, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| | - Carlos Parra-López
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia.
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundacion Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia; Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia.
| |
Collapse
|
17
|
Heger L, Heidkamp GF, Amon L, Nimmerjahn F, Bäuerle T, Maier A, Erber R, Hartmann A, Hack CC, Ruebner M, Huebner H, Fasching P, Beckmann MW, Dudziak D. Unbiased high-dimensional flow cytometry identified NK and DC immune cell signature in Luminal A-type and triple negative breast cancer. Oncoimmunology 2023; 13:2296713. [PMID: 38170155 PMCID: PMC10761100 DOI: 10.1080/2162402x.2023.2296713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is the most common malignancy in women worldwide and a highly heterogeneous disease. Four different subtypes are described that differ in the expression of hormone receptors as well as the growth factor receptor HER2. Treatment modalities and survival rate depend on the subtype of breast cancer. However, it is still not clear which patients benefit from immunotherapeutic approaches such as checkpoint blockade. Thus, we aimed to decipher the immune cell signature of the different breast cancer subtypes based on high-dimensional flow cytometry followed by unbiased approaches. Here, we show that the frequency of NK cells is reduced in Luminal A and B as well as triple negative breast cancer and that the phenotype of residual NK cells is changed toward regulatory CD11b-CD16- NK cells. Further, we found higher frequencies of PD-1+ CD4+ and CD8+ T cells in triple negative breast cancer. Moreover, while Luminal A-type breast cancer was enriched for CD14+ cDC2 (named type 3 DC (DC3)), CD14- cDC2 (named DC2) were more frequent in triple negative breast cancer. In contrast, HER2-enriched breast cancer did not show major alterations in the composition of the immune cell compartment in the tumor microenvironment. These findings suggest that patients with Luminal A- and B-type as well as triple negative breast cancer might benefit from immunotherapeutic approaches targeting NK cells.
Collapse
Affiliation(s)
- Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gordon F. Heidkamp
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Amon
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Maier
- Chair of Computer Science 5 (Pattern Recognition), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ramona Erber
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| | - Carolin C. Hack
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Ruebner
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hanna Huebner
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Fasching
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias W. Beckmann
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
- Comprehensive Cancer Center Central Germany Jena/Leipzig, Jena, Germany
| |
Collapse
|
18
|
Feng C, Li Y, Tai Y, Zhang W, Wang H, Lian S, Jin-Si-Han EEMBK, Liu Y, Li X, Chen Q, He M, Lu Z. A neutrophil extracellular traps-related classification predicts prognosis and response to immunotherapy in colon cancer. Sci Rep 2023; 13:19297. [PMID: 37935721 PMCID: PMC10630512 DOI: 10.1038/s41598-023-45558-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023] Open
Abstract
Neutrophil extracellular traps (NETs) have been categorized as a form of inflammatory cell death mode of neutrophils (NETosis) involved in natural immunity and the regulation of adaptive immunity. More and more studies revealed the ability of NETs to reshape the tumor immune microenvironment (TIME) by limiting antitumor effector cells, which may impair the efficacy of immunotherapy. To explore whether NETs-related genes make vital impacts on Colon carcinoma (COAD), we have carried out a systematic analysis and showed several findings in the present work. First, we obtained the patient's data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) dataset, aiming to detect two NETs-associated subtypes by consensus clustering. For the purpose of annotating the roles of NETs-related pathways, gene ontology enrichment analyses were adopted. Next, we constructed a 6 novel NETs-related genes score using the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression model. We found that the NETs risk score was notably upregulated in COAD patient samples, and its levels were notably correlated with tumor clinicopathological and immune traits. Then, according to NETs-associated molecular subtypes and the risk signature, this study compared immune cell infiltration calculated through the estimate, CIBERSORT, TIMER, ssGSEA algorithms, tumor immune dysfunction, as well as exclusion (TIDE). Furthermore, we confirm that MPO(myeloperoxidase) was significantly upregulated in COAD patient samples, and its levels were significantly linked to tumor malignancy and clinic outcome. Moreover, multiplex immunohistochemistry (mIHC) spatial analysis confirmed that MPO was closely related to Treg and PD-1 + Treg in spatial location which suggested MPO may paly an important role in TIME formation. Altogether, the obtained results indicated that a six NETs-related genes prognostic signature was conducive to estimating the prognosis and response of chemo-/immuno-therapy of COAD patients.
Collapse
Affiliation(s)
- Cheng Feng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yuan Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yi Tai
- Department of Musculoskeletal Oncology, Sun Yat-Senen University Cancer Center, Guangzhou, 510515, Guangdong, China
| | - Weili Zhang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Hao Wang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Shaopu Lian
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - E-Er-Man-Bie-Ke Jin-Si-Han
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China
| | - Yuanyuan Liu
- Department of Radiation Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Xinghui Li
- Department of Cardiology General Hospital of Xinjiang Military Command, No. 359 Youhao North Road, Saybak District, Urumqi, 830001, Xinjiang, China
| | - Qifeng Chen
- Department of Minimally Invasive Interventional Therapy, Liver Cancer Study and Service Group, Sun Yat-Sen University Cancer Center, Guangzhou, 510515, Guangdong, China.
| | - Meng He
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China.
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
19
|
Choi JE, Ahn AR, Zhang J, Kim KM, Park HS, Lee H, Chung MJ, Moon WS, Jang KY. FAM83H Expression Is Associated with Tumor-Infiltrating PD1-Positive Lymphocytes and Predicts the Survival of Breast Carcinoma Patients. Diagnostics (Basel) 2023; 13:2959. [PMID: 37761326 PMCID: PMC10529262 DOI: 10.3390/diagnostics13182959] [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: 08/09/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND FAM83H has been implicated in cancer progression, and PD1 is an important target for anti-cancer immune checkpoint therapy. Recent studies suggest an association between FAM83H expression and immune infiltration. However, studies on the roles of FAM83H and its relationship with PD1 in breast carcinomas have been limited. METHODS Immunohistochemical expression of FAM83H and PD1 and their prognostic significance were evaluated in 198 breast carcinomas. RESULTS The expression of FAM83H in cancer cells was significantly associated with the presence of PD1-positive lymphoid cells within breast carcinoma tissue. Individual and co-expression patterns of nuclear FAM83H and PD1 were significantly associated with shorter survival of breast carcinomas in univariate analysis. In multivariate analysis, the expression of nuclear FAM83H (overall survival, p < 0.001; relapse-free survival, p = 0.003), PD1 (overall survival, p < 0.001; relapse-free survival, p = 0.003), and co-expression patterns of nuclear FAM83H and PD1 (overall survival, p < 0.001; relapse-free survival, p < 0.001) were the independent indicators of overall survival and relapse-free survival of breast carcinoma patients. CONCLUSIONS This study suggests a close association between FAM83H expression and the infiltration of PD1-positive lymphoid cells in breast carcinomas and their expression as the prognostic indicators for breast carcinoma patients, and further studies are needed to clarify this relationship.
Collapse
Affiliation(s)
- Ji Eun Choi
- Department of Pathology, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Ae Ri Ahn
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
| | - Junyue Zhang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
| | - Kyoung Min Kim
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Research Institute, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Ho Sung Park
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Research Institute, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Ho Lee
- Department of Forensic Medicine, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea;
| | - Myoung Ja Chung
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Research Institute, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Research Institute, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (A.R.A.); (J.Z.); (K.M.K.); (H.S.P.); (M.J.C.); (W.S.M.)
- Research Institute of Clinical Medicine, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Research Institute, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| |
Collapse
|
20
|
Shin K, Kim J, Park SJ, Kim H, Lee MA, Kim O, Park J, Kang N, Kim IH. Early Increase in Circulating PD-1 +CD8 + T Cells Predicts Favorable Survival in Patients with Advanced Gastric Cancer Receiving Chemotherapy. Cancers (Basel) 2023; 15:3955. [PMID: 37568771 PMCID: PMC10417033 DOI: 10.3390/cancers15153955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
The clinical significance of PD-1 expression in circulating CD8+ T cells in patients with gastric cancer (GC) receiving chemotherapy remains unelucidated. Therefore, we aimed to examine its prognostic significance in blood samples of 68 patients with advanced GC who received platinum-based chemotherapy. The correlation between peripheral blood mononuclear cells, measured using fluorescence-activated cell sorting, was evaluated. Patients were divided into two groups according to the changes in PD-1+CD8+ T-cell frequencies between day 0 and 7. They were categorized as increased or decreased PD-1+CD8+ T-cell groups. The increased PD-1+CD8+ T-cell group showed longer progression-free survival (PFS) and overall survival (OS) than the decreased PD-1+CD8+ T-cell group (PFS: 8.7 months vs. 6.1 months, p = 0.007; OS: 20.7 months vs. 10.8 months, p = 0.003). The mean duration of response was significantly different between the groups (5.7 months vs. 2.5 months, p = 0.041). Multivariate analysis revealed that an increase in PD-1+CD8+ T-cell frequency was an independent prognostic factor. We concluded that the early increase in PD-1+CD8+ T-cell frequency is a potential predictor of favorable prognoses and durable responses in patients with advanced GC receiving chemotherapy.
Collapse
Affiliation(s)
- Kabsoo Shin
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.S.); (J.K.); (S.J.P.); (M.A.L.)
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Joori Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.S.); (J.K.); (S.J.P.); (M.A.L.)
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Se Jun Park
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.S.); (J.K.); (S.J.P.); (M.A.L.)
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Hyunho Kim
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Myung Ah Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.S.); (J.K.); (S.J.P.); (M.A.L.)
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Okran Kim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Juyeon Park
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - Nahyeon Kang
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.S.); (J.K.); (S.J.P.); (M.A.L.)
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (O.K.); (J.P.); (N.K.)
| |
Collapse
|
21
|
Kaya NA, Tai D, Lim X, Lim JQ, Lau MC, Goh D, Phua CZJ, Wee FYT, Joseph CR, Lim JCT, Neo ZW, Ye J, Cheung L, Lee J, Loke KSH, Gogna A, Yao F, Lee MY, Shuen TWH, Toh HC, Hilmer A, Chan YS, Lim TKH, Tam WL, Choo SP, Yeong J, Zhai W. Multimodal molecular landscape of response to Y90-resin microsphere radioembolization followed by nivolumab for advanced hepatocellular carcinoma. J Immunother Cancer 2023; 11:e007106. [PMID: 37586766 PMCID: PMC10432632 DOI: 10.1136/jitc-2023-007106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Combination therapy with radioembolization (yttrium-90)-resin microspheres) followed by nivolumab has shown a promising response rate of 30.6% in a Phase II trial (CA209-678) for advanced hepatocellular carcinoma (HCC); however, the response mechanisms and relevant biomarkers remain unknown. METHODS By collecting both pretreatment and on-treatment samples, we performed multimodal profiling of tissue and blood samples and investigated molecular changes associated with favorable responses in 33 patients from the trial. RESULTS We found that higher tumor mutation burden, NCOR1 mutations and higher expression of interferon gamma pathways occurred more frequently in responders. Meanwhile, non-responders tended to be enriched for a novel Asian-specific transcriptomic subtype (Kaya_P2) with a high frequency of chromosome 16 deletions and upregulated cell cycle pathways. Strikingly, unlike other cancer types, we did not observe any association between T-cell populations and treatment response, but tumors from responders had a higher proportion of CXCL9+/CXCR3+ macrophages. Moreover, biomarkers discovered in previous immunotherapy trials were not predictive in the current cohort, suggesting a distinctive molecular landscape associated with differential responses to the combination therapy. CONCLUSIONS This study unraveled extensive molecular changes underlying distinctive responses to the novel treatment and pinpointed new directions for harnessing combination therapy in patients with advanced HCC.
Collapse
Affiliation(s)
- Neslihan Arife Kaya
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
| | - David Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
- Duke NUS Medical School, Singapore
| | - Xinru Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jia Qi Lim
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
| | - Mai Chan Lau
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Bioinformatics Institute (BII), Agency of Science Technology and Research, Singapore
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Cheryl Zi Jin Phua
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
| | - Felicia Yu Ting Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Zhen Wei Neo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Lawrence Cheung
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Joycelyn Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
- Duke NUS Medical School, Singapore
| | - Kelvin S H Loke
- Duke NUS Medical School, Singapore
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore
| | - Apoorva Gogna
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore
| | - Fei Yao
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
| | - May Yin Lee
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
| | | | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Axel Hilmer
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Koln, Cologne, Germany
| | - Yun Shen Chan
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province, China
| | - Tony Kiat-Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore (GIS), Agency for Science(A*STAR), Technology and Research, Singapore
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
Jia K, Chen Y, Xie Y, Chong X, Li Y, Wu Y, Yuan J, Li Y, Feng X, Hu Y, Sun Y, Gong J, Zhang X, Li J, Shen L. Multidimensional immune profiling in Gastric Cancer Multiplex Immunohistochemistry Atlas from Peking University Cancer Hospital project informs PD-1/PD-L1 blockade efficacy. Eur J Cancer 2023; 189:112931. [PMID: 37343324 DOI: 10.1016/j.ejca.2023.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Immunotherapy has resulted in impressive objective response rates and durable tumour remission, but only in a subset of gastric cancer (GC) patients. The PD-L1 combined positive score is the most widely used tissue-based biomarker for anti-PD-1/PD-L1 therapy; however, this unidimensional method has limitations. Next-generation exploration of tissue-based biomarkers for GC requires characterisation of various cellular markers and key immunoregulatory molecule expression in situ. Thus, a complete, stepwise solution covering the entire process from staining samples to cross-site utilisation of pathomics data is urgently needed. METHODS With the advanced multispectral imaging analysis method, web-based data repository, and interactive sharing technology, we conducted a project entitled Gastric Cancer Multiplex Immunohistochemistry Atlas from Peking University Cancer Hospital (GMAP). We propose a standard pipeline covering sample collection, staining, scanning multispectral images, constructing a spectral library, identifying and phenotyping cells, positioning each element, and quantitatively extracting immune features. We designed an open-access relational database to explore tissue-based biomarkers to determine PD-1/PD-L1 blockade efficacy. RESULTS The GMAP project detected the functional status and spatial location of more than 50 million cells using 15 markers in 80 GC patients, based on which billions of cell pairs were recognised, highlighting the rich spatial arrangement information and the fine tumour microenvironment structure. We generated a tumour-immune atlas using the count and spatial features of 65 immune cell types. We eventually selected the indicators and built a comprehensive risk-scoring system. Patients with higher risk score showed superior immunotherapy-related progression-free survival (irPFS) (hazard ratio [HR]: 3.19; P < 0.001; median irPFS: 4.87 versus 19.87months, respectively) and immunotherapy-related overall survival (HR: 3.10; P = 0.001; median irPFS: 10.03 versus 24.87months, respectively) compared with lower risk patients, demonstrating their potential for guiding anti-PD-1/PD-L1-based immunotherapy. Importantly, an easy-to-use and versatile web server was built to promote tissue-based biomarker exploration in GC. CONCLUSION The GMAP project highlighted the clinical value of tissue-based immune features as biomarkers for immunotherapeutic decision-making. We present a well-designed, detailed workflow for the orderly generation and use of a high-quality, spatially resolved pathological database.
Collapse
Affiliation(s)
- Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yilin Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yajie Hu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
23
|
Cheung CCL, Seah YHJ, Fang J, Orpilla NHC, Lau MC, Lim CJ, Lim X, Lee JNLW, Lim JCT, Lim S, Cheng Q, Toh HC, Choo SP, Lee SY, Lee JJX, Liu J, Lim TKH, Tai D, Yeong J. Immunohistochemical scoring of LAG-3 in conjunction with CD8 in the tumor microenvironment predicts response to immunotherapy in hepatocellular carcinoma. Front Immunol 2023; 14:1150985. [PMID: 37342338 PMCID: PMC10277502 DOI: 10.3389/fimmu.2023.1150985] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Abstract
Introduction Immune checkpoint blockade (ICB) is a systemic therapeutic option for advanced hepatocellular carcinoma (HCC). However, low patient response rates necessitate the development of robust predictive biomarkers that identify individuals who will benefit from ICB. A 4-gene inflammatory signature, comprising CD8, PD-L1, LAG-3, and STAT1, was recently shown to be associated with a better overall response to ICB in various cancer types. Here, we examined whether tissue protein expression of CD8, PD-L1, LAG-3, and STAT1 predicts response to ICB in HCC. Methods HCC samples from 191 Asian patients, comprising resection specimens from 124 patients (ICB-naïve) and pre-treatment specimens from 67 advanced HCC patients treated with ICB (ICB-treated), were analyzed for CD8, PD-L1, LAG-3, and STAT1 tissue expression using multiplex immunohistochemistry followed by statistical and survival analyses. Results Immunohistochemical and survival analyses of ICB-naïve samples showed that high LAG-3 expression was associated with shorter median progression-free survival (mPFS) and overall survival (mOS). Analysis of ICB-treated samples revealed that high proportions of LAG-3+ and LAG-3+CD8+ cells pre-treatment were most closely associated with longer mPFS and mOS. Using a log-likelihood model, adding the total LAG-3+ cell proportion to the total CD8+ cell proportion significantly increased the predictive values for mPFS and mOS, compared with the total CD8+ cell proportion alone. Moreover, levels of CD8 and STAT1, but not PD-L1, were significantly correlated with better responses to ICB. After analyzing viral-related and non-viral HCC samples separately, only the LAG3+CD8+ cell proportion was significantly associated with responses to ICB regardless of viral status. Conclusion Immunohistochemical scoring of pre-treatment levels of LAG-3 and CD8 in the tumor microenvironment may help predict ICB benefits in HCC patients. Furthermore, immunohistochemistry-based techniques offer the advantage of being readily translatable in the clinical setting.
Collapse
Affiliation(s)
- Chun Chau Lawrence Cheung
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Yong Hock Justin Seah
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juntao Fang
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Mai Chan Lau
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Chun Jye Lim
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Xinru Lim
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Justina Nadia Li Wen Lee
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Sherlly Lim
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Qing Cheng
- Duke-NUS Medical School, Singapore, Singapore
- Center of Statistical Research, School of Statistics, Southwestern University of Finance and Economics, Chengdu, Sichuan, China
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Suat Ying Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joycelyn Jie Xin Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Jin Liu
- Duke-NUS Medical School, Singapore, Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - David Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joe Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
- Singapore Immunology Network (SIgN), Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
24
|
Sun GY, Zhang J, Wang BZ, Jing H, Fang H, Tang Y, Song YW, Jin J, Liu YP, Tang Y, Qi SN, Chen B, Lu NN, Li N, Li YX, Ying JM, Wang SL. The prognostic value of tumour-infiltrating lymphocytes, programmed cell death protein-1 and programmed cell death ligand-1 in Stage I-III triple-negative breast cancer. Br J Cancer 2023; 128:2044-2053. [PMID: 36966236 PMCID: PMC10205737 DOI: 10.1038/s41416-023-02218-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Tumour-infiltrating lymphocytes (TILs) represent a robust biological prognostic biomarker in triple-negative breast cancer (TNBC); however, the contribution of different subsets of immune cells is unclear. We investigated the prognostic value of immune markers, including stromal TILs (sTILs), CD8+T and FOPX3+T cells, PD-1 and PD-L1 in non-metastatic TNBC. METHODS In total, 259 patients with Stage I-III TNBC were reviewed. The density of sTILs along with the presence of total (t), stromal (s), and intratumoral (i) CD8+T cells and FOPX3+T cells were evaluated by haematoxylin and eosin and immunohistochemical staining. Immunohistochemical staining of PD-1, PD-L1 was also conducted. RESULTS All immune markers were positively correlated with each other (P < 0.05). In the multivariate analysis, sTILs (P = 0.046), tCD8+T cells (P = 0.024), iCD8+T cells (P = 0.050) and PD-1 (P = 0.039) were identified as independent prognostic factors for disease-free survival (DFS). Further analysis showed that tCD8+T cells (P = 0.026), iCD8+T cells (P = 0.017) and PD-1 (P = 0.037) increased the prognostic value for DFS beyond that of the classic clinicopathological factors and sTILs. CONCLUSIONS In addition to sTILs, inclusion of tCD8+T, iCD8+T cells, or PD-1 may further refine the prognostic model for non-metastatic TNBC beyond that including classical factors alone.
Collapse
Affiliation(s)
- Guang-Yi Sun
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Jing Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, 350014, Fuzhou, China
| | - Bing-Zhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Hao Jing
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Hui Fang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yu Tang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yong-Wen Song
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Jing Jin
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yue-Ping Liu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yuan Tang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Shu-Nan Qi
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Bo Chen
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Ning-Ning Lu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Ning Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Ye-Xiong Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Jian-Ming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - Shu-Lian Wang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
25
|
Stenmark Tullberg A, Sjöström M, Tran L, Niméus E, Killander F, Kovács A, Lundstedt D, Holmberg E, Karlsson P. Combining histological grade, TILs, and the PD-1/PD-L1 pathway to identify immunogenic tumors and de-escalate radiotherapy in early breast cancer: a secondary analysis of a randomized clinical trial. J Immunother Cancer 2023; 11:e006618. [PMID: 37208129 PMCID: PMC10201214 DOI: 10.1136/jitc-2022-006618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND The implementation of immunological biomarkers for radiotherapy (RT) individualization in breast cancer requires consideration of tumor-intrinsic factors. This study aimed to investigate whether the integration of histological grade, tumor-infiltrating lymphocytes (TILs), programmed cell death protein-1 (PD-1), and programmed death ligand-1 (PD-L1) can identify tumors with aggressive characteristics that can be downgraded regarding the need for RT. METHODS The SweBCG91RT trial included 1178 patients with stage I-IIA breast cancer, randomized to breast-conserving surgery with or without adjuvant RT, and followed for a median time of 15.2 years. Immunohistochemical analyses of TILs, PD-1, and PD-L1 were performed. An activated immune response was defined as stromal TILs ≥10% and PD-1 and/or PD-L1 expression in ≥1% of lymphocytes. Tumors were categorized as high-risk or low-risk using assessments of histological grade and proliferation as measured by gene expression. The risk of ipsilateral breast tumor recurrence (IBTR) and benefit of RT were then analyzed with 10 years follow-up based on the integration of immune activation and tumor-intrinsic risk group. RESULTS Among high-risk tumors, an activated immune infiltrate was associated with a reduced risk of IBTR (HR 0.34, 95% CI 0.16 to 0.73, p=0.006). The incidence of IBTR in this group was 12.1% (5.6-25.0) without RT and 4.4% (1.1-16.3) with RT. In contrast, the incidence of IBTR in the high-risk group without an activated immune infiltrate was 29.6% (21.4-40.2) without RT and 12.8% (6.6-23.9) with RT. Among low-risk tumors, no evidence of a favorable prognostic effect of an activated immune infiltrate was seen (HR 2.0, 95% CI 0.87 to 4.6, p=0.100). CONCLUSIONS Integrating histological grade and immunological biomarkers can identify tumors with aggressive characteristics but a low risk of IBTR despite a lack of RT boost and systemic therapy. Among high-risk tumors, the risk reduction of IBTR conferred by an activated immune infiltrate is comparable to treatment with RT. These findings may apply to cohorts dominated by estrogen receptor-positive tumors.
Collapse
Affiliation(s)
- Axel Stenmark Tullberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Martin Sjöström
- Department of Radiation Oncology, UCSF, San Francisco, California, USA
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Lena Tran
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
| | - Emma Niméus
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Fredrika Killander
- Department of Clinical Sciences Lund, Oncology/Pathology and Surgery, Lund University, Lund, Sweden
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dan Lundstedt
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Erik Holmberg
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Per Karlsson
- Department of Oncology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| |
Collapse
|
26
|
Clinical relevance of PD-1 positive CD8 T-cells in gastric cancer. Gastric Cancer 2023; 26:393-404. [PMID: 36781556 PMCID: PMC10115710 DOI: 10.1007/s10120-023-01364-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 01/11/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND We evaluated the relevance of PD-1+CD8+ T-cells in gastric cancer (GC) including prognostic significance, association with chemotherapy and immunotherapy sensitivity and correlations with the tumor microenvironment (TME). METHODS Discovery cohort: GC samples were evaluated for AE1/3, CD8, PD-1, Ki-67 and Granzyme-B expression with fluorescence-based multiplex immunohistochemistry (mIHC). Validation cohorts: we analyzed bulk RNAseq GC datasets from TCGA, the "3G" chemotherapy trial and an immunotherapy phase 2 trial. The cox proportional hazards model was used to identify factors that influenced overall survival (OS). To study the TME, we analyzed single-cell RNAseq performed on GCs. RESULTS In the discovery cohort of 350 GCs, increased PD-1 expression of CD8 T-cells was prognostic for OS (HR 0.822, p = 0.042). PD-1 expression in CD8 T-cells highly correlated with cytolytic [Granzyme-B+] (r = 0.714, p < 0.001) and proliferative [Ki-67+] (r = 0.798, p < 0.001) activity. Analysis of bulk RNAseq datasets showed tumors with high PD-1 and CD8A expression levels had improved OS when treated with immunotherapy (HR 0.117, p = 0.036) and chemotherapy (HR 0.475, p = 0.017). Analysis of an scRNAseq dataset of 152,423 cells from 40 GCs revealed that T-cell and NK-cell proportions were higher (24% vs 18% and 19% vs 15%, p < 0.0001), while macrophage proportions were lower (7% vs 11%, p < 0.0001) in CD8PD-1high compared to CD8PD-1low tumors. CONCLUSION This is one of the largest GC cohorts of mIHC combined with analysis of multiple datasets providing orthogonal validation of the clinical relevance of PD-1+CD8+ T-cells being associated with improved OS. CD8PD-1high tumors have distinct features of an immunologically active, T-cell inflamed TME.
Collapse
|
27
|
Yeong J, Goh D, Tan TJ, Tan B, Sivaraj H, Koh V, Tatt Lim JC, Joseph CR, Ye J, Yong Tay TK, Chan Lau M, Chan JY, Ng C, Iqbal J, Teh BT, Dent RA, Tan PH. Early Triple-Negative Breast Cancers in a Singapore Cohort Exhibit High PIK3CA Mutation Rates Associated With Low PD-L1 Expression. Mod Pathol 2023; 36:100056. [PMID: 36788078 DOI: 10.1016/j.modpat.2022.100056] [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: 07/20/2022] [Revised: 11/20/2022] [Accepted: 11/30/2022] [Indexed: 01/11/2023]
Abstract
Mutations in the PI3K pathway, particularly PIK3CA, were reported to be intimately associated with triple-negative breast cancer (TNBC) progression and the development of treatment resistance. We profiled PIK3CA and other genes on 166 early-stage TNBC tumors from Singapore for comparison to publicly available TNBC cohorts. These tumors were profiled transcriptionally using a NanoString panel of immune genes and multiplex immunohistochemistry, then manually scored for PD-L1-positivity using 2 clinically relevant clones, SP142 and 22C3. We discovered a higher rate of PIK3CA mutations in our TNBC cohort than in non-Asian cohorts, along with TP53, BRCA1, PTPN11, and MAP3K1 alterations. PIK3CA mutations did not affect overall or recurrence-free survival, and when compared with PIK3CAWT tumors, there were no differences in immune infiltration. Using 2 clinically approved antibodies, PIK3CAmut tumors were associated with PD-L1 negativity. Analysis of comutation frequencies further revealed that PIK3CA mutations tended to be accompanied by MAP kinase pathway mutation. The mechanism and impact of PIK3CA alterations on the TNBC tumor immune microenvironment and PD-L1 positivity warrant further study.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Tira J Tan
- Duke-NUS Medical School, Singapore; National Cancer Centre Singapore, Singapore
| | - Benedict Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | | | - Valerie Koh
- Division of Pathology, Singapore General Hospital, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Jiangfeng Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | | | - Mai Chan Lau
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | | | - Cedric Ng
- National Cancer Centre Singapore, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, Singapore
| | | | | | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore; KK Women's and Children's Hospital, Singapore; Luma Women's Imaging Centre/Medical Centre, Singapore.
| |
Collapse
|
28
|
Wang X, Guo S, Zhou H, Sun Y, Gan J, Zhang Y, Zheng W, Zhang C, Zhao X, Xiao J, Wang L, Gao Y, Ning S. Immune Pathways with Aging Characteristics Improve Immunotherapy Benefits and Drug Prediction in Human Cancer. Cancers (Basel) 2023; 15:cancers15020342. [PMID: 36672292 PMCID: PMC9856581 DOI: 10.3390/cancers15020342] [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: 11/17/2022] [Revised: 12/15/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
(1) Background: Perturbation of immune-related pathways can make substantial contributions to cancer. However, whether and how the aging process affects immune-related pathways during tumorigenesis remains largely unexplored. (2) Methods: Here, we comprehensively investigated the immune-related genes and pathways among 25 cancer types using genomic and transcriptomic data. (3) Results: We identified several pathways that showed aging-related characteristics in various cancers, further validated by conventional aging-related gene sets. Genomic analysis revealed high mutation burdens in cytokines and cytokines receptors pathways, which were strongly correlated with aging in diverse cancers. Moreover, immune-related pathways were found to be favorable prognostic factors in melanoma. Furthermore, the expression level of these pathways had close associations with patient response to immune checkpoint blockade therapy in melanoma and non-small cell lung cancer. Applying a net-work-based method, we predicted immune- and aging-related genes in pan-cancer and utilized these genes for potential immunotherapy drug discovery. Mapping drug target data to our top-ranked genes identified potential drug targets, FYN, JUN, and SRC. (4) Conclusions: Taken together, our systematic study helped interpret the associations among immune-related pathways, aging, and cancer and could serve as a resource for promoting clinical treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yue Gao
- Correspondence: (Y.G.); (S.N.)
| | | |
Collapse
|
29
|
D’Angelo A, Kilili H, Chapman R, Generali D, Tinhofer I, Luminari S, Donati B, Ciarrocchi A, Giannini R, Moretto R, Cremolini C, Pietrantonio F, Sobhani N, Bonazza D, Prins R, Song SG, Jeon YK, Pisignano G, Cinelli M, Bagby S, Urrutia AO. Immune-related pan-cancer gene expression signatures of patient survival revealed by NanoString-based analyses. PLoS One 2023; 18:e0280364. [PMID: 36649303 PMCID: PMC9844904 DOI: 10.1371/journal.pone.0280364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
The immune system plays a central role in the onset and progression of cancer. A better understanding of transcriptional changes in immune cell-related genes associated with cancer progression, and their significance in disease prognosis, is therefore needed. NanoString-based targeted gene expression profiling has advantages for deployment in a clinical setting over RNA-seq technologies. We analysed NanoString PanCancer Immune Profiling panel gene expression data encompassing 770 genes, and overall survival data, from multiple previous studies covering 10 different cancer types, including solid and blood malignancies, across 515 patients. This analysis revealed an immune gene signature comprising 39 genes that were upregulated in those patients with shorter overall survival; of these 39 genes, three (MAGEC2, SSX1 and ULBP2) were common to both solid and blood malignancies. Most of the genes identified have previously been reported as relevant in one or more cancer types. Using Cibersort, we investigated immune cell levels within individual cancer types and across groups of cancers, as well as in shorter and longer overall survival groups. Patients with shorter survival had a higher proportion of M2 macrophages and γδ T cells. Patients with longer overall survival had a higher proportion of CD8+ T cells, CD4+ T memory cells, NK cells and, unexpectedly, T regulatory cells. Using a transcriptomics platform with certain advantages for deployment in a clinical setting, our multi-cancer meta-analysis of immune gene expression and overall survival data has identified a specific transcriptional profile associated with poor overall survival.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Life Sciences, University of Bath, Bath, United Kingdom
- Oncology Department, Royal United Hospital, Bath, United Kingdom
- * E-mail:
| | - Huseyin Kilili
- Milner Centre, Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Robert Chapman
- Department of Medicine, The Princess Alexandra Hospital, Harlow, United Kingdom
| | - Daniele Generali
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charite´ University Hospital, Berlin, Germany
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS, Reggio Emilia, Italy
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | - Benedetta Donati
- Translational Research Laboratory, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Translational Research Laboratory, Azienda USL-IRCCS, Reggio Emilia, Italy
| | - Riccardo Giannini
- Department of Surgery, Clinical, Molecular and Critical Care Pathology, University of Pisa, Pisa, Italy
| | - Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Debora Bonazza
- Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Robert Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Seung Geun Song
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | | | - Mattia Cinelli
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Stefan Bagby
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Araxi O. Urrutia
- Milner Centre, Department of Life Sciences, University of Bath, Bath, United Kingdom
- Instituto de Ecologia, UNAM, Ciudad de Mexico, Mexico
| |
Collapse
|
30
|
Acar E, Esendağlı G, Yazıcı O, Dursun A. Tumor-Infiltrating Lymphocytes (TIL), Tertiary Lymphoid Structures (TLS), and Expression of PD-1, TIM-3, LAG-3 on TIL in Invasive and In Situ Ductal Breast Carcinomas and Their Relationship with Prognostic Factors. Clin Breast Cancer 2022; 22:e901-e915. [PMID: 36089459 DOI: 10.1016/j.clbc.2022.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/03/2022] [Accepted: 08/14/2022] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Immunotherapy has been determined as an important choice in breast carcinomas, especially in tumors with markedly inflammatory response. About this promising subject, tumor-infiltrating lymphocytes (TIL) and the expression of immune control point receptors on TIL have gained importance. MATERIALS AND METHODS In this study, stromal TIL and tertiary lymphoid structures (TLS) were determined in tumor tissues of 312 invasive and 68 in situ breast cancer patients. Expression rates of PD-1, LAG-3, and TIM-3 on intratumoral and stromal TIL were immunohistochemically evaluated. RESULTS In invasive breast carcinomas, stromal TIL was found to be significantly associated with lymph node metastasis, HR and HER2 expression, and basal-like phenotype, as the presence of TLS with neoadjuvant therapy, recurrence, death, and expression of HR and HER2. PD-1, LAG-3, and TIM-3 expressions were found to be associated with HR and HER2 status, stromal TIL rates, and TLS. In multivariate analysis, high stromal TIL and PD-1 expression in intratumoral TIL were found to be independent prognostic factors in terms of overall survival and disease-free survival. CONCLUSION Evaluation of TIL and immune control point receptor expressions in breast cancer is particularly important in terms of planning the therapeutic approaches based on immunotherapy protocols.
Collapse
Affiliation(s)
- Elif Acar
- Department of Medical Pathology, Ömer Halis Demir University, Niğde, Turkey.
| | - Güldal Esendağlı
- Department of Medical Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ozan Yazıcı
- Department of Medical Oncology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ayşe Dursun
- Department of Medical Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
31
|
Gao L, Jiang W, Yue Q, Ye R, Li Y, Hong J, Zhang M. Radiomic model to predict the expression of PD-1 and overall survival of patients with ovarian cancer. Int Immunopharmacol 2022; 113:109335. [DOI: 10.1016/j.intimp.2022.109335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
32
|
Pomatto-Watson LCD, Bodogai M, Carpenter M, Chowdhury D, Krishna P, Ng S, Bosompra O, Kato J, Wong S, Reyes-Sepulveda C, Bernier M, Price NL, Biragyn A, de Cabo R. Replenishment of myeloid-derived suppressor cells (MDSCs) overrides CR-mediated protection against tumor growth in a murine model of triple-negative breast cancer. GeroScience 2022; 44:2471-2490. [PMID: 35996062 PMCID: PMC9768076 DOI: 10.1007/s11357-022-00635-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/28/2022] [Indexed: 01/06/2023] Open
Abstract
Caloric restriction (CR) is the leading non-pharmacological intervention to delay induced and spontaneous tumors in pre-clinical models. These effects of CR are largely attributed to canonical inhibition of pro-growth pathways. However, our recent data suggest that CR impairs primary tumor growth and cancer progression in the murine 4T1 model of triple negative breast cancer (TNBC), at least in part, through reduced frequency of the myeloid-derived suppressor cells (MDSC). In the present study, we sought to determine whether injection of excess MDSCs could block regression in 4T1 tumor growth and metastatic spread in BALB/cJ female mice undergoing daily CR. Our findings show that MDSC injection impeded CR-mediated protection against tumor growth without increasing lung metastatic burden. Overall, these results reveal that CR can slow cancer progression by affecting immune suppressive cells.Impact statement: Inoculation of MDSCs from donor mice effectively impedes the ability of calorie restriction to protect against primary tumor growth without impacting lung metastatic burden in recipient animals.
Collapse
Affiliation(s)
- Laura C D Pomatto-Watson
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Melissa Carpenter
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Dolly Chowdhury
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Priya Krishna
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sandy Ng
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Oye Bosompra
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jonathan Kato
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sarah Wong
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Carlos Reyes-Sepulveda
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
33
|
Wen Z, Yang C, Zou D, Liu J, Wang S, Liu X, Zhang Y, Zhang Y. Pan-cancer analysis of PSAP identifies its expression and clinical relevance in gastric cancer. Pathol Res Pract 2022; 238:154027. [PMID: 36084426 DOI: 10.1016/j.prp.2022.154027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Prosaposin (PSAP) plays a critical role in sphingolipid and cancer metabolism. Reports have shown that PSAP was involved in proliferation, tumorigenesis, and metastasis. However, the expression pattern of PSAP and its prognostic roles in gastric cancer remain elusive. PSAP expression pattern and its prognostic roles in gastric cancer (GC) were explored using data from the TCGA and Kaplan-Meier Plotter. Immunohistochemical staining of GC tissues was performed to validate the prognostic role of PSAP. TISIDB was used to analyze its correlation with immunomodulators. PSAP-associated genes, PDCD1, TGFB1, and CSF1R were used to build a risk model to evaluate immunotherapy outcomes of patients with stomach adenocarcinoma (STAD). Results showed that PSAP was highly expressed in GC. High PSAP expression in GC patients also significantly indicated a poor prognosis. The results of immunohistochemical staining showed that PSAP was an independent prognostic factor in GC patients. Based on three PSAP-associated genes, a risk model that could predict the prognosis and immunotherapy outcome of STAD was bulit. PSAP was an independent prognostic factor in GC. Our results have identified three prognosis-related genes which were useful to evaluate immunotherapy outcomes of STAD patients.
Collapse
Affiliation(s)
- Zhenpeng Wen
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Dan Zou
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, Liaoning Province 110042, PR China.
| | - Jiaqing Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Song Wang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Xuqin Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Yi Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| |
Collapse
|
34
|
Mortezaee K, Majidpoor J. Extracellular vesicle-based checkpoint regulation and immune state in cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:225. [PMID: 36175741 DOI: 10.1007/s12032-022-01837-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/28/2022] [Indexed: 10/14/2022]
Abstract
Tumor cells exploit several mechanisms for hijacking an immunosuppressive tumor ecosystem in order to evade immune surveillance and to progress toward metastasis. Equipment of extracellular vesicles (EVs) with checkpoints is an example of cancer control over anti-tumor responses from immune system. Programmed death-ligand 1 (PD-L1) is a checkpoint highly expressed in a tumor at progressive stage. Interactions between PD-L1 with its receptor programmed death-1 receptor (PD-1) expressed on T cells will block the effector function of CD8+ T cells, known as one of the most important defensive cells against cancer. Evaluation of circulatory exosomal PD-L1 can be a prognostic biomarker in tumor diagnosis and responses to the immune checkpoint inhibitor (ICI) therapy, and can be considered as a tool in clinical practice for exploiting personalized therapy. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is also a checkpoint that its engagement with CD80/CD86 expressed on antigen-presenting cells (APCs), such as dendritic cells (DCs) hamper the priming phase of CD4+ and CD8+ T cells. Harvesting EVs from tumor and their modification with desired anti-checkpoint antibodies can be a promising strategy in cancer immunotherapy. The aim of this review is to discuss about EV roles in checkpoint regulation, cancer diagnosis and ICI responses, and to survey possible application of such vesicles in cancer immunotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
35
|
Goh D, Lim JCT, Fernaíndez SB, Joseph CR, Edwards SG, Neo ZW, Lee JN, Caballero SG, Lau MC, Yeong JPS. Case report: Persistence of residual antigen and RNA of the SARS-CoV-2 virus in tissues of two patients with long COVID. Front Immunol 2022; 13:939989. [PMID: 36131932 PMCID: PMC9483160 DOI: 10.3389/fimmu.2022.939989] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
The World Health Organization has defined long COVID-19 (LC) as a condition that occurs in individuals with a history of SARS-CoV-2 infection who exhibit persistent symptoms after its acute phase that last for at least two months and cannot be explained by an alternative diagnosis. Since we had previously reported residual viral antigens in tissues of convalescent patients, we aimed to assess the presence of such antigens in long COVID tissues. Here, we established the presence of the residual virus in the appendix, skin, and breast tissues of 2 patients who exhibited LC symptoms 163 and 426 days after symptom onset. With multiplex immunohistochemistry, we detected viral nucleocapsid protein in all three tissues. The nucleocapsid protein was further observed to colocalize with macrophage marker CD68, suggesting that immune cells were direct targets of SARS-CoV-2. Additionally, using RNAscope, the presence of viral RNA was also detected. Our positive finding in the breast tissue is corroborated by the recent reports of immunocompromised patients experiencing LC symptoms and persistent viral replication. Overall, our findings and emerging LC studies raise the possibility that the gastrointestinal tract may function as a reservoir for SARS-CoV-2.
Collapse
Affiliation(s)
- Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Zhen Wei Neo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Justina Nadia Lee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Mai Chan Lau
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- *Correspondence: Joe Poh Sheng Yeong,
| |
Collapse
|
36
|
Solek J, Chrzanowski J, Cieslak A, Zielinska A, Piasecka D, Braun M, Sadej R, Romanska HM. Subtype-Specific Tumour Immune Microenvironment in Risk of Recurrence of Ductal Carcinoma In Situ: Prognostic Value of HER2. Biomedicines 2022; 10:1061. [PMID: 35625798 PMCID: PMC9138378 DOI: 10.3390/biomedicines10051061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing evidence suggests that the significance of the tumour immune microenvironment (TIME) for disease prognostication in invasive breast carcinoma is subtype-specific but equivalent studies in ductal carcinoma in situ (DCIS) are limited. The purpose of this paper is to review the existing data on immune cell composition in DCIS in relation to the clinicopathological features and molecular subtype of the lesion. We discuss the value of infiltration by various types of immune cells and the PD-1/PD-L1 axis as potential markers of the risk of recurrence. Analysis of the literature available in PubMed and Medline databases overwhelmingly supports an association between densities of infiltrating immune cells, traits of immune exhaustion, the foci of microinvasion, and overexpression of HER2. Moreover, in several studies, the density of immune infiltration was found to be predictive of local recurrence as either in situ or invasive cancer in HER2-positive or ER-negative DCIS. In light of the recently reported first randomized DCIS trial, relating recurrence risk with overexpression of HER2, we also include a closing paragraph compiling the latest mechanistic data on a functional link between HER2 and the density/composition of TIME in relation to its potential value in the prognostication of the risk of recurrence.
Collapse
Affiliation(s)
- Julia Solek
- Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland; (J.S.); (A.Z.); (M.B.)
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 90-419 Lodz, Poland; (J.C.); (A.C.)
| | - Jedrzej Chrzanowski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 90-419 Lodz, Poland; (J.C.); (A.C.)
| | - Adrianna Cieslak
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 90-419 Lodz, Poland; (J.C.); (A.C.)
| | - Aleksandra Zielinska
- Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland; (J.S.); (A.Z.); (M.B.)
| | - Dominika Piasecka
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland; (J.S.); (A.Z.); (M.B.)
| | - Rafal Sadej
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Hanna M. Romanska
- Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland; (J.S.); (A.Z.); (M.B.)
| |
Collapse
|
37
|
Istl AC, Gronchi A. Neoadjuvant Therapy for Primary Resectable Retroperitoneal Sarcomas-Looking Forward. Cancers (Basel) 2022; 14:1831. [PMID: 35406603 PMCID: PMC8998126 DOI: 10.3390/cancers14071831] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 01/25/2023] Open
Abstract
The cornerstone of therapy for primary retroperitoneal sarcomas (RPS) is complete surgical resection, best achieved by resecting the tumor en bloc with adherent structures even if not overtly infiltrated. Until recently, trials designed to elucidate the role of neoadjuvant radiation or chemotherapy for RPS have been unable to achieve sufficient enrollment. The completion of the STRASS trial, which explored neoadjuvant radiotherapy for primary resectable RPS, is a major milestone in RPS research, but has prompted further questions about histology-driven treatment paradigms for RPS. Though it was ultimately a negative trial with respect to its primary endpoint of abdominal recurrence-free survival, STRASS produced a signal that suggested improved abdominal recurrence-free survival with neoadjuvant radiotherapy (RT) for patients with liposarcoma (LPS). No effect was seen for leiomyosarcoma (LMS) or high-grade dedifferentiated (DD) LPS, consistent with recent literature suggesting LMS and high-grade DD-LPS have a predominant pattern of distant rather than local failure. These results, along with those from other recent studies conducted at the bench and the bedside, emphasize the importance of a histology-specific approach to RPS research. Recent evidence for patterns of distant failure in LMS and high-grade DD-LPS has prompted the initiation of STRASS2, a study of neoadjuvant chemotherapy for these histologies. As this study unfolds, evidence may emerge for novel systemic therapy options in specific sarcoma histotypes given the explosion in targeted and immunotherapeutic applications over the last decade. This article reviews current and recent evidence around neoadjuvant radiation and chemotherapy as well as avenues for future study to optimize these treatment approaches.
Collapse
Affiliation(s)
- Alexandra C. Istl
- Division of Surgical Oncology, Johns Hopkins Hospital, Baltimore, MD 21287, USA;
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| |
Collapse
|
38
|
Ge Y, Liu H, Zhang Y, Liu J, Yan R, Xiao Z, Fan X, Huang X, An G. Inhibition of DCLK1 kinase reverses epithelial-mesenchymal transition and restores T-cell activity in pancreatic ductal adenocarcinoma. Transl Oncol 2022; 17:101317. [PMID: 34998236 PMCID: PMC8739467 DOI: 10.1016/j.tranon.2021.101317] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy has recently become a promising cancer therapy with extensive applications of immune checkpoint inhibitors (ICIs). However, pancreatic ductal adenocarcinoma (PDAC) appears to be unresponsive to immunotherapy due to the immunosuppressive microenvironment. Recent studies showed that cancer stem cell marker DCLK1 promoted the initiation and development of PDAC. Nevertheless, the mechanism driving this process remains unclear. Here, by performing gain-of-function investigations in PDAC cell lines, we demonstrate that both DCLK1 long (DCLK1-iso1, DCLK1-AS) and short (DCLK1-iso4, DCLK1-BL) isoforms can efficiently activate EMT leading to tumor migration and invasion. Consistent with experiments in vitro, bioinformatic analysis demonstrates that DCLK1 may act as a driver of EMT activation in PDAC. Further analysis showed that EMT was associated with an immunosuppressive microenvironment, which includes more immunosuppressive cells and chemokines, and patients with a higher EMT score were less sensitive to immune checkpoint inhibitors according to the TIDE (Tumor Immune Dysfunction and Exclusion) algorithm. Multiplexed immunofluorescence results demonstrated the close correlation between DCLK1, EMT and immunosuppression in PDAC patients. The findings were further confirmed in vivo reflected by decreased CD4+, CD8+ T cells and increased M2 macrophages as well as E-cad loss in DCLK1-overexpressing subcutaneous tumors. Importantly, the highly-specific DCLK1 inhibitor (DCLK1-IN-1) was able to effectively block EMT process and restore T-cell activity. Altogether, our data demonstrate that DCLK1 is strongly associated with tumor immune escape in PDAC and inhibiting DCLK1 kinase activity may be a promising therapeutic modality.
Collapse
Affiliation(s)
- Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Heshu Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuanyuan Zhang
- Department of Oncology, Beijing Huai-Rou Hospital, Beijing 101400, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Rui Yan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zeru Xiao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaona Fan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xuying Huang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
39
|
St-Denis-Bissonnette F, Khoury R, Mediratta K, El-Sahli S, Wang L, Lavoie JR. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:451. [PMID: 35053616 PMCID: PMC8773485 DOI: 10.3390/cancers14020451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and refractory subtype of breast cancer, often occurring in younger patients with poor clinical prognosis. Given the current lack of specific targets for effective intervention, the development of better treatment strategies remains an unmet medical need. Over the last decade, the field of extracellular vesicles (EVs) has grown tremendously, offering immense potential for clinical diagnosis/prognosis and therapeutic applications. While TNBC-EVs have been shown to play an important role in tumorigenesis, chemoresistance and metastasis, they could be repurposed as potential biomarkers for TNBC diagnosis and prognosis. Furthermore, EVs from various cell types can be utilized as nanoscale drug delivery systems (NDDS) for TNBC treatment. Remarkably, EVs generated from specific immune cell subsets have been shown to delay solid tumour growth and reduce tumour burden, suggesting a new immunotherapy approach for TNBC. Intrinsically, EVs can cross the blood-brain barrier (BBB), which holds great potential to treat the brain metastases diagnosed in one third of TNBC patients that remains a substantial clinical challenge. In this review, we present the most recent applications of EVs in TNBC as diagnostic/prognostic biomarkers, nanoscale drug delivery systems and immunotherapeutic agents, as well as discuss the associated challenges and future directions of EVs in cancer immunotherapy.
Collapse
Affiliation(s)
- Frederic St-Denis-Bissonnette
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Rachil Khoury
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jessie R. Lavoie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
40
|
Chong PY, Iqbal J, Yeong J, Aw TC, Chan KS, Chui P. Immune Response in Myocardial Injury: In Situ Hybridization and Immunohistochemistry Techniques for SARS-CoV-2 Detection in COVID-19 Autopsies. Front Mol Biosci 2021; 8:658932. [PMID: 34765640 PMCID: PMC8576174 DOI: 10.3389/fmolb.2021.658932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 09/28/2021] [Indexed: 01/18/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) is caused by the newly discovered coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While the lung remains the primary target site of COVID-19 injury, damage to myocardium, and other organs also contribute to the morbidity and mortality of this disease. There is also increasing demand to visualize viral components within tissue specimens. Here we discuss the cardiac autopsy findings of 12 intensive care unit (ICU) naïve and PCR-positive COVID-19 cases using a combination of histological, Immunohistochemical/immunofluorescent and molecular techniques. We performed SARS-CoV-2 qRT-PCR on fresh tissue from all cases; RNA-ISH and IHC for SARS-CoV-2 were performed on selected cases using FFPE tissue from heart. Eight of these patients also had positive post-mortem serology for SARS-CoV-2. Histopathologic changes in the coronary vessels and inflammation of the myocardium as well as in the endocardium were documented which support the reports of a cardiac component to the viral infection. As in the pulmonary reports, widespread platelet and fibrin thrombi were also identified in the cardiac tissue. In keeping with vaccine-induced activation of virus-specific CD4+ and CD8+ T cells, and release of cytokines such as interferon-gamma (IFNγ), we observed similar immune cellular distribution and cytokines in these patients. Immunohistochemical and immunofluorescent localisation for the viral Spike (S-protein) protein and the nucleocapsid protein (NP) were performed; presence of these aggregates may possibly contribute to cardiac ischemia and even remodelling.
Collapse
Affiliation(s)
- Pek Yoon Chong
- Department of Pathology, Sengkang General Hospital, Singapore, Singapore
| | - Jabed Iqbal
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joe Yeong
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
| | - Tar Choon Aw
- Department of Pathology, Sengkang General Hospital, Singapore, Singapore
| | - Kian Sing Chan
- Department of Molecular Pathology, Singapore General Hospital, Singapore, Singapore
| | - Paul Chui
- Health Science Authority, Singapore, Singapore
| |
Collapse
|
41
|
Cancer-Testis Antigens in Triple-Negative Breast Cancer: Role and Potential Utility in Clinical Practice. Cancers (Basel) 2021; 13:cancers13153875. [PMID: 34359776 PMCID: PMC8345750 DOI: 10.3390/cancers13153875] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer cells commonly express tumour-associated antigens that can induce immune responses to eradicate the tumour. Triple-negative breast cancer (TNBC) is a form of breast cancer lacking the expression of hormone receptors and cerbB2 (HER2) and tends to be more aggressive and associated with poorer prognoses due to the limited treatment options. Characterisation of biomarkers or treatment targets is thus of great significance in revealing additional therapeutic options. Cancer-testis antigens (CTAs) are tumour-associated antigens that have garnered strong attention as potential clinical biomarkers in targeted immunotherapy due to their cancer-restricted expressions and robust immunogenicity. Previous clinical studies reported that CTAs correlated with negative hormonal status, advanced tumour behaviour and a poor prognosis in a variety of cancers. Various studies also demonstrated the oncogenic potential of CTAs in cell proliferation by inhibiting cell death and inducing metastasis. Multiple clinical trials are in progress to evaluate the role of CTAs as treatment targets in various cancers. CTAs hold great promise as potential treatment targets and biomarkers in cancer, and further research could be conducted on elucidating the mechanism of actions of CTAs in breast cancer or combination therapy with other immune modulators. In the current review, we summarise the current understandings of CTAs in TNBC, addressing the role and utility of CTAs in TNBC, as well as discussing the potential applications and advantage of incorporating CTAs in clinical practise.
Collapse
|
42
|
Activated T cell-derived exosomal PD-1 attenuates PD-L1-induced immune dysfunction in triple-negative breast cancer. Oncogene 2021; 40:4992-5001. [PMID: 34172932 PMCID: PMC8342306 DOI: 10.1038/s41388-021-01896-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 12/20/2022]
Abstract
Programmed cell death 1 (PD-1) is widely expressed in tumor-infiltrating lymphocytes (TILs) of triple-negative breast cancer (TNBC). As a dominant inhibitory immune checkpoint (ICP) receptor, cell surface PD-1 is well-known to transduce negative signaling of effector T cell activity during cell–cell contact. However, despite its well-documented inhibitory effects, higher PD-1 expression in TILs is significantly associated with longer survival in TNBC patients. This phenomenon raises an interesting question whether PD-1 harbors positive activity to enhance anti-tumor immunity. Here, we show that PD-1 is secreted in an exosomal form by activated T cells and can remotely interact with either cell surface or exosomal programmed death-ligand 1 (PD-L1), induce PD-L1 internalization via clathrin-mediated endocytosis, and thereby prevent subsequent cellular PD-L1: PD-1 interaction, restoring tumor surveillance through attenuating PD-L1-induced suppression of tumor-specific cytotoxic T cell activity. Our results, through revealing an anti-PD-L1 function of exosomal PD-1, provide a positive role to enhance cytotoxic T cell activity and a potential therapeutic strategy of modifying the exosome surface with membrane-bound inhibitory ICP receptors to attenuate the suppressive tumor immune microenvironment.
Collapse
|
43
|
Immune checkpoint inhibitors for triple-negative breast cancer: From immunological mechanisms to clinical evidence. Int Immunopharmacol 2021; 98:107876. [PMID: 34146865 DOI: 10.1016/j.intimp.2021.107876] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/09/2021] [Indexed: 12/25/2022]
Abstract
Breast cancer is the most common cancer type in women worldwide. Triple-negative breast cancer (TNBC), which is characterized by the absence of estrogen receptor/progesterone receptor (ER/PR) and human epidermal growth factor receptor 2 (Her2) expressions, has a poorer prognosis compared with non-TNBC breast tumors. Until recently systemic treatment for TNBC was confined to chemotherapy owing to the lack of actionable targets. Immune checkpoint molecules are expressed on malignant cells or tumor-infiltrating immune cells and can inhibit anti-cancer immune responses. Immune checkpoint inhibitors (ICI), including anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), anti-programmed cell death protein 1 (PD-1), and anti-programmed cell death 1 ligand 1 (PD-L1), induce immune responses in different types of neoplasms. They have recently gained attention for their possible role in TNBC treatment. Several clinical trials have been conducted on the role of immune checkpoint blockade in different settings for TNBC treatment. Available evidence justifies the application of ICI and chemotherapy combination in the management of metastatic TNBC and early-stage TNBC in neoadjuvant setting. This study aims to provide information on the mechanisms of action of ICIs, review the efficacy results of clinical trials using ICIs for TNBC treatment, and assess the side effects of such drugs.
Collapse
|
44
|
Nagarajan D, Pearson J, Brentville V, Metheringham R, Pockley AG, Durrant L, McArdle SE. ImmunoBody®-HAGE derived vaccine induces immunity to HAGE and delays the growth and metastasis of HAGE-expressing tumours in vivo. Immunol Cell Biol 2021; 99:972-989. [PMID: 34105800 DOI: 10.1111/imcb.12485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 12/01/2022]
Abstract
The management of patients with triple-negative breast cancer (TNBC) continues to pose a significant clinical challenge. Less than 30% of women with metastatic TNBC survive 5 years, despite adjuvant chemotherapy and the initial higher rates of clinical response that can be achieved with neoadjuvant chemotherapy. ImmunoBody® is a plasmid DNA designed to encode a human antibody molecule with complementary determining regions (CDRs) engineered to express cytotoxic and helper T cell epitopes derived from the cancer antigen of interest. HAGE is a Cancer Testis Antigen, which is expressed in TNBC. Herein, we have identified a 30-amino-acid-long HAGE-derived sequence containing HLA-A2 and HLA-DR1 restricted epitopes and demonstrated that the use of this sequence as peptide (with CpG/IFA) or incorporated into an ImmunoBody® vaccine can generate specific IFNγ secreting splenocytes in HHDII/DR1 mice. T-cell responses elicited by the ImmunoBody®-HAGE vaccine were superior to peptide immunisation. Moreover, splenocytes from ImmunoBody®-HAGE vaccinated mice stimulated in vitro could recognise HAGE+ tumour cells and the human TNBC cell line MDA-MB-231. More importantly, the growth of implanted B16/HHDII/DR1/HAGE+ cells was significantly delayed by the ImmunoBody®-HAGE vaccine in both prophylactic and experimental metastasis settings. Overall, we demonstrate the potential of HAGE-derived vaccines for treating HAGE-expressing cancers and that such vaccines could be considered as therapeutic options for patients with HAGE+ TNBC after conventional treatment to prevent disease recurrence.
Collapse
Affiliation(s)
- Divya Nagarajan
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Joshua Pearson
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Victoria Brentville
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - Rachael Metheringham
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - A Graham Pockley
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Lindy Durrant
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - Stephanie E McArdle
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
45
|
Baram T, Erlichman N, Dadiani M, Balint-Lahat N, Pavlovski A, Meshel T, Morzaev-Sulzbach D, Gal-Yam EN, Barshack I, Ben-Baruch A. Chemotherapy Shifts the Balance in Favor of CD8+ TNFR2+ TILs in Triple-Negative Breast Tumors. Cells 2021; 10:cells10061429. [PMID: 34201054 PMCID: PMC8229590 DOI: 10.3390/cells10061429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is primarily treated via chemotherapy; in parallel, efforts are made to introduce immunotherapies into TNBC treatment. CD4+ TNFR2+ lymphocytes were reported as Tregs that contribute to tumor progression. However, our published study indicated that TNFR2+ tumor-infiltrating lymphocytes (TNFR2+ TILs) were associated with improved survival in TNBC patient tumors. Based on our analyses of the contents of CD4+ and CD8+ TILs in TNBC patient tumors, in the current study, we determined the impact of chemotherapy on CD4+ and CD8+ TIL subsets in TNBC mouse tumors. We found that chemotherapy led to (1) a reduction in CD4+ TNFR2+ FOXP3+ TILs, indicating that chemotherapy decreased the content of CD4+ TNFR2+ Tregs, and (2) an elevation in CD8+ TNFR2+ and CD8+ TNFR2+ PD-1+ TILs; high levels of these two subsets were significantly associated with reduced tumor growth. In spleens of tumor-bearing mice, chemotherapy down-regulated CD4+ TNFR2+ FOXP3+ cells but the subset of CD8+ TNFR2+ PD-1+ was not present prior to chemotherapy and was not increased by the treatment. Thus, our data suggest that chemotherapy promotes the proportion of protective CD8+ TNFR2+ TILs and that, unlike other cancer types, therapeutic strategies directed against TNFR2 may be detrimental in TNBC.
Collapse
Affiliation(s)
- Tamir Baram
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Nofar Erlichman
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Maya Dadiani
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Nora Balint-Lahat
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
| | - Anya Pavlovski
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
| | - Tsipi Meshel
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
| | - Dana Morzaev-Sulzbach
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Einav Nili Gal-Yam
- Sheba Medical Center, Breast Oncology Institute, Ramat Gan 5211401, Israel; (M.D.); (D.M.-S.); (E.N.G.-Y.)
| | - Iris Barshack
- Sheba Medical Center, Pathology Institute, Ramat Gan 5211401, Israel; (N.B.-L.); (A.P.); (I.B.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978-01, Israel
| | - Adit Ben-Baruch
- George S. Wise Faculty of Life Sciences, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv 69978-01, Israel; (T.B.); (N.E.); (T.M.)
- Correspondence: ; Tel.: +972-3-6407933 or +972-3-6405491; Fax: +972-3-6422046
| |
Collapse
|
46
|
Ying H, Lin A, Liang J, Zhang J, Luo P. Association Between FSIP2 Mutation and an Improved Efficacy of Immune Checkpoint Inhibitors in Patients With Skin Cutaneous Melanoma. Front Mol Biosci 2021; 8:629330. [PMID: 34113648 PMCID: PMC8186463 DOI: 10.3389/fmolb.2021.629330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have shown remarkable success in treating skin cutaneous melanoma (SKCM); however, the response to treatment varies greatly between patients. Considering that the efficacy of ICI treatment is influenced by many factors, we selected the Fibrosheath interacting protein 2 (FSIP2) gene and systematically analyzed its potential to predict the efficacy of ICI treatment. Methods Patient data were collected from an ICI treatment cohort (n = 120) and a The Cancer Genome Atlas (TCGA)-SKCM cohort (n = 467). The data were divided into an FSIP2-mutant (MT) group and FSIP2-wild-type (WT) group according to FSIP2 mutation status. In this study, we analyzed the patients' overall survival rate, tumor mutational burden (TMB), neoantigen load (NAL), copy number variation (CNV), cell infiltration data and immune-related genes. We used gene set enrichment analysis (GSEA) to delineate biological pathways and processes associated with the efficacy of immunotherapy. Results The efficacy of ICI treatment of SKCM patients with FSIP2 mutation was significantly better than that of patients without FSIP2 mutation. The patients in the FSIP2-MT group had higher tumor immunogenicity and lower regulatory T cell (Treg) infiltration. Results of GSEA showed that pathways related to tumor progression (MAPK and FGFR), immunomodulation, and IL-2 synthesis inhibition were significantly downregulated in the FSIP2-MT group. Conclusion Our research suggests that the FSIP2 gene has the potential to predict the efficacy of ICI treatment. The high tumor immunogenicity and low Treg levels observed may be closely related to the fact that patients with FSIP2-MT can benefit from ICI treatment.
Collapse
Affiliation(s)
- Haoxuan Ying
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Liang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Li X, Cao Y, Yu X, Jin F, Li Y. A novel autophagy-related genes prognostic risk model and validation of autophagy-related oncogene VPS35 in breast cancer. Cancer Cell Int 2021; 21:265. [PMID: 34001111 PMCID: PMC8130280 DOI: 10.1186/s12935-021-01970-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/30/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Accumulating evidence implies that autophagy plays a critical role in breast cancer development and progression. It is crucial to screen out autophagy-related encoding genes (ARGs) with prognostic value in breast cancer and reveal their biological properties in the aggressiveness of breast cancer. METHODS Univariate and multivariate Cox proportional hazards analyses were used to identify a prognostic risk model of ARGs from The Cancer Genome Atlas (TCGA). Kaplan-Meier analysis, univariate and multivariate Cox regression analyses and receiver operating characteristic (ROC) curve analysis were performed to validate the risk model. Western blot and immunohistochemistry (IHC) were conducted to assess the expression of VPS35 (one of ARGs in risk model). CCK8, Colony formation assay, Transwell migration/invasion assays and autophagy flux assay were used to confirm biological function of VPS35 in breast cancer. RESULTS In this study, the prognostic risk model consisting of six ARGs (VPS35, TRIM21, PRKAB2, RUFY4, MAP1LC3A and LARP1) in breast cancer were identified. The risk model was further verified as a novel independent prognostic factor for breast cancer patients. We also clarified that vacuolar protein sorting-associated protein 35 (VPS35), one of ARGs in the risk model, was upregulated in breast cancer samples and cell lines. VPS35 overexpression was correlated with more aggressive phenotype of breast cancer and indicated worse prognosis in both progression-free survival and overall survival analyses. Meanwhile, VPS35 knockdown inhibited breast cancer cell proliferation, migration and invasion, suggesting that VPS35 promoted the progression of breast cancer. VPS35 silence also influenced autophagy process, indicating that VPS35 was essential for autophagy completion. CONCLUSION Taken together, the six ARGs risk model has a remarkably prognostic value for breast cancer. Among them, VPS35 might exert as a significant oncogenic and prognostic factor for breast cancer and could be a promising autophagy-related therapeutic target in clinical practice.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, Shenyang, 110001, China.,Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yu Cao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, Shenyang, 110001, China
| | - Xinmiao Yu
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, Shenyang, 110001, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, Shenyang, 110001, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
48
|
Yeong J, Suteja L, Simoni Y, Lau KW, Tan AC, Li HH, Lim S, Loh JH, Wee FYT, Nerurkar SN, Takano A, Tan EH, Lim TKH, Newell EW, Tan DSW. Intratumoral CD39 +CD8 + T Cells Predict Response to Programmed Cell Death Protein-1 or Programmed Death Ligand-1 Blockade in Patients With NSCLC. J Thorac Oncol 2021; 16:1349-1358. [PMID: 33975004 DOI: 10.1016/j.jtho.2021.04.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/09/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Programmed cell death protein-1 (PD-1) and programmed death-ligand 1 (PD-L1) blockade is currently widely used in the treatment of metastatic NSCLC. Despite available biomarker stratification, clinical responses vary. Thus, the search for novel biomarkers with improved response prediction is ongoing. Previously, using mass cytometry or cytometry by time-of-flight (CyTOF), our group demonstrated that CD39+CD8+ immune cells represent tumor antigen-specific, cytotoxic T cells in treatment-naive NSCLC. We hypothesized that accurate quantitation of this T cell subset would predict immunotherapy outcome. METHODS To translate this to a clinical setting, the present study compared CyTOF data with a range of clinically relevant methods, including conventional immunohistochemistry (IHC), multiplex IHC or immunofluorescence (mIHC), and gene expression assay by NanoString. RESULTS Quantification using mIHC but not conventional IHC or NanoString correlated with the CyTOF results. The specificity and sensitivity of mIHC were then evaluated in a separate retrospective NSCLC cohort. CD39+CD8+ T cell proportion, as determined by mIHC, successfully stratified responders and nonresponders to PD-1 or PD-L1 inhibitors (objective response rate of 63.6%, compared with 0% for the negative group). This predictive capability was independent from other confounding factors, such as total CD8+ T cell proportion, CD39+ lymphocyte proportion, PD-L1 positivity, EGFR mutation status, and other clinicopathologic parameters. CONCLUSIONS Our results suggest that the mIHC platform is a clinically relevant method to evaluate CD39+CD8+ T cell proportion and that this marker can serve as a potential biomarker that predicts response to PD-1 or PD-L1 blockade in patients with NSCLC. Further validation in additional NSCLC cohorts is warranted.
Collapse
Affiliation(s)
- Joe Yeong
- Institute of Molecular Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore; Division of Pathology, Singapore General Hospital, Singapore
| | - Lisda Suteja
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Yannick Simoni
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kah Weng Lau
- Institute of Molecular Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore; Division of Pathology, Singapore General Hospital, Singapore
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Hui Hua Li
- Division of Medicine, Singapore General Hospital, Singapore; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Sherlly Lim
- Institute of Molecular Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Jie Hua Loh
- Division of Pathology, Singapore General Hospital, Singapore
| | - Felicia Y T Wee
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Angela Takano
- Division of Pathology, Singapore General Hospital, Singapore
| | - Eng Huat Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Tony K H Lim
- Division of Pathology, Singapore General Hospital, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
49
|
Tien TZ, Lee JNLW, Lim JCT, Chen XY, Thike AA, Tan PH, Yeong JPS. Delineating the breast cancer immune microenvironment in the era of multiplex immunohistochemistry/immunofluorescence. Histopathology 2021; 79:139-159. [PMID: 33400265 DOI: 10.1111/his.14328] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common malignancy and the leading cause of cancer death in females worldwide. Treatment is challenging, especially for those who are triple-negative. Increasing evidence suggests that diverse immune populations are present in the breast tumour microenvironment, which opens up avenues for personalised drug targets. Historically, our investigations into the immune constitution of breast tumours have been restricted to analyses of one or two markers at a given time. Recent technological advances have allowed simultaneous labelling of more than 35 markers and detailed profiling of tumour-immune infiltrates at the single-cell level, as well as determining the cellular composition and spatial analysis of the entire tumour architecture. In this review, we describe emerging technologies that have contributed to the field of breast cancer diagnosis, and discuss how to interpret the vast data sets obtained in order to effectively translate them for clinically relevant use.
Collapse
Affiliation(s)
- Tracy Z Tien
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Justina N L W Lee
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jeffrey C T Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xiao-Yang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joe P S Yeong
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
50
|
Graeser M, Feuerhake F, Gluz O, Volk V, Hauptmann M, Jozwiak K, Christgen M, Kuemmel S, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Kolberg-Liedtke C, Kates R, Wuerstlein R, Nitz U, Kreipe HH, Harbeck N. Immune cell composition and functional marker dynamics from multiplexed immunohistochemistry to predict response to neoadjuvant chemotherapy in the WSG-ADAPT-TN trial. J Immunother Cancer 2021; 9:e002198. [PMID: 33963012 PMCID: PMC8108653 DOI: 10.1136/jitc-2020-002198] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The association of early changes in the immune infiltrate during neoadjuvant chemotherapy (NACT) with pathological complete response (pCR) in triple-negative breast cancer (TNBC) remains unexplored. METHODS Multiplexed immunohistochemistry was performed in matched tumor biopsies obtained at baseline and after 3 weeks of NACT from 66 patients from the West German Study Group Adjuvant Dynamic Marker-Adjusted Personalized Therapy Trial Optimizing Risk Assessment and Therapy Response Prediction in Early Breast Cancer - Triple Negative Breast Cancer (WSG-ADAPT-TN) trial. Association between CD4, CD8, CD73, T cells, PD1-positive CD4 and CD8 cells, and PDL1 levels in stroma and/or tumor at baseline, week 3 and 3-week change with pCR was evaluated with univariable logistic regression. RESULTS Compared with no change in immune cell composition and functional markers, transition from 'cold' to 'hot' (below-median and above-median marker level at baseline, respectively) suggested higher pCR rates for PD1-positive CD4 (tumor: OR=1.55, 95% CI 0.45 to 5.42; stroma: OR=2.65, 95% CI 0.65 to 10.71) and PD1-positive CD8 infiltrates (tumor: OR=1.77, 95% CI 0.60 to 5.20; stroma: OR=1.25, 95% CI 0.41 to 3.84; tumor+stroma: OR=1.62, 95% CI 0.51 to 5.12). No pCR was observed after 'hot-to-cold' transition in PD1-positive CD8 cells. pCR rates appeared lower after hot-to-cold transitions in T cells (tumor: OR=0.26, 95% CI 0.03 to 2.34; stroma: OR=0.35, 95% CI 0.04 to 3.25; tumor+stroma: OR=0.00, 95% CI 0.00 to 1.04) and PD1-positive CD4 cells (tumor: OR=0.60, 95% CI 0.11 to 3.35; stroma: OR=0.22, 95% CI 0.03 to 1.92; tumor+stroma: OR=0.32, 95% CI 0.04 to 2.94). Higher pCR rates collated with 'altered' distribution (levels below-median and above-median in tumor and stroma, respectively) of T cell (OR=3.50, 95% CI 0.84 to 14.56) and PD1-positive CD4 cells (OR=4.50, 95% CI 1.01 to 20.14). CONCLUSION Our exploratory findings indicate that comprehensive analysis of early immune infiltrate dynamics complements currently investigated predictive markers for pCR and may have a potential to improve guidance for individualized de-escalation/escalation strategies in TNBC.
Collapse
Affiliation(s)
- Monika Graeser
- West German Study Group, Moenchengladbach, Germany
- Breast Center Niederrhein, Bethesda Protestant Hospital Monchengladbach, Monchengladbach, Germany
- Department of Gynecology, University Medical Center Hamburg, Hamburg, Germany
| | - Friedrich Feuerhake
- Institute of Pathology, Medical School Hannover, Hannover, Germany
- Institute of Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany
- Breast Center Niederrhein, Bethesda Protestant Hospital Monchengladbach, Monchengladbach, Germany
- University Clinics Cologne, Cologne, Germany
| | - Valery Volk
- Institute of Pathology, Medical School Hannover, Hannover, Germany
| | - Michael Hauptmann
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Katarzyna Jozwiak
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | | | - Sherko Kuemmel
- West German Study Group, Moenchengladbach, Germany
- Breast Unit, Kliniken Essen-Mitte, Essen, Germany
- University Hospital Charité, Humboldt University, Berlin, Germany
| | | | | | - Michael Braun
- Breast Center, Rotkreuz Clinics Munich, Munich, Germany
| | - Mathias Warm
- Breast Center, City Hospital Holweide, Cologne, Germany
| | | | | | - Bahriye Aktas
- Women's Clinic, University Clinics Essen, Essen, Germany
- Women's Clinic, University Clinics Leipzig, Leipzig, Germany
| | | | - Cornelia Kolberg-Liedtke
- University Hospital Charité, Humboldt University, Berlin, Germany
- Women's Clinic, University Clinics Essen, Essen, Germany
| | - Ronald Kates
- West German Study Group, Moenchengladbach, Germany
| | - Rachel Wuerstlein
- West German Study Group, Moenchengladbach, Germany
- Breast Center, Department of Gynecology and Obstetrics and CCCLMU, LMU University Hospital, Munich, Germany
| | - Ulrike Nitz
- West German Study Group, Moenchengladbach, Germany
- Breast Center Niederrhein, Bethesda Protestant Hospital Monchengladbach, Monchengladbach, Germany
| | | | - Nadia Harbeck
- West German Study Group, Moenchengladbach, Germany
- Breast Center, Department of Gynecology and Obstetrics and CCCLMU, LMU University Hospital, Munich, Germany
| |
Collapse
|