1
|
Guo J, Si G, Song X, Si F. Association of B cells and the risk of Esophageal cancer: a bidirectional two-sample mendelian randomization study. BMC Cancer 2024; 24:1416. [PMID: 39550560 DOI: 10.1186/s12885-024-13166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Currently, research on the role of B cells in esophageal cancer (EC) is limited, and existing studies on their impact are controversial. Therefore, this study was conducted to elucidate the complex causal relationship between B cells and EC, expand the understanding of esophageal cancer immunology. METHODS Bidirectional two-sample Mendelian randomization (MR) was performed to assess the causal relationships between 190 B cell phenotypes and EC. To complement the MR analysis, Bayesian Weighted Mendelian Randomization (BWMR) was employed, and sensitivity analyses were conducted to evaluate the robustness of the findings. Positive results were further validated in independent cohorts of esophageal cancer studies. In addition, RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) were utilized for validation, incorporating B cell-related gene expression analysis and functional enrichment analysis to support the MR findings. RESULTS In the primary analysis, significant causal relationships were observed between 5 B cell types and the risk of EC; the onset of EC was causally linked to 3 B cell phenotypes. Validation in other cohorts revealed that 4 outcomes aligned with the primary analysis, included were CD19 on IgD + CD38-, CD20 on IgD- CD27-, CD20 on IgD- CD38br, and CD38 on PB/PC. Further validation using RNA-seq data showed that CD38 mRNA was significantly overexpressed in EC tissues, whereas CD19 and MS4A1 mRNA levels did not differ significantly between tumor and normal tissues. Functional enrichment analysis revealed that CD19, MS4A1, and CD38 are involved in multiple tumor-related immune pathways, suggesting their pivotal role in regulating the tumor immune microenvironment. CONCLUSIONS Our study suggests a potential connection between B cell phenotypes and EC through bidirectional two-sample MR combined with BWMR analysis, providing a preliminary basis for future research.
Collapse
Affiliation(s)
- Jinzhou Guo
- Academy of Zhongjing, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
- Laboratory of TCM Syndrome and Prescription Signaling, Academy of zhongjing, Zhengzhou, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint, Zhengzhou, China
| | - Gao Si
- Department of Orthopedic, Peking University Third Hospital, Beijing, China
| | - Xuejie Song
- Academy of Zhongjing, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
- Laboratory of TCM Syndrome and Prescription Signaling, Academy of zhongjing, Zhengzhou, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint, Zhengzhou, China
| | - Fuchun Si
- Academy of Zhongjing, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China.
- Laboratory of TCM Syndrome and Prescription Signaling, Academy of zhongjing, Zhengzhou, China.
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint, Zhengzhou, China.
| |
Collapse
|
2
|
Guo J, Si G, Si F. Association of immune cells and the risk of esophageal cancer: A Mendelian randomization study in a East Asian population. Medicine (Baltimore) 2024; 103:e38064. [PMID: 38701252 PMCID: PMC11062746 DOI: 10.1097/md.0000000000038064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Immunotherapy has been used in esophageal cancer (EC), but the causal relationship between EC and immune cells is not clear. Although the cellular phenotype has been reported as a biomarker for immunotherapy, the biomarker studies for immunotherapy in EC still face great challenges. Comprehensive 2-sample Mendelian randomization (MR) analysis was performed to determine the causal association between immune cell signatures and EC in this study. Based on publicly available genetic data, we explored causal associations between 731 immune cell signatures and EC risk. EC had no statistically significant effect on immunophenotypes. Nine immunophenotype types were positively associated with the risk of EC: CD20-%B cell, CD20% lymphocytes, CD25 on IgD- CD27-, CD25 on IgD+ CD24+, CD27 on IgD+ CD24+, CD28+ CD45RA- CD8br AC, CD3 on TD CD8br, IgD-CD38dim%B cells, and Mo MDSC AC. In addition, a total of 15 immunophenotypes were identified as causally associated with EC. IgD+ CD38- %B cell, IgD- CD24- %lymphocyte, CD19 on IgD- CD38dim, CD20 on IgD+ CD24+, CD62L-myeloid DC AC, CD4+ AC, Lymphocyte %leukocyte, CD3 on HLA-DR+ T cell, CD3 on CD45RA- CD4+, HVEM on naive CD4+ AC, HVEM on CD45RA- CD4+, CD4 on TD CD4+, CD4 on CD4 Treg, and CD4 on CD39+ resting Treg, and CD4 on activated & secreting Treg. Our study has demonstrated the close connection between immune cells and EC by genetic means, thus providing guidance for future clinical research.
Collapse
Affiliation(s)
- Jinzhou Guo
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Laboratory of TCM Syndrome and Prescription Signaling, Academy of Zhongjing, Zhengzhou, Henan, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint, Zhengzhou, Henan, China
| | - Gao Si
- Department of Orthopedic, Peking University Third Hospital, Beijing, China
| | - Fuchun Si
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Laboratory of TCM Syndrome and Prescription Signaling, Academy of Zhongjing, Zhengzhou, Henan, China
- Henan Key Laboratory of TCM Syndrome and Prescription Signaling, Henan International Joint, Zhengzhou, Henan, China
| |
Collapse
|
3
|
Sundar R, Huang KK, Kumar V, Ramnarayanan K, Demircioglu D, Her Z, Ong X, Bin Adam Isa ZF, Xing M, Tan ALK, Tai DWM, Choo SP, Zhai W, Lim JQ, Das Thakur M, Molinero L, Cha E, Fasso M, Niger M, Pietrantonio F, Lee J, Jeyasekharan AD, Qamra A, Patnala R, Fabritius A, De Simone M, Yeong J, Ng CCY, Rha SY, Narita Y, Muro K, Guo YA, Skanderup AJ, So JBY, Yong WP, Chen Q, Göke J, Tan P. Epigenetic promoter alterations in GI tumour immune-editing and resistance to immune checkpoint inhibition. Gut 2022; 71:1277-1288. [PMID: 34433583 PMCID: PMC9185816 DOI: 10.1136/gutjnl-2021-324420] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Epigenomic alterations in cancer interact with the immune microenvironment to dictate tumour evolution and therapeutic response. We aimed to study the regulation of the tumour immune microenvironment through epigenetic alternate promoter use in gastric cancer and to expand our findings to other gastrointestinal tumours. DESIGN Alternate promoter burden (APB) was quantified using a novel bioinformatic algorithm (proActiv) to infer promoter activity from short-read RNA sequencing and samples categorised into APBhigh, APBint and APBlow. Single-cell RNA sequencing was performed to analyse the intratumour immune microenvironment. A humanised mouse cancer in vivo model was used to explore dynamic temporal interactions between tumour kinetics, alternate promoter usage and the human immune system. Multiple cohorts of gastrointestinal tumours treated with immunotherapy were assessed for correlation between APB and treatment outcomes. RESULTS APBhigh gastric cancer tumours expressed decreased levels of T-cell cytolytic activity and exhibited signatures of immune depletion. Single-cell RNAsequencing analysis confirmed distinct immunological populations and lower T-cell proportions in APBhigh tumours. Functional in vivo studies using 'humanised mice' harbouring an active human immune system revealed distinct temporal relationships between APB and tumour growth, with APBhigh tumours having almost no human T-cell infiltration. Analysis of immunotherapy-treated patients with GI cancer confirmed resistance of APBhigh tumours to immune checkpoint inhibition. APBhigh gastric cancer exhibited significantly poorer progression-free survival compared with APBlow (median 55 days vs 121 days, HR 0.40, 95% CI 0.18 to 0.93, p=0.032). CONCLUSION These findings demonstrate an association between alternate promoter use and the tumour microenvironment, leading to immune evasion and immunotherapy resistance.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore .,Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore.,Singapore Gastric Cancer Consortium, Singapore
| | - Kie-Kyon Huang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Vikrant Kumar
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | | | - Deniz Demircioglu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Xuewen Ong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Zul Fazreen Bin Adam Isa
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore,Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore,Diagnostic Development Hub (DxD), Agency for Science, Technology and Research, Singapore
| | - Manjie Xing
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore,Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore,Diagnostic Development Hub (DxD), Agency for Science, Technology and Research, Singapore
| | - Angie Lay-Keng Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | | | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre, Singapore,Curie Oncology, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Jia Qi Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Meghna Das Thakur
- Department of Development Sciences, Genentech, San Francisco, California, USA
| | - Luciana Molinero
- Department of Development Sciences, Genentech, San Francisco, California, USA
| | - Edward Cha
- Department of Development Sciences, Genentech, San Francisco, California, USA
| | - Marcella Fasso
- Department of Development Sciences, Genentech, San Francisco, California, USA
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Anand D Jeyasekharan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Aditi Qamra
- Statistical Programming and Analytics, Roche Canada, Mississauga, Ontario, Canada,University Health Network, Toronto, Ontario, Canada
| | | | | | | | - Joe Yeong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Cedric Chuan Young Ng
- Laboratory of Cancer Epigenome, Department of Medical Sciences, National Cancer Centre, Singapore
| | - Sun Young Rha
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea,Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Yukiya Narita
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yu Amanda Guo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | | | - Jimmy Bok Yan So
- Singapore Gastric Cancer Consortium, Singapore,Department of Surgery, National University Hospital, Singapore,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore,Singapore Gastric Cancer Consortium, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jonathan Göke
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore .,Singapore Gastric Cancer Consortium, Singapore.,Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
4
|
Güler İ, Kuzucu İ, Baklacı D, Kum RO, Özhamam Uçaryılmaz E, Özcan M. Evaluation of Nivolumab for Ototoxic Effects: An Animal Study in Rats. J Int Adv Otol 2021; 16:218-221. [PMID: 32510456 DOI: 10.5152/iao.2020.7123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES The aim of this study was to assess whether nivolumab is ototoxic in rats and whether this ototoxicity is dose-dependent. MATERIALS AND METHODS Twelve rats were divided into two groups: Group 1 (control group, 6 rats, 12 ears) received intraperitoneal saline for 14 days. Group 2 (study group, 6 rats, 12 ears) and received two doses of 3 mg/kg intraperitoneal nivolumab within 14 days. Auditory brainstem responses (ABRs) were performed preoperatively and 4 and 8 weeks postoperatively. We compared between the groups, morphologic appearance of spiral ganglion cells and organ of Corti and density of spiral ganglion cells (measured with conventional light microscope connected to a personal computer). RESULTS In our control group, both spiral ganglion and organ of corti had a normal morphological appearance. In our study group, spiral ganglion cells had a normal morphological appearance. However, some sections showed possibly mild degenerative changes in the organ of corti. Of 12 samples in the study group, four had a significant loss of density of spiral ganglion cells compared to the control group. The baseline ABR thresholds did not significantly differ between the groups (p=0.713). There was no statistically significant difference between the groups regarding ABR thresholds at week 4 (p=0.347). However, a statistically significant difference was observed in the ABR thresholds at week 8 (p=0.045). CONCLUSION The results of our study showed that nivolumab treatment has ototoxic effects. Based on our results, we recommend monitoring the changes in the hearing ability of chemotherapy patients.
Collapse
Affiliation(s)
- İsmail Güler
- Deparment of Otorhinolaryngology, Ankara Medipol University School of Medicine, Ankara, Turkey
| | - İhsan Kuzucu
- Department of Otorhinolaryngology, Aksaray University School of Medicine, Aksaray, Turkey
| | - Deniz Baklacı
- Clinic of Otorhinolaryngology, Kahramankazan State Hospital, Ankara, Turkey
| | - Rauf Oğuzhan Kum
- Clinic of Otorhinolaryngology, Ankara City Hospital, Ankara, Turkey
| | | | - Müge Özcan
- Clinic of Otorhinolaryngology, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
5
|
You W, Shang B, Sun J, Liu X, Su L, Jiang S. Mechanistic insight of predictive biomarkers for antitumor PD‑1/PD‑L1 blockade: A paradigm shift towards immunome evaluation (Review). Oncol Rep 2020; 44:424-437. [PMID: 32627031 PMCID: PMC7336519 DOI: 10.3892/or.2020.7643] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
Checkpoint inhibitor-based immunotherapy has exhibited unprecedented success in the treatment of advanced-stage cancer in recent years. Several therapeutic antibodies targeting programmed death-1 (PD-1) or its ligand (PD-L1) have received regulatory approvals for the treatment of multiple malignancies, including melanoma, non-small cell lung cancer, kidney cancer and Hodgkin's lymphoma. However, a substantial proportion of patients still do not benefit from these agents, let alone the risk of immune-associated toxicities and financial burden. Therefore, it is imperative to identify valid predictive biomarkers which can help optimize the selection of patients. In this review, a mechanism-based interpretation of tumor PD-L1 expression and other candidate biomarkers of response to antitumor PD-1/PD-L1 blockade was provided, particularly for the tumor microenvironment-derived ‘immunomes’, and the challenges faced in their clinical use was addressed. Directions for future biomarker development and the potential of combined biomarker strategies were also proposed.
Collapse
Affiliation(s)
- Wenjie You
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Bin Shang
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jian Sun
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xueqing Liu
- Department of Respiratory and Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Lili Su
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shujuan Jiang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
6
|
Zhang M, Yang J, Hua W, Li Z, Xu Z, Qian Q. Monitoring checkpoint inhibitors: predictive biomarkers in immunotherapy. Front Med 2019; 13:32-44. [PMID: 30680606 DOI: 10.1007/s11684-018-0678-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
Immunotherapy has become the fourth cancer therapy after surgery, chemotherapy, and radiotherapy. In particular, immune checkpoint inhibitors are proved to be unprecedentedly in increasing the overall survival rates of patients with refractory cancers, such as advanced melanoma, non-small cell lung cancer, and renal cell carcinoma. However, inhibitor therapies are only effective in a small proportion of patients with problems, such as side effects and high costs. Therefore, doctors urgently need reliable predictive biomarkers for checkpoint inhibitor therapies to choose the optimal therapies. Here, we review the biomarkers that can serve as potential predictors of the outcomes of immune checkpoint inhibitor treatment, including tumor-specific profiles and tumor microenvironment evaluation and other factors.
Collapse
Affiliation(s)
- Min Zhang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Jingwen Yang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Wenjing Hua
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zhong Li
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zenghui Xu
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| | - Qijun Qian
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| |
Collapse
|
7
|
Transcriptional analysis of immune genes in Epstein-Barr virus-associated gastric cancer and association with clinical outcomes. Gastric Cancer 2018; 21:1064-1070. [PMID: 29915957 DOI: 10.1007/s10120-018-0851-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Epstein-Barr virus-associated gastric cancer (EBVaGC) has traditionally been associated with high expression of PD-L1 and immune infiltration. Correlations between PD-L1 and other immune-related gene (IRG) expressions in EBVaGC have not been previously described. METHODS We performed NanoString® transcriptomic profiling and PD-L1 immunohistochemistry (IHC) (using the FDA approved Dako PD-L1 IHC 22C3) on EBVaGC samples from gastric cancer patients undergoing primary tumor resections at Samsung Medical Centre, South Korea. For controls, EBV-negative samples from the previously reported Asian Cancer Research Group (EBVnegACRG) cohort were used. Genes tested included PD-L1 and other IRGs related to intra-tumoral cytolytic activity, cytokines and immune checkpoints. Samples with PD-L1 expression > 34th percentile were defined as PD-L1high and the remaining as PD-L1low. RESULTS We identified 71 cases of EBVaGC and 193 EBV-negative ACRG samples as controls. EBVaGC showed higher expression of all queried immune genes compared to EBVnegACRG samples (p < 0.01). PD-L1 immunohistochemistry expression correlated with PD-L1 transcript expression (r = 0.63, p < 0.001). Tumor-infiltrating lymphocyte patterns were also found to be different between PD-L1low and PD-L1high groups. PD-L1low EBVaGC samples (n = 24, 34%) had consistently decreased expression of all other immune genes, such as CD8A, GZMA and PRF1 and PD-1 (p < 0.001). PD-L1low EBVaGC samples were also associated with worse disease-free survival (HR 5.03, p = 0.032) compared to PD-L1high EBVaGC samples. CONCLUSIONS A substantial proportion of EBVaGC does not express high levels of PD-L1 and other immune genes. EBVaGCs which have lower transcriptomic expression of PD-L1 tend to have a similarly low expression of other immune genes, IHC scores and a poorer prognosis.
Collapse
|
8
|
Zou MX, Guo KM, Lv GH, Huang W, Li J, Wang XB, Jiang Y, She XL. Clinicopathologic implications of CD8 +/Foxp3 + ratio and miR-574-3p/PD-L1 axis in spinal chordoma patients. Cancer Immunol Immunother 2018; 67:209-224. [PMID: 29051990 PMCID: PMC11028121 DOI: 10.1007/s00262-017-2080-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/14/2017] [Indexed: 12/11/2022]
Abstract
Currently, little is known about the interactions between microRNAs (miRNAs) and the PD-1/PD-L1 signaling pathway in chordoma, and data discussing the role of the immune milieu in chordoma prognosis are limited. We aimed to analyze the relationship between PD-L1, miR-574-3p, microenvironmental tumor-infiltrating lymphocytes (TILs) and clinicopathological features of spinal chordoma patients. PD-L1 expression and TILs (including Foxp3+, CD8+, PD-1+ and PD-L1+) were assessed by immunohistochemistry in tumor specimens of 54 spinal chordoma patients. MiRNAs microarray and bioinformatical analysis were used to identify miRNAs potentially regulating PD-L1 expression, which were further validated by quantitative RT-PCR. miR-574-3p was identified to potentially regulate PD-L1 expression in chordoma, which inversely correlated with PD-L1. Positive PD-L1 expression on tumor cells was associated with advanced stages (P = 0.041) and TILs infiltration (P = 0.005), whereas decreased miR-574-3p level correlated with higher muscle invasion (P = 0.012), more severe tumor necrosis (P = 0.022) and poor patient survival. Importantly, a patient subgroup with PD-L1+/miR-574-3plow chordoma phenotype was significantly associated with worse local recurrence-free survival (LRFS) (P = 0.026). PD-1+ TILs density was associated with surrounding muscle invasion (P = 0.014), and independently portended poor LRFS (P = 0.040), while PD-L1+ TILs showed tendencies of less aggressive clinical outcomes. Multivariate analysis of OS only found CD8+/Foxp3+ ratio to be independent prognostic factor (P = 0.022). These findings may be useful to stratify patients into prognostic groups and provide a rationale for the use of checkpoint blockade therapy, possibly by administering miR-574-3p mimics, in spinal chordoma.
Collapse
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Ke-Miao Guo
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China.
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China.
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
9
|
Zheng P, Li J, Kros JM. Breakthroughs in modern cancer therapy and elusive cardiotoxicity: Critical research-practice gaps, challenges, and insights. Med Res Rev 2018; 38:325-376. [PMID: 28862319 PMCID: PMC5763363 DOI: 10.1002/med.21463] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/16/2022]
Abstract
To date, five cancer treatment modalities have been defined. The three traditional modalities of cancer treatment are surgery, radiotherapy, and conventional chemotherapy, and the two modern modalities include molecularly targeted therapy (the fourth modality) and immunotherapy (the fifth modality). The cardiotoxicity associated with conventional chemotherapy and radiotherapy is well known. Similar adverse cardiac events are resurging with the fourth modality. Aside from the conventional and newer targeted agents, even the most newly developed, immune-based therapeutic modalities of anticancer treatment (the fifth modality), e.g., immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapy, have unfortunately led to potentially lethal cardiotoxicity in patients. Cardiac complications represent unresolved and potentially life-threatening conditions in cancer survivors, while effective clinical management remains quite challenging. As a consequence, morbidity and mortality related to cardiac complications now threaten to offset some favorable benefits of modern cancer treatments in cancer-related survival, regardless of the oncologic prognosis. This review focuses on identifying critical research-practice gaps, addressing real-world challenges and pinpointing real-time insights in general terms under the context of clinical cardiotoxicity induced by the fourth and fifth modalities of cancer treatment. The information ranges from basic science to clinical management in the field of cardio-oncology and crosses the interface between oncology and onco-pharmacology. The complexity of the ongoing clinical problem is addressed at different levels. A better understanding of these research-practice gaps may advance research initiatives on the development of mechanism-based diagnoses and treatments for the effective clinical management of cardiotoxicity.
Collapse
Affiliation(s)
- Ping‐Pin Zheng
- Cardio‐Oncology Research GroupErasmus Medical CenterRotterdamthe Netherlands
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| | - Jin Li
- Department of OncologyShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Johan M Kros
- Department of PathologyErasmus Medical CenterRotterdamthe Netherlands
| |
Collapse
|
10
|
Sundar R, Chénard-Poirier M, Collins DC, Yap TA. Imprecision in the Era of Precision Medicine in Non-Small Cell Lung Cancer. Front Med (Lausanne) 2017; 4:39. [PMID: 28443282 PMCID: PMC5385461 DOI: 10.3389/fmed.2017.00039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/22/2017] [Indexed: 12/29/2022] Open
Abstract
Over the past decade, major advances have been made in the management of advanced non-small cell lung cancer (NSCLC). There has been a particular focus on the identification and targeting of putative driver aberrations, which has propelled NSCLC to the forefront of precision medicine. Several novel molecularly targeted agents have now achieved regulatory approval, while many others are currently in late-phase clinical trial testing. These antitumor therapies have significantly impacted the clinical outcomes of advanced NSCLC and provided patients with much hope for the future. Despite this, multiple deficiencies still exist in our knowledge of this complex disease, and further research is urgently required to overcome these critical issues. This review traces the path undertaken by the different therapeutics assessed in NSCLC and the impact of precision medicine in this disease. We also discuss the areas of "imprecision" that still exist in NSCLC and the modern hypothesis-testing studies being conducted to address these key challenges.
Collapse
Affiliation(s)
- Raghav Sundar
- Royal Marsden Hospital, London, UK
- Department of Haematology-Oncology, National University Cancer Institute of Singapore, Singapore, Singapore
| | | | | | - Timothy A. Yap
- Royal Marsden Hospital, London, UK
- The Institute of Cancer Research, London, UK
| |
Collapse
|