151
|
Xiu-Ying H, Yue-Xiang Z, Hui-Si Y, Hong-Zhou Y, Qing-Jie X, Ting-Hua W. PDGFBB facilitates tumorigenesis and malignancy of lung adenocarcinoma associated with PI3K-AKT/MAPK signaling. Sci Rep 2024; 14:4191. [PMID: 38378786 PMCID: PMC10879171 DOI: 10.1038/s41598-024-54801-7] [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: 09/17/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Lung adenocarcinoma (LUAD) remains one of the most aggressive tumors and the efficacy of conventional treatment has been bleak. Nowadays, gene-targeted therapy has become a new favorite in tumor therapy. Herein, we investigated the effect of platelet derived growth factor BB (PDGFBB) on LUAD. Firstly, PDGFBB was upregulated in LUAD patients and closely linked with poor survival. Furthermore, the expression of PDGFBB and PDGFRα/β in LUAD cells was higher than that in normal lung cells. By loss-of-function with herpes simplex virus (HSV)-PDGFi-shRNA, we found that PDGFBB knockdown caused a significant decrease in proliferation and migration, but evoked apoptosis of LUAD cells in vitro. Conversely, exogenous PDGFBB held adverse effect. Additionally, A549 cells with PDGFBB knockdown had a low probability of tumorigenesis in vivo. Moreover, PDGFBB knockdown restrained the growth of xenografts derived from normal A549 cells. Mechanistically, PDGFBB knockdown suppressed PI3K/AKT and Ras/MAPK signaling, while PDGFBB was the opposite. Therefore, we concluded that PDGFBB might facilitate the tumorigenesis and malignancy of LUAD through its functional downstream nodes-PI3K/AKT and Ras/MAPK signaling, which supported that PDGFBB could serve as a rational therapeutic target for LUAD.
Collapse
Affiliation(s)
- He Xiu-Ying
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zheng Yue-Xiang
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yang Hui-Si
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yu Hong-Zhou
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xia Qing-Jie
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wang Ting-Hua
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China.
| |
Collapse
|
152
|
Hu H, Zhang M. Correlation analysis between peripheral blood dendritic cell subsets and PD-1 in patients with peritoneal adenocarcinoma. Braz J Med Biol Res 2024; 57:e13192. [PMID: 38381884 PMCID: PMC10880883 DOI: 10.1590/1414-431x2023e13192] [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: 10/23/2023] [Accepted: 12/27/2023] [Indexed: 02/23/2024] Open
Abstract
The aim of this study was to explore the association between differential percentages of dendritic cell (DC) subsets in peripheral blood and malignancy (grade and lymph node metastasis) of peritoneal adenocarcinoma patients and the frequencies of dendritic cell subsets in the normal controls. The peripheral blood of 30 patients with peritoneal adenocarcinoma and 12 healthy controls were collected for multicolor flow cytometry analysis. Peritoneal adenocarcinoma patients were grouped according to the malignant degree (grade and lymph node metastasis). Percentages of myeloid DCs (mDCs) and its subsets MDC1 and MDC2 in DCs were lower in peripheral blood of patients with peritoneal adenocarcinoma than in normal controls. The percentages of plasmacytoid dendritic cells (pDCs) and CD16+mDCs in DCs were higher than in normal controls. Compared with poor differentiation grade, patients with well/moderate differentiation grade had an increased percentage of CD16+mDCs. Contrary to CD16+mDCs, the percentage of MDC1 was lower in the well/moderate differentiation grade group. In patients with no lymph node metastasis, pDCs and CD16+mDCs levels were higher compared with patients with lymph node metastasis. mDCs and MDC1 levels had opposite results. pDCs were positively correlated with CD16+mDCs in peripheral blood of peritoneal patients, as was mDCs and MDC1. CD16+mDCs were negatively correlated with MDC1. The percentages of pDCs and CD16+mDCs in DCs were positively correlated with CD3+CD8+T cells, and pDCs also positively correlated with CD8+PD-1+T cells. Our results revealed that DCs subsets correlated with peritoneal adenocarcinoma malignancy. Dendritic cells play an independent role in the immune function of peritoneal adenocarcinoma.
Collapse
Affiliation(s)
- Huihui Hu
- Department of Clinical Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Man Zhang
- Department of Clinical Laboratory, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- Clinical Laboratory Medicine, Peking University Ninth School of Clinical Medicine, Beijing, China
| |
Collapse
|
153
|
Liu XS, Chen YX, Wan HB, Wang YL, Wang YY, Gao Y, Wu LB, Pei ZJ. TRIP6 a potential diagnostic marker for colorectal cancer with glycolysis and immune infiltration association. Sci Rep 2024; 14:4042. [PMID: 38369589 PMCID: PMC10874967 DOI: 10.1038/s41598-024-54670-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
Thyroid hormone receptor interactor 6 (TRIP6) it is an adaptor protein belonging to the zyxin family of LIM proteins, participating in signaling events through interactions with various molecules. Despite this, TRIP6's role in colorectal cancer (CRC), particularly its correlation with glucose metabolism and immune cell infiltration, remains unclear. Through the TCGA and GEO databases, we obtained RNA sequencing data to facilitate our in-depth study and analysis of TRIP6 expression. To investigate the prognostic value of TRIP6 in CRC, we also used univariate Cox regression analysis. In addition, this study also covered a series of analyses, including clinicopathological analysis, functional enrichment analysis, glycolysis correlation analysis, immunoinfiltration analysis, immune checkpoint analysis, and angiogenesis correlation analysis, to gain a comprehensive and in-depth understanding of this biological phenomenon. It has been found that TRIP6 expression is significantly upregulated in CRC and correlates with the stage of the disease. Its overexpression portends a worse survival time. Functional enrichment analysis reveals that TRIP6 is associated with focal adhesion and glycolysis. Mechanistically, TRIP6 appears to exert its tumorigenic effect by regulating the glycolysis-related gene GPI. A higher level of expression of TRIP6 is associated with an increase in the number of iDC immune cells and a decrease in the number of Th1 immune cells. Also, TRIP6 may promote angiogenesis in tumor cells by promoting the expression of JAG2. Our study uncovers the upregulation of TRIP6 in CRC, illuminating its prognostic and diagnostic value within this context. Furthermore, we examine the relationship between TRIP6 expression levels, glycolysis, angiogenesis and immune cell infiltration. This underscores its potential as a biomarker for CRC treatment and as a therapeutic target.
Collapse
Affiliation(s)
- Xu-Sheng Liu
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China
| | - Yu-Xuan Chen
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Hua-Bing Wan
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Ya-Lan Wang
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yang-Yang Wang
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yan Gao
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Li-Bing Wu
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| | - Zhi-Jun Pei
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for precision Diagnosis and Treatment of liver cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, Hubei, China.
| |
Collapse
|
154
|
Zhang H, Lu X, Lu B, Gullo G, Chen L. Measuring the composition of the tumor microenvironment with transcriptome analysis: past, present and future. Future Oncol 2024; 20:1207-1220. [PMID: 38362731 PMCID: PMC11318690 DOI: 10.2217/fon-2023-0658] [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: 07/31/2023] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Interactions between tumor cells and immune cells in the tumor microenvironment (TME) play a vital role the mechanisms of immune evasion, by which cancer cells escape immune elimination. Thus, the characterization and quantification of different components in the TME is a hot topic in molecular biology and drug discovery. Since the development of transcriptome sequencing in bulk tissue, single cells and spatial dimensions, there are increasing methods emerging to deconvolute and subtype the TME. This review discusses and compares such computational strategies and downstream subtyping analyses. Integrative analyses of the transcriptome with other data, such as epigenetics and T-cell receptor sequencing, are needed to obtain comprehensive knowledge of the dynamic TME.
Collapse
Affiliation(s)
- Han Zhang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Binfeng Lu
- Center for Discovery & Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Giuseppe Gullo
- Department of Obstetrics & Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146, Palermo, Italy
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| |
Collapse
|
155
|
Yang J, Liu Q, Shyr Y. A Large-Scale Meta-Analysis Reveals Positive Feedback between Macrophages and T Cells That Sensitizes Tumors to Immunotherapy. Cancer Res 2024; 84:626-638. [PMID: 38117502 PMCID: PMC10867621 DOI: 10.1158/0008-5472.can-23-2006] [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: 07/08/2023] [Revised: 10/24/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
Although considerable efforts have been dedicated to identifying predictive signatures for immune checkpoint inhibitor (ICI) treatment response, current biomarkers suffer from poor generalizability and reproducibility across different studies and cancer types. The integration of large-scale multiomics studies holds great promise for discovering robust biomarkers and shedding light on the mechanisms of immune resistance. In this study, we conducted the most extensive meta-analysis involving 3,037 ICI-treated patients with genetic and/or transcriptomics profiles across 14 types of solid tumor. The comprehensive analysis uncovered both known and novel reliable signatures associated with ICI treatment outcomes. The signatures included tumor mutational burden (TMB), IFNG and PDCD1 expression, and notably, interactions between macrophages and T cells driving their activation and recruitment. Independent data from single-cell RNA sequencing and dynamic transcriptomic profiles during the ICI treatment provided further evidence that enhanced cross-talk between macrophages and T cells contributes to ICI response. A multivariable model based on eight nonredundant signatures significantly outperformed existing models in five independent validation datasets representing various cancer types. Collectively, this study discovered biomarkers predicting ICI response that highlight the contribution of immune cell networks to immunotherapy efficacy and could help guide patient treatment. SIGNIFICANCE Identification of robust immunogenomic connections, particularly macrophage T-cell interactions, in a large-scale pan-cancer meta-analysis and development of a predictive model for immunotherapy response that outperformed existing models could facilitate clinical decision-making.
Collapse
Affiliation(s)
- Jing Yang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
156
|
Ilangumaran S, Gui Y, Shukla A, Ramanathan S. SOCS1 expression in cancer cells: potential roles in promoting antitumor immunity. Front Immunol 2024; 15:1362224. [PMID: 38415248 PMCID: PMC10897024 DOI: 10.3389/fimmu.2024.1362224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a potent regulator immune cell responses and a proven tumor suppressor. Inhibition of SOCS1 in T cells can boost antitumor immunity, whereas its loss in tumor cells increases tumor aggressivity. Investigations into the tumor suppression mechanisms so far focused on tumor cell-intrinsic functions of SOCS1. However, it is possible that SOCS1 expression in tumor cells also regulate antitumor immune responses in a cell-extrinsic manner via direct and indirect mechanisms. Here, we discuss the evidence supporting the latter, and its implications for antitumor immunity.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
157
|
Cong XZ, Feng J, Zhang HJ, Zhang LZ, Lin TY, Chen G, Zhang ZL. Microfluidic Device-Based In Vivo Detection of PD-L1-Positive Small Extracellular Vesicles and Its Application for Tumor Monitoring. Anal Chem 2024; 96:2658-2665. [PMID: 38311857 DOI: 10.1021/acs.analchem.3c05418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Liquid biopsy is of great significance in tumor early diagnosis and treatment stratification. PD-L1-positive small extracellular vesicles (PD-L1+ sEVs) are closely related to tumor growth and immunotherapy response, which are considered valuable liquid biopsy biomarkers. In contrast to conventional in vitro detection, in vivo detection has the ability to improve the detection efficiency and enable continuous or real-time dynamic monitoring. However, in vivo detection of PD-L1+ sEVs has multiple difficulties, such as high cell background, complex blood environments, and lack of a specific and stable detection method. Herein, the in vivo detection of PD-L1+ sEVs method was constructed, which efficiently separated sEVs based on the microfluidic device and quantitatively analyzed PD-L1+ sEVs by aptamer recognition and hybridization chain reaction. The concentration of PD-L1+ sEVs was continuously monitored, and significant differences at different stages of tumor as well as a correlation with tumor volume were found. Diseased and healthy individuals could also be effectively distinguished based on the concentration of PD-L1+ sEVs. The method with good stability, biocompatibility, and detection performance provided a powerful means for in vivo detection of PD-L1+ sEVs, contributing to the clinical diagnosis and treatment of tumor.
Collapse
Affiliation(s)
- Xi-Zhu Cong
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Jiao Feng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - He-Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Lin-Zhou Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Tian-Yang Lin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
158
|
Asleh K, Ouellette RJ. Tumor Copy Number Alteration Burden as a Predictor for Resistance to Immune Checkpoint Blockade across Different Cancer Types. Cancers (Basel) 2024; 16:732. [PMID: 38398121 PMCID: PMC10886982 DOI: 10.3390/cancers16040732] [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: 01/15/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Immune checkpoint blockade (ICB) benefits only a subset of advanced cancer patients, and predictive biomarkers for immunotherapy response are needed. Recently, copy number alteration (CNA) burden has been proposed to predict ICB resistance. We assessed this finding using the publicly accessible data for 1661 ICB-treated patients whose tumors were profiled by MSK-IMPACT, an approved targeted assay in clinical care. We tested the hypothesis that the continuous increase in CNA burden is associated with poor overall survival following ICB. In addition, we hypothesized that the combinatorial biomarkers of tumor mutational burden (TMB) and CNA burden would better stratify patients for immune status and ICB response. Of the 1661 cases, 79% (n = 1307) were treated with anti PD-1/PD-L1 and the remaining 21% (n = 354) with anti CTLA-4 or the combination of both. In a multivariate analysis, increase in CNA burden was associated with poor overall survival [HR = 1.52, 95% CI (1.01-2.30), p = 0.04]. The combination of biomarkers TMB and CNA burden stratified patients into four clinically distinct subsets among which "LowTMB/HighCNA" showed the worst survival (p < 0.0001). The four patient subsets had unique CNA profiles and enriched pathways, which could predict transcriptional and phenotypic effects related to immune signaling and CD8+ T-cell abundance in the tumor microenvironment. CNA burden was associated with poor overall survival in patients receiving ICB and could improve patient stratification when incorporated with TMB. These findings may guide patient selection for immunotherapy or alternative strategies.
Collapse
Affiliation(s)
- Karama Asleh
- Department of Pathology and Laboratory Medicine, Halifax, NS B3H 1V8, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada;
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Rodney J. Ouellette
- Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 0A2, Canada;
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Dr. Georges L. Dumont University Hospital, Vitalité Health Network, Moncton, NB E1C 2Z3, Canada
| |
Collapse
|
159
|
Wang J, Wang S, Zhang Y, Zhang W. Bibliometric analysis of evolutionary trajectory and prospective directions of LAG-3 in cancer. Front Immunol 2024; 15:1329775. [PMID: 38390331 PMCID: PMC10881671 DOI: 10.3389/fimmu.2024.1329775] [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: 10/29/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Objectives Perform a bibliometric analysis on the role of LAG-3 in the domain of cancer, elucidate the prevailing areas of research, and visually depict the evolutionary trajectory and prospective directions of LAG-3 research over the past twenty-three decades. Materials and methods Between 2000 and 2023, a comprehensive review of scholarly articles pertaining to LAG-3 research in the context of cancer was carried out using the Web of Science Core Collection (WoSCC) database. Bibliometric analysis can be conducted by taking advantage of VOSviewer (version 1.6.16) and CiteSpace (version 6.2.R4). Create a network diagram to visually represent various authors, countries, and organizations while assessing the publishing years, journals, references, and keywords. Results In conclusion, 1841 records were identified and published in 587 publications. These records were authored by 12,849 individuals affiliated with 2491 institutes across 74 countries. There has been a substantial surge in publications subsequent to 2013. The USA, China, and Germany gave the majority of records, amounting to 69.69%. American institutions actively engage in collaboration with institutions located in other countries. Triebel, F., Vignali, Dario A. A., Workman, Creg J. Drake, Charles G., and Elkord, Eyad are highly regarded authors in their respective fields. However, it is worth noting that Triebel exhibits limited collaboration with other writers. The examination of the role of LAG-3 in cancer and its potential for use in clinical settings is a discernible trend, as seen by keyword analysis. Conclusion The scientific interest in and attention towards LAG-3 has experienced a significant rise since 2013. The United States is leading the way, with China following closely behind. Promoting collaboration among writers, nations, and institutions with varied backgrounds is imperative. The discipline of immunotherapy is currently seeing ongoing progress. A thorough investigation of the distinctive cis ligand TCR-CD3 complex of LAG-3 and its signal transduction mechanism is necessary. Additionally, it is worthwhile to explore novel combinations of LAG-3 therapy.
Collapse
Affiliation(s)
| | | | | | - Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
160
|
Noel OD, Hassouneh Z, Svatek RS, Mukherjee N. Innate Lymphoid Cells in Bladder Cancer: From Mechanisms of Action to Immune Therapies. Cancer Immunol Res 2024; 12:149-160. [PMID: 38060011 PMCID: PMC11492724 DOI: 10.1158/2326-6066.cir-23-0414] [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: 05/19/2023] [Revised: 07/10/2023] [Accepted: 10/24/2023] [Indexed: 12/08/2023]
Abstract
Bladder tumors have a high mutational burden and tend to be responsive to immune therapies; however, response rates remain modest. To date, immunotherapy in bladder cancer has largely focused on enhancing T-cell immune responses in the bladder tumor microenvironment. It is anticipated that other immune cells, including innate lymphoid cells (ILC), which play an important role in bladder oncogenesis and tumor suppression, could be targeted to improve response to existing therapies. ILCs are classified into five groups: natural killer cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer cells. ILCs are pleiotropic and play dual and sometimes paradoxical roles in cancer development and progression. Here, a comprehensive discussion of the current knowledge and recent advancements in understanding the role of ILCs in bladder cancer is provided. We discuss the multifaceted roles that ILCs play in bladder immune surveillance, tumor protection, and immunopathology of bladder cancer. This review provides a rationale for targeting ILCs in bladder cancer, which is relevant for other solid tumors.
Collapse
Affiliation(s)
- Onika D.V. Noel
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas
| | - Zaineb Hassouneh
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas
| | - Robert S. Svatek
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas
| | - Neelam Mukherjee
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
161
|
Ajay AK, Gasser M, Hsiao LL, Böldicke T, Waaga-Gasser AM. TLR2 and TLR9 Blockade Using Specific Intrabodies Inhibits Inflammation-Mediated Pancreatic Cancer Cell Growth. Antibodies (Basel) 2024; 13:11. [PMID: 38390872 PMCID: PMC10885114 DOI: 10.3390/antib13010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) remains a deadly cancer worldwide with a need for new therapeutic approaches. A dysregulation in the equilibrium between pro- and anti-inflammatory responses with a predominant immunosuppressive inflammatory reaction in advanced stage tumors seem to contribute to tumor growth and metastasis. The current therapies do not include strategies against pro-tumorigenic inflammation in cancer patients. We have shown that the upregulated cell surface expression of Toll-like Receptor (TLR) 2 and of TLR9 inside PDAC cells maintain chronic inflammatory responses, support chemotherapeutic resistance, and mediate tumor progression in human pancreatic cancer. We further demonstrated intracellular TLR2 and TLR9 targeting using specific intrabodies, which resulted in downregulated inflammatory signaling. In this study, we tested, for the first time, an intrabody-mediated TLR blockade in human TLR2- and TLR9-expressing pancreatic cancer cells for its effects on inflammatory signaling-mediated tumor growth. Newly designed anti-TLR2- and anti-TLR9-specific intrabodies inhibited PDAC growth. Co-expression analysis of the intrabodies and corresponding human TLRs showed efficient retention and accumulation of both intrabodies within the endoplasmic reticulum (ER), while co-immunoprecipitation studies indicated both intrabodies interacting with their cognate TLR antigen within the pancreatic cancer cells. Cancer cells with attenuated proliferation expressing accumulated TLR2 and TRL9 intrabodies demonstrated reduced STAT3 phosphorylation signaling, while apoptotic markers Caspases 3 and 8 were upregulated. To conclude, our results demonstrate the TLR2 and TLR9-specific intrabody-mediated signaling pathway inhibition of autoregulatory inflammation inside cancer cells and their proliferation, resulting in the suppression of pancreatic tumor cell growth. These findings underscore the potential of specific intrabody-mediated TLR inhibition in the ER relevant for tumor growth inhibition and open up a new therapeutic intervention strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Amrendra K Ajay
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Martin Gasser
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Li-Li Hsiao
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Böldicke
- Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Ana Maria Waaga-Gasser
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
162
|
Yamasaki S, Shimizu K, Fujii SI. Tumor epitope spreading by a novel multivalent therapeutic cellular vaccine targeting cancer antigens to invariant NKT-triggered dendritic cells in situ. Front Immunol 2024; 15:1345037. [PMID: 38361934 PMCID: PMC10867576 DOI: 10.3389/fimmu.2024.1345037] [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: 11/27/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction Cancer is categorized into two types based on the microenvironment: cold and hot tumors. The former is challenging to stimulate through immunity. The immunogenicity of cancer relies on the quality and quantity of cancer antigens, whether recognized by T cells or not. Successful cancer immunotherapy hinges on the cancer cell type, antigenicity and subsequent immune reactions. The T cell response is particularly crucial for secondary epitope spreading, although the factors affecting these mechanisms remain unknown. Prostate cancer often becomes resistant to standard therapy despite identifying several antigens, placing it among immunologically cold tumors. We aim to leverage prostate cancer antigens to investigate the potential induction of epitope spreading in cold tumors. This study specifically focuses on identifying factors involved in secondary epitope spreading based on artificial adjuvant vector cell (aAVC) therapy, a method established as invariant natural killer T (iNKT) -licensed DC therapy. Methods We concentrated on three prostate cancer antigens (prostate-specific membrane antigen (PSMA), prostate-specific antigen (PSA), and prostatic acid phosphatase (PAP)). By introducing allogeneic cells with the antigen and murine CD1d mRNA, followed by α-galactosylceramide (α-GalCer) loading, we generated five types of aAVCs, i.e, monovalent, divalent and trivalent antigen-expressing aAVCs and four types of prostate antigen-expressing cold tumors. We evaluated iNKT activation and antigen-specific CD8+ T cell responses against tumor cells prompted by the aAVCs. Results Our study revealed that monovalent aAVCs, expressing a single prostate antigen, primed T cells for primary tumor antigens and also induced T cells targeting additional tumor antigens by triggering a tumor antigen-spreading response. When we investigated the immune response by trivalent aAVC (aAVC-PROS), aAVC-PROS therapy elicited multiple antigen-specific CD8+ T cells simultaneously. These CD8+ T cells exhibited both preventive and therapeutic effects against tumor progression. Conclusions The findings from this study highlight the promising role of tumor antigen-expressing aAVCs, in inducing efficient epitope spreading and generating robust immune responses against cancer. Our results also propose that multivalent antigen-expressing aAVCs present a promising therapeutic option and could be a more comprehensive therapy for treating cold tumors like prostate cancer.
Collapse
Affiliation(s)
- Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medical Science (IMS), Yokohama, Japan
- aAVC Drug Translational Unit, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Research Center for Integrative Medical Science (IMS), Yokohama, Japan
- aAVC Drug Translational Unit, RIKEN Center for Integrative Medical Science (IMS), Yokohama, Japan
- RIKEN Program for Drug Discovery and Medical Technology Platforms, Yokohama, Japan
| |
Collapse
|
163
|
Xie X, Zhang J, Wang Y, Shi W, Tang R, Tang Q, Sun S, Wu R, Xu S, Wang M, Liang X, Cui L. Nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. Mater Today Bio 2024; 24:100926. [PMID: 38179429 PMCID: PMC10765306 DOI: 10.1016/j.mtbio.2023.100926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024] Open
Abstract
Immunotherapy as a milestone in cancer treatment has made great strides in the past decade, but it is still limited by low immune response rates and immune-related adverse events. Utilizing bioeffects of ultrasound to enhance tumor immunotherapy has attracted more and more attention, including sonothermal, sonomechanical, sonodynamic and sonopiezoelectric immunotherapy. Moreover, the emergence of nanomaterials has further improved the efficacy of ultrasound mediated immunotherapy. However, most of the summaries in this field are about a single aspect of the biological effects of ultrasound, which is not comprehensive and complete currently. This review proposes the recent progress of nanomaterials augmented bioeffects of ultrasound in cancer immunotherapy. The concept of immunotherapy and the application of bioeffects of ultrasound in cancer immunotherapy are initially introduced. Then, according to different bioeffects of ultrasound, the representative paradigms of nanomaterial augmented sono-immunotherapy are described, and their mechanisms are discussed. Finally, the challenges and application prospects of nanomaterial augmented ultrasound mediated cancer immunotherapy are discussed in depth, hoping to pave the way for cancer immunotherapy and promote the clinical translation of ultrasound mediated cancer immunotherapy through the reasonable combination of nanomaterials augmented ultrasonic bioeffects.
Collapse
Affiliation(s)
- Xinxin Xie
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yuan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Wanrui Shi
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Rui Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Qingshuang Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ruiqi Wu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Shuyu Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Mengxin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Ligang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, P.R. China
| |
Collapse
|
164
|
Xin Y, Liu CG, Zang D, Chen J. Gut microbiota and dietary intervention: affecting immunotherapy efficacy in non-small cell lung cancer. Front Immunol 2024; 15:1343450. [PMID: 38361936 PMCID: PMC10867196 DOI: 10.3389/fimmu.2024.1343450] [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: 11/23/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80-85% of all lung cancers. In recent years, treatment with immune checkpoint inhibitors (ICIs) has gradually improved the survival rate of patients with NSCLC, especially those in the advanced stages. ICIs can block the tolerance pathways that are overexpressed by tumor cells and maintain the protective activity of immune system components against cancer cells. Emerging clinical evidence suggests that gut microbiota may modulate responses to ICIs treatment, possibly holding a key role in tumor immune surveillance and the efficacy of ICIs. Studies have also shown that diet can influence the abundance of gut microbiota in humans, therefore, dietary interventions and the adjustment of the gut microbiota is a novel and promising treatment strategy for adjunctive cancer therapy. This review comprehensively summarizes the effects of gut microbiota, antibiotics (ATBs), and dietary intervention on the efficacy of immunotherapy in NSCLC, with the aim of informing the development of novel strategies in NSCLC immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
165
|
Tang C, Yang B, Liu Y. The efficacy of neoadjuvant immunotherapy combined with chemotherapy in resectable stage II-IV non-small cell lung cancer: a preliminary study. Transl Cancer Res 2024; 13:290-298. [PMID: 38410203 PMCID: PMC10894338 DOI: 10.21037/tcr-23-1276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/24/2023] [Indexed: 02/28/2024]
Abstract
Background Lung cancer is currently the world's leading malignancy in terms of morbidity and mortality. Neoadjuvant therapy is widely used in clinic to improve R0 resection rates and long-term survival after surgery, and patients with locally resectable non-small cell lung cancer (NSCLC) may benefit from neoadjuvant therapy. Methods Data from 78 patients with stage II to IV NSCLC who had received neoadjuvant immunotherapy combined with chemotherapy from January 2019 to May 2022 were collected. The patients were categorized into groups based on their eligibility for posttreatment surgery, the level of pathological remission, and receipt of adjuvant therapy. The progression-free survival (PFS) and survival rates of patients in each group were compared. Efforts were made to identify the factors that influence patients' prognoses. Results The incidence of adverse events in patients who received neoadjuvant immunotherapy combined with chemotherapy was 19%. The proportion of patients receiving neoadjuvant immunotherapy and chemotherapy undergoing surgery was 83.33%, and the rate of R0 resection was 64.10%. The pathological complete response (pCR) and major pathological response (MPR) rates were 26.25% and 21.87%, respectively. Patients who received adjuvant therapy were less likely to experience recurrent metastases than were those who did not receive adjuvant therapy (χ2=7.183; P=0.007<0.05). Conclusions Neoadjuvant immunotherapy combined with chemotherapy has a low incidence of adverse events in resectable stage II-IV NSCLC, does not significantly increase the difficulty of surgery, and provides greater benefit in terms of PFS for patients who receive operation and adjuvant therapy.
Collapse
Affiliation(s)
- Chengbin Tang
- School of Medicine, Nankai University, Tianjin, China
- Department of Thoracic Surgery, First Medical Center, General Hospital of Chinese People Liberation Army, Beijing, China
| | - Bo Yang
- Department of Thoracic Surgery, First Medical Center, General Hospital of Chinese People Liberation Army, Beijing, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center, General Hospital of Chinese People Liberation Army, Beijing, China
| |
Collapse
|
166
|
He J, Zhou Y, Sun L. Emerging mechanisms of the unfolded protein response in therapeutic resistance: from chemotherapy to Immunotherapy. Cell Commun Signal 2024; 22:89. [PMID: 38297380 PMCID: PMC10832166 DOI: 10.1186/s12964-023-01438-0] [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: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 02/02/2024] Open
Abstract
The accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and activates the unfolded protein response (UPR). As an adaptive cellular response to hostile microenvironments, such as hypoxia, nutrient deprivation, oxidative stress, and chemotherapeutic drugs, the UPR is activated in diverse cancer types and functions as a dynamic tumour promoter in cancer development; this role of the UPR indicates that regulation of the UPR can be utilized as a target for tumour treatment. T-cell exhaustion mainly refers to effector T cells losing their effector functions and expressing inhibitory receptors, leading to tumour immune evasion and the loss of tumour control. Emerging evidence suggests that the UPR plays a crucial role in T-cell exhaustion, immune evasion, and resistance to immunotherapy. In this review, we summarize the molecular basis of UPR activation, the effect of the UPR on immune evasion, the emerging mechanisms of the UPR in chemotherapy and immunotherapy resistance, and agents that target the UPR for tumour therapeutics. An understanding of the role of the UPR in immune evasion and therapeutic resistance will be helpful to identify new therapeutic modalities for cancer treatment. Video Abstract.
Collapse
Affiliation(s)
- Jiang He
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Huan, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central, South University, Xiangya Hospital, Changsha, 410008, China.
| | - You Zhou
- Department of Pathology, Tongji Medical College Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Huan, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central, South University, Xiangya Hospital, Changsha, 410008, China.
| |
Collapse
|
167
|
Hu B, Zhang X, Zhu S, Wang C, Deng Z, Wang T, Wu Y. Identification and validation of an individualized metabolic prognostic signature for predicting the biochemical recurrence of prostate cancer based on the immune microenvironment. Eur J Med Res 2024; 29:92. [PMID: 38297388 PMCID: PMC10829481 DOI: 10.1186/s40001-024-01672-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/13/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the most prevalent genitourinary malignancy in men, with a significant proportion of patients developing biochemical recurrence (BCR) after treatment. The immune microenvironment and metabolic alterations have crucial implications for the tumorigenesis and progression of PCa. Therefore, identifying metabolic genes associated with the immune microenvironment holds promise for predicting BCR and improving PCa prognosis. METHODS In this study, ssGSEA and hierarchical clustering analysis were first conducted to evaluate and group PCa samples, followed by the use of the ESTIMATE and CIBERSORT algorithms to characterize the immunophenotypes and tumor microenvironment. The differential metabolic genes (MTGs) between groups were utilized to develop a prognostic-related signature. The predictive performance of the signature was assessed by principal component analysis (PCA), receiver operating characteristic (ROC) curve analysis, survival analysis, and the TIDE algorithm. A miRNA-MTGs regulatory network and predictive nomogram were constructed. Moreover, the expression of prognostic MTGs in PCa was detected by RT‒qPCR. RESULTS PCa samples from the TCGA cohort were separated into two groups: the immune-low group and immune-high group. Forty-eight differentially expressed MTGs between the groups were identified, including 37 up-regulated and 11 down-regulated MTGs. Subsequently, CEL, CYP3A4, and PDE6G were identified as the genes most strongly associated with the BCR of PCa patients and these genes were utilized to establish the MTGs-based prognostic signatures. PCA, ROC curves analysis, Kaplan-Meier survival analysis, and the nomogram all showed the good predictive ability of the signature regardless of clinical variables. Furthermore, the MTGs-based signature was indicated as a potential predictive biomarker for immunotherapy response. Nine miRNAs involved in the regulation of prognostic MTGs were determined. In addition to the CEL gene, the PDE6G and CYP3A4 genes were expressed at higher levels in PCa samples. CONCLUSIONS The MTGs-based signature represents a novel approach with promising potential for predicting BCR in PCa patients.
Collapse
Affiliation(s)
- Bintao Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xi Zhang
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiqing Zhu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengwei Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiyao Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, Guangdong, China.
| | - Yue Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
168
|
Jiang F, Mao M, Jiang S, Jiao Y, Cao D, Xiang Y. PD-1 and TIGIT coexpressing CD8 + CD103 + tissue-resident memory cells in endometrial cancer as potential targets for immunotherapy. Int Immunopharmacol 2024; 127:111381. [PMID: 38150880 DOI: 10.1016/j.intimp.2023.111381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Immunotherapy has shown promise in treating various cancers; however, its efficacy in endometrial cancer (EC) remains suboptimal owing to the complex dynamics of the tumour immune microenvironment. This study focuses on exploring the potential of targeting the programmed cell death protein 1 gene (PD-1) and the T cell Immunoreceptor with Ig and ITIM domains gene (TIGIT) coexpressing tissue-resident memory cells in EC. METHODS A comprehensive approach, utilizing RNA sequencing, single-cell RNA sequencing, mass cytometry, and flow cytometry, was employed to analyse the expression patterns of PD-1 and TIGIT in the EC tumor environment and to characterize the phenotypic properties of tumor-infiltrating lymphocytes (TILs), particularly tissue-resident memory (TRM) cells. Additionally, in vitro cell experiments were conducted to assess the functional impact of PD-1 and TIGIT blockade on T-cell activity. RESULTS Our analysis identified a significant co-expression of PD-1 and TIGIT in TRM cells within the EC tumor microenvironment. These TRM cells displayed an exhausted phenotype with impaired cytotoxicity, enhanced proliferative capacity, and diminished cytotoxic activity. In vitro T-cell assays showed that a dual blockade of PD-1 and TIGIT more effectively restored T-cell functionality compared to single blockade, suggesting enhanced therapeutic potential. CONCLUSIONS TRM cells co-expressing PD-1 and TIGIT represent potential targets for EC immunotherapy. Dual immune checkpoint blockade targeting PD-1 and TIGIT may offer an effective therapeutic strategy for EC, providing valuable insights for the development of immunotherapeutic approaches.
Collapse
Affiliation(s)
- Fang Jiang
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Mingyi Mao
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Shiyang Jiang
- Ovarian Cancer Program, Department of Gynaecologic Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuhao Jiao
- Department of Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Dongyan Cao
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China
| | - Yang Xiang
- Department of Obstetrics and Gynaecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynaecologic Diseases, Beijing, China.
| |
Collapse
|
169
|
Xuan M, Gu X, Liu Y, Yang L, Li Y, Huang D, Li J, Xue C. Intratumoral microorganisms in tumors of the digestive system. Cell Commun Signal 2024; 22:69. [PMID: 38273292 PMCID: PMC10811838 DOI: 10.1186/s12964-023-01425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024] Open
Abstract
Tumors of the digestive system pose a significant threat to human health and longevity. These tumors are associated with high morbidity and mortality rates, leading to a heavy economic burden on healthcare systems. Several intratumoral microorganisms are present in digestive system tumors, and their sources and abundance display significant heterogeneity depending on the specific tumor subtype. These microbes have a complex and precise function in the neoplasm. They can facilitate tumor growth through various mechanisms, such as inducing DNA damage, influencing the antitumor immune response, and promoting the degradation of chemotherapy drugs. Therefore, these microorganisms can be targeted to inhibit tumor progression for improving overall patient prognosis. This review focuses on the current research progress on microorganisms present in the digestive system tumors and how they influence the initiation, progression, and prognosis of tumors. Furthermore, the primary sources and constituents of tumor microbiome are delineated. Finally, we summarize the application potential of intratumoral microbes in the diagnosis, treatment, and prognosis prediction of digestive system tumors. Video Abstract.
Collapse
Affiliation(s)
- Mengjuan Xuan
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yingru Liu
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China
| | - Li Yang
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China
| | - Yi Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Juan Li
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China.
| | - Chen Xue
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China.
| |
Collapse
|
170
|
Le Y, Gao H, Le J, Hornick JL, Bleday R, Wee J, Zhu Z. VentX promotes tumor specific immunity and efficacy of immune checkpoint inhibitors. iScience 2024; 27:108731. [PMID: 38299030 PMCID: PMC10829883 DOI: 10.1016/j.isci.2023.108731] [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: 07/12/2023] [Revised: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 02/02/2024] Open
Abstract
Immune suppression within tumor microenvironments (TME) have been implicated in limited efficacy of immune check point inhibitors (ICIs) against solid tumors. Down-regulated VentX expression in tumor associated macrophages (TAMs) underlies phagocytotic anergic phenotype of TAMs, which govern immunological state of TME. In this study, using a tumor immune microenvironment enabling model system (TIME-EMS) of non-small cell lung cancer (NSCLC), we found that PD-1 antibody modestly activates cytotoxic T lymphocytes (CTLs) within the NSCLC-TME but not the status of TIME. We showed that the restoration of VentX expression in TAMs reignites the phagocytotic function of TAMs, which in turn, transforms TIME, activates CTLs in a tumor-specific manner and promotes efficacy of PD-1 antibody against NSCLC but not toxicity on normal lung epithelial cells. Supported by in vivo data on NSG-PDX models of primary human NSCLC, our study revealed potential venues to promote the efficacy of ICI against solid tumors through VentX-based mechanisms.
Collapse
Affiliation(s)
- Yi Le
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hong Gao
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Joanna Le
- Department of Obstetrics and Gynecology, University of Massachusetts Medical Center
| | - Jason L. Hornick
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ronald Bleday
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jon Wee
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Zhenglun Zhu
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
171
|
Sears T, Pagadala M, Castro A, Lee KH, Kong J, Tanaka K, Lippman S, Zanetti M, Carter H. Integrated germline and somatic features reveal divergent immune pathways driving ICB response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575430. [PMID: 38293085 PMCID: PMC10827124 DOI: 10.1101/2024.01.12.575430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Immune Checkpoint Blockade (ICB) has revolutionized cancer treatment, however mechanisms determining patient response remain poorly understood. Here we used machine learning to predict ICB response from germline and somatic biomarkers and interpreted the learned model to uncover putative mechanisms driving superior outcomes. Patients with higher T follicular helper infiltrates were robust to defects in the class-I Major Histocompatibility Complex (MHC-I). Further investigation uncovered different ICB responses in MHC-I versus MHC-II neoantigen reliant tumors across patients. Despite similar response rates, MHC-II reliant responses were associated with significantly longer durable clinical benefit (Discovery: Median OS=63.6 vs. 34.5 months P=0.0074; Validation: Median OS=37.5 vs. 33.1 months, P=0.040). Characteristics of the tumor immune microenvironment reflected MHC neoantigen reliance, and analysis of immune checkpoints revealed LAG3 as a potential target in MHC-II but not MHC-I reliant responses. This study highlights the value of interpretable machine learning models in elucidating the biological basis of therapy responses.
Collapse
Affiliation(s)
- Timothy Sears
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA USA
| | - Meghana Pagadala
- Biomedical Sciences Program, University of California San Diego, La Jolla, CA,, USA
| | - Andrea Castro
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Ko-han Lee
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA USA
| | - JungHo Kong
- Division of Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Kairi Tanaka
- School of Biological Sciences, University of California San Diego, La Jolla, CA USA
| | - Scott Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, CA USA
| | - Maurizio Zanetti
- Moores Cancer Center, University of California San Diego, La Jolla, CA USA
- The Laboratory of Immunology, Moores Cancer Center and Department of Medicine, University of California San Diego, La Jolla, CA USA
| | - Hannah Carter
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA USA
- The Laboratory of Immunology, Moores Cancer Center and Department of Medicine, University of California San Diego, La Jolla, CA USA
| |
Collapse
|
172
|
Shi M, Nan XR, Liu BQ. The Multifaceted Role of FUT8 in Tumorigenesis: From Pathways to Potential Clinical Applications. Int J Mol Sci 2024; 25:1068. [PMID: 38256141 PMCID: PMC10815953 DOI: 10.3390/ijms25021068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
FUT8, the sole glycosyltransferase responsible for N-glycan core fucosylation, plays a crucial role in tumorigenesis and development. Aberrant FUT8 expression disrupts the function of critical cellular components and triggers the abnormality of tumor signaling pathways, leading to malignant transformations such as proliferation, invasion, metastasis, and immunosuppression. The association between FUT8 and unfavorable outcomes in various tumors underscores its potential as a valuable diagnostic marker. Given the remarkable variation in biological functions and regulatory mechanisms of FUT8 across different tumor types, gaining a comprehensive understanding of its complexity is imperative. Here, we review how FUT8 plays roles in tumorigenesis and development, and how this outcome could be utilized to develop potential clinical therapies for tumors.
Collapse
Affiliation(s)
| | | | - Bao-Qin Liu
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China; (M.S.); (X.-R.N.)
| |
Collapse
|
173
|
Xiang J, Zheng B, Zhao L, He Y, Lou F, Li R, Fu M, Huang X, Zhang W, Hong X, Xiao L, Hu T. Exo70 Promotes the Invasion of Pancreatic Cancer Cells via the Regulation of Exosomes. Cancers (Basel) 2024; 16:336. [PMID: 38254825 PMCID: PMC10813805 DOI: 10.3390/cancers16020336] [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: 12/04/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Pancreatic cancer (PC) is an aggressive and fatal malignant tumor, and exosomes have been reported to be closely related to PC invasion and metastasis. Here we found that Exo70, a key subunit of the exocyst complex, promoted PC metastasis by regulating the secretion of tumor exosomes. Clinical sample studies showed that Exo70 was highly expressed in PC and negatively correlated with patients' survival. Exo70 promoted PC cell lines' invasion and migration. Interestingly, knockdown of Exo70, or using an Exo70 inhibitor (ES2) inhibited the secretion of tumor exosomes and increased the accumulation of cellular vesicles. Furthermore, Exo70 was found to accumulate in the exosomes, which then fused with neighboring PC cells and promoted their invasion. Moreover, Exo70 increased the expression of exosomal PD-L1, leading to the immune escape of PC cells. In vivo, knockdown of Exo70 or treatment with ES2 both decreased the tumor metastasis of PC cells in mice. This study provides new insight into the mechanism of invasion and metastasis in PC and identifies Exo70 as a potential prognostic factor and therapeutic target for PC.
Collapse
Affiliation(s)
- Jingzhou Xiang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
| | - Bowen Zheng
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361102, China
| | - Lingying Zhao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
| | - Yuting He
- Department of Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; (Y.H.); (F.L.); (X.H.)
| | - Fanzhuoran Lou
- Department of Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; (Y.H.); (F.L.); (X.H.)
| | - Runyang Li
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361102, China
| | - Miao Fu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
| | - Xintian Huang
- Department of Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; (Y.H.); (F.L.); (X.H.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
| | - Xiaoting Hong
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
| | - Li Xiao
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
- Department of Oncology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; (Y.H.); (F.L.); (X.H.)
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (J.X.); (B.Z.); (L.Z.); (R.L.); (M.F.); (W.Z.); (X.H.)
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| |
Collapse
|
174
|
Sheng Y, Lei K, Sun C, Liu J, Tu Z, Zhu X, Huang K. Aberrant RBMX expression is relevant for cancer prognosis and immunotherapy response. Aging (Albany NY) 2024; 16:226-245. [PMID: 38214653 PMCID: PMC10817375 DOI: 10.18632/aging.205363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/03/2023] [Indexed: 01/13/2024]
Abstract
Cancer accounts for the highest rates of morbidity and mortality worldwide. RNA binding motif protein X-linked (RBMX) is a nuclear RNA-binding protein, associated with certain types of cancer by participating in the integration of sister chromatids and a combination of ribonucleoprotein complexes. However, the specific role of RBMX in cancer immunity remains unknown. This study presents the aberrant expression levels, single-cell distributions, effective prognostic roles, immune cell infiltration associations, and immunotherapy responses of RBMX as a biomarker in various types of cancer. Moreover, it validates the aberrant expression of RBMX in clinical cancer samples. Furthermore, we also evaluated the relationships between RBMX expression and myeloid-derived suppressor cells in clinical samples by immunofluorescent staining. The results showed that knockdown of RBMX can impair the proliferation, migration, and invasion of liver cancer cells. Finally, we indicated that RBMX may play an immunoregulatory role in cancer progression, affecting the therapeutic effects of immune checkpoint inhibitors in patients with cancer.
Collapse
Affiliation(s)
- Yilei Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- The HuanKui Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Kunjian Lei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, P.R. China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, P.R. China
| | - Chengpeng Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- The HuanKui Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Jia Liu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06511, USA
| | - Zewei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, P.R. China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, P.R. China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, P.R. China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, P.R. China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006, Jiangxi, P.R. China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- JXHC Key Laboratory of Neurological Medicine, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
175
|
Song D, Ding Y. A new target of radiotherapy combined with immunotherapy: regulatory T cells. Front Immunol 2024; 14:1330099. [PMID: 38259489 PMCID: PMC10800811 DOI: 10.3389/fimmu.2023.1330099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Radiotherapy is one important treatment for malignant tumours. It is widely believed today that radiotherapy has not only been used as a local tumour treatment method, but also can induce systemic anti-tumour responses by influencing the tumour microenvironment, but its efficacy is limited by the tumour immunosuppression microenvironment. With the advancement of technology, immunotherapy has entered a golden age of rapid development, gradually occupying a place in clinical tumour treatment. Regulatory T cells (Tregs) widely distributing in the tumour microenvironment play an important role in mediating tumour development. This article analyzes immunotherapy, the interaction between Tregs, tumours and radiotherapy. It briefly introduces immunotherapies targeting Tregs, aiming to provide new strategies for radiotherapy combined with Immunotherapy.
Collapse
Affiliation(s)
| | - Yun Ding
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
176
|
Zhao X, Liu M, Li C, Liu X, Zhao J, Ma H, Zhang S, Qu J. High dose Vitamin C inhibits PD-L1 by ROS-pSTAT3 signal pathway and enhances T cell function in TNBC. Int Immunopharmacol 2024; 126:111321. [PMID: 38041955 DOI: 10.1016/j.intimp.2023.111321] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Vitamin C (VitC) presents excellent anti-tumor effect for long time. Recently, high dose VitC achieved by intravenous administration manifests superior anti-tumor effect. However, the functions and detailed mechanisms of high dose VitC's role in cancer immunity are not fully understood. This study investigates the effect of high dose VitC on PD-L1 expression in triple negative breast cancer (TNBC) and the potential mechanism. Results showed VitC inhibited PD-L1 expression in breast cancer cell lines and enhanced anti-tumor effects of T cells. Furthermore, we found VitC inhibited PD-L1 transcription through ROS-pSTAT3 signal pathways. Consistent with in vitro results, in vivo study showed VitC suppressed tumor growth in immunocompetent mice and enhanced CD8+ T cells infiltration and function in tumor microenvironment. Our findings identify the effects of high dose VitC on PD-L1 expression and provide a rationale for the use of high dose VitC as immunomodulator for cancer therapy.
Collapse
Affiliation(s)
- Xixi Zhao
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Mengjie Liu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Chaofan Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Jiaqi Zhao
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Hongbing Ma
- Department of Radiation Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China.
| | - Jingkun Qu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West Fifth Street, Xi'an 710004, Shaanxi, PR China.
| |
Collapse
|
177
|
Liu Y, Zhu J, Shen J, Lu Y, Pan K, Tong C, Wang Y. A pan-cancer analysis of the prognostic implication and oncogenic role of tubulin epsilon and delta complex 2 (TEDC2) in human tumors. Front Immunol 2024; 14:1272108. [PMID: 38239349 PMCID: PMC10794491 DOI: 10.3389/fimmu.2023.1272108] [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: 08/03/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Tubulin epsilon and delta complex 2 (TEDC2) is widely expressed in various human tissues and primarily governs centriole stability. However, the biological significance of TEDC2 in pan-cancer is unclear. Methods In this study, we employed R software and various online bioinformatics analysis tools to investigate the functional attributes of TEDC2 in human tumours and its potential involvement in immune response. The status of TEDC2 expression was evaluated in samples from the TCGA and GEO datasets, as well as in tumour and corresponding normal samples from the TCGA database. Subsequently, Kaplan-Meier estimates, clinical correlations, and univariate Cox regressions were used to analyze the 33 types of tumors from TCGA and determine the prognostic significance of TEDC2. Moreover, nomogram models were formulated using three distinct tumours, namely kidney renal clear cell carcinoma (KIRC), lung adenocarcinoma (LUAD), and liver hepatocellular carcinoma (LIHC), to evaluate the prognostic significance of TEDC2 in tumours. Furthermore, TEDC2 was investigated for its correlation with the levels of immune cell infiltration, and a functional enrichment analysis was conducted to identify potential signalling pathways involving TEDC2. Results Differential analysis revealed that 16 tumour types expressed TEDC2 to a greater extent than normal tissues. The abnormal expression of TEDC2 can predict survival outcomes in patients with adrenocortical carcinoma (ACC), KIRC, kidney renal papillary cell carcinoma (KIRP), LUAD, LIHC, lower grade glioma (LGG), and thymoma (THYM). Subsequent results indicated that TEDC2 has the ability to influence ECM regulators, cell cycle, and Immune checkpoint-associated signalling pathways, which could potentially lead to a poor prognosis and tumour progression. Discussion TEDC2 has been identified as a potential therapeutic target that could predict the prognosis of multiple tumour types, making it a promising target for reversing tumour development.
Collapse
Affiliation(s)
- Yang Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jie Zhu
- Senior Departments of Urology, the Third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jing Shen
- Department of Endocrinology, the Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yuting Lu
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ke Pan
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chuan Tong
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yao Wang
- Department of Bio-therapeutic, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
178
|
Xing T, Li L, Rao X, Zhao J, Chen Y, Ju G, Xu Y, Gao X, Dong G, Xia X, Guan Y, Zhang L, Wen Z, Liang J. ARID1A deficiency promotes progression and potentiates therapeutic antitumour immunity in hepatitis B virus-related hepatocellular carcinoma. BMC Gastroenterol 2024; 24:11. [PMID: 38166741 PMCID: PMC10759659 DOI: 10.1186/s12876-023-03059-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Exploring predictive biomarkers and therapeutic strategies of ICBs has become an urgent need in clinical practice. Increasing evidence has shown that ARID1A deficiency might play a critical role in sculpting tumor environments in various tumors and might be used as pan-cancer biomarkers for immunotherapy outcomes. The current study aims to explored the immune-modulating role of ARID1A deficiency in Hepatitis B virus (HBV) related hepatocellular carcinoma (HBV-HCC) and its potential immunotherapeutic implications. METHODS In the current study, we performed a comprehensive analysis using bioinformatics approaches and pre-clinical experiments to evaluate the ARID1A regulatory role on the biological behavior, and immune landscape of Hepatitis B virus (HBV) related hepatocellular carcinoma (HBV-HCC). A total of 425 HBV-related hepatocellular carcinoma patients from TCGA-LIHC, AMC and CHCC-HBV cohort were enrolled in bioinformatics analysis. Immunohistochemical staining of HBV-HCC specimens and ARID1A deficiency cellular models were used to validate the results of the analysis. RESULTS Our results have shown that ARID1A deficiency promoted tumor proliferation and metastasis. More importantly, ARID1A deficiency in HBV-HCC was associated with the higher TMB, elevated immune activity, and up-regulated expression of immune checkpoint proteins, especially TIM-3 in HBV-HCC. Further, the expression of Galectin-9, which is the ligand of TIM-3, was elevated in the ARID1A knockout HBV positive cell line. CONCLUSION To conclude, we have shown that the ARID1A deficiency was correlated with more active immune signatures and higher expression of immune checkpoints in HBV-HCC. Additionally, the present study provides insights to explore the possibility of the predictive role of ARID1A in HBV-HCC patients responsive to immunotherapy.
Collapse
Affiliation(s)
- Tao Xing
- Departments of Oncology, Peking University International Hospital, 1 Life Park Road, Life Science Park of Zhongguancun, Changping, Beijing, 102206, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China
| | - Li Li
- Departments of Oncology, Peking University International Hospital, 1 Life Park Road, Life Science Park of Zhongguancun, Changping, Beijing, 102206, China
| | - Xiaosong Rao
- HAINAN YILING Medical Industry Development Co.,Ldt, Qionghai, Hainan, 571442, China
| | - Jing Zhao
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, 72074, Germany
| | - Yiran Chen
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Gaoda Ju
- Departments of Oncology, Peking University International Hospital, 1 Life Park Road, Life Science Park of Zhongguancun, Changping, Beijing, 102206, China
| | - Yaping Xu
- Geneplus-Beijing Institute, Beijing, 102206, China
| | - Xuan Gao
- Geneplus-Beijing Institute, Beijing, 102206, China
| | - Guilan Dong
- Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Xuefeng Xia
- Geneplus-Beijing Institute, Beijing, 102206, China
| | - Yanfang Guan
- Geneplus-Beijing Institute, Beijing, 102206, China
| | - Lingling Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Zhenping Wen
- Inner Mongolia Cancer Hospital, 42 Zhaowuda Road, Saihan District, Hohhot, Inner Mongolia, 010020, P. R. China.
| | - Jun Liang
- Departments of Oncology, Peking University International Hospital, 1 Life Park Road, Life Science Park of Zhongguancun, Changping, Beijing, 102206, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, No. 52, Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
179
|
Chen Y, Shen C, Wu J, Yan X, Huang Q. Role of immune related genes in predicting prognosis and immune response in patients with hepatocellular carcinoma. J Biochem Mol Toxicol 2024; 38:e23519. [PMID: 37665680 DOI: 10.1002/jbt.23519] [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: 10/20/2022] [Revised: 06/25/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Abstract
Immunotherapy has developed rapidly in recent years. This study aimed to establish a prognostic signature for immune-related genes (IRGs) and explore related potential immunotherapies. The RNA-seq transcriptome profiles and clinicopathological information of patients were obtained from The Cancer Genome Atlas. Differentially expressed IRGs in tumors and normal tissues were screened and a risk score signature was constructed to predict the prognosis in patients with hepatocellular carcinoma (HCC). Receiver operating characteristic curves, survival analyses, and correlation analyses were used to explore the clinical application of this model. We further analyzed the differences in clinical characteristics, immune infiltration, somatic mutations, and treatment sensitivity between the high- and low-risk populations characterized by the prognostic models. The immune cell infiltration score and immune-related pathway activity were calculated using the single sample gene set enrichment analysis (ssGSEA) set enrichment analysis. Gene ontology (GO), Kyoto encyclopedia of genes and genomes, and GSEA were used to explore the underlying mechanisms. We constructed a nine-IRG formula to predict the prognosis in HCC patients. The higher the risk score, the higher the malignancy of the tumor and the worse the prognosis. There were significant differences in immune related processes between the high- and low-risk groups. TP53 and CTNNB1 mutations were significantly different between different risk groups. The expression of model gene was closely related to the sensitivity of tumor cells to chemotherapeutic drugs. This risk score model, which is helpful for the individualized treatment of patients with different risk factors, could be a reliable prognostic tool for HCC patients.
Collapse
Affiliation(s)
- Yi Chen
- Departments of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People's Republic of China
| | - Chuchen Shen
- Departments of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People's Republic of China
| | - Juju Wu
- Departments of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People's Republic of China
| | - Xiaodan Yan
- Departments of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People's Republic of China
| | - Qin Huang
- Departments of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, People's Republic of China
| |
Collapse
|
180
|
Nguyen CTT, Van TNK, Huong PT. Predictability of Neutrophile to Lymphocyte Ratio and Platelet to Lymphocyte Ratio on the Effectiveness of Immune Checkpoint Inhibitors in Non-small Cell Lung Cancer patients: A Meta-Analysis. Cancer Control 2024; 31:10732748241285474. [PMID: 39285591 PMCID: PMC11406641 DOI: 10.1177/10732748241285474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVES The associations between the neutrophil-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) with the responses of non-small cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICI) and the NLR/PLR predictive potential were evaluated via meta-analysis. METHODS A systematic review was conducted using the PubMed, Embase, and The Cochrane Library databases until October 2021. The relationship between NLR/PLR and overall survival (OS) and progression-free survival (PFS) was evaluated using pooled hazard ratios (HR). The relationship between NLR/PLR and overall response rate (ORR) and disease control rate (DCR) was assessed via pooled odds ratios (OR). Heterogeneity between studies, publication bias, subgroup and sensitivity analyses, trim and fill meta-analysis, and the contour-enhanced funnel plot were performed using the R software. RESULTS A total of 44 (out of 875) studies met the eligibility criteria, providing a sample size of 4597 patients. Patients with a high NLR were statistically significantly associated with worse outcomes, including OS (pooled HR = 2.44; P < 0.001), PFS (pooled HR = 2.06; P < 0.001), DCR (pooled OR = 0.71; P < 0.001), and ORR (pooled OR = 0.33; P < 0.001). Similarly, a high PLR was associated with poorer outcomes in response to ICI drugs, including OS (pooled HR = 2.13; P < 0.001) and PFS (pooled HR = 1.61; P < 0.001). CONCLUSION High NLR and PLR were associated with a statistically significant reduction in the efficacy of ICI drugs in NSCLC patients. Thereby, it is possible to use NLR and PLR as potential and available biomarkers in the clinical practice to predict the outcome of ICI treatment in NSCLC patients.
Collapse
Affiliation(s)
- Cuc Thi Thu Nguyen
- Faculty of Pharmaceutical Management and Economics, Hanoi University of Pharmacy, Hanoi, Vietnam
| | | | - Phung Thanh Huong
- Faculty of Biotechnology, Hanoi University of Pharmacy, Hanoi, Vietnam
| |
Collapse
|
181
|
Kikuchi Y, Shimada H, Hatanaka Y, Kinoshita I, Ikarashi D, Nakatsura T, Kitano S, Naito Y, Tanaka T, Yamashita K, Oshima Y, Nanami T. Clinical practice guidelines for molecular tumor markers, 2nd edition review part 1. Int J Clin Oncol 2024; 29:1-19. [PMID: 38019341 DOI: 10.1007/s10147-023-02430-x] [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: 08/31/2023] [Accepted: 10/14/2023] [Indexed: 11/30/2023]
Abstract
With advances in gene and protein analysis technologies, many target molecules that may be useful in cancer diagnosis have been reported. Therefore, the "Tumor Marker Study Group" was established in 1981 with the aim of "discovering clinically" useful molecules. Later, the name was changed to "Japanese Society for Molecular Tumor Marker Research" in 2000 in response to the remarkable progress in gene-related research. Currently, the world of cancer treatment is shifting from the era of representative tumor markers of each cancer type used for tumor diagnosis and treatment evaluation to the study of companion markers for molecular-targeted therapeutics that target cancer cells. Therefore, the first edition of the Molecular Tumor Marker Guidelines, which summarizes tumor markers and companion markers in each cancer type, was published in 2016. After publication of the first edition, the gene panel testing using next-generation sequencing became available in Japan in June 2019 for insured patients. In addition, immune checkpoint inhibitors have been indicated for a wide range of cancer types. Therefore, the 2nd edition of the Molecular Tumor Marker Guidelines was published in September 2021 to address the need to revise the guidelines. Here, we present an English version of the review (Part 1) of the Molecular Tumor Marker Guidelines, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan.
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Hokkaido, Japan
| | - Daiki Ikarashi
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shigehisa Kitano
- Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Chiba, Japan
| | - Toshimichi Tanaka
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Tokyo, Japan
| | - Keishi Yamashita
- Division of Advanced Surgical Oncology, Department of Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Tokyo, Japan
| | - Yoko Oshima
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| | - Tatsuki Nanami
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| |
Collapse
|
182
|
Ramamurthy C, Wheeler KM, Trecarten S, Hassouneh Z, Ji N, Lee Y, Svatek RS, Mukherjee N. Perioperative Immune Checkpoint Blockade for Muscle-Invasive and Metastatic Bladder Cancer. JOURNAL OF CANCER IMMUNOLOGY 2024; 6:29-39. [PMID: 38784962 PMCID: PMC11113005 DOI: 10.33696/cancerimmunol.6.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Checkpoint inhibitors offer promise in treating muscle-invasive and metastatic bladder cancer, but the optimal timing of their administration-neoadjuvant or adjuvant-remains unclear. To determine the efficacy of combining checkpoint inhibition with standard cisplatin-based chemotherapy, we conducted a phase II trial of neoadjuvant anti-PD-1 (αPD-1) and anti-CTLA-4 (αCTLA-4), in combination with cisplatin-gemcitabine, for patients with muscle-invasive bladder cancer prior to radical cystectomy. In addition, a novel murine model of spontaneous metastatic bladder cancer was used to compare the efficacy of neoadjuvant versus adjuvant anti-PD-L1 (αPD-L1) treatment. The clinical trial was closed prematurely due to the industry's withdrawal of drug provision. Adverse events were observed in all patients; however, serious adverse events were not observed in any patient. A complete pathologic response was observed in 50% of the 4 patients enrolled. Response to treatment was significantly associated with elevated urinary T cells including CD8+ and IFNγ+ CD4+ T cells, suggesting potential reinforcement of immune responses by neoadjuvant αPD-1 and αCTLA-4 against bladder tumor cells. These findings suggest that combining chemotherapy and immunotherapy in the neoadjuvant setting could be safe. However, the complete response rate of this four-drug regimen was modest and emphasizes the need for randomized controlled trials to properly assess immunotherapy efficacy in the neoadjuvant setting. In corresponding murine studies, the MB49-met model consistently displayed widespread metastasis, including tumor growth in the lungs, liver, and bowel mesentery, within 20 days of subcutaneous transplantation. Mice receiving surgery plus neoadjuvant αPD-L1 or adjuvant αPD-L1 exhibited improved survival compared to those receiving only αPD-L1. However, no significant difference in survival was observed between the neoadjuvant and adjuvant αPD-L1 cohorts. Furthermore, the timing of neoadjuvant therapy administration (early vs. late) did not significantly impact survival. This study highlights the potential of perioperative immunotherapy in the treatment of locally advanced and metastatic bladder cancer.
Collapse
Affiliation(s)
- Chethan Ramamurthy
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Karen M. Wheeler
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Shaun Trecarten
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Zaineb Hassouneh
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Niannian Ji
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Yifen Lee
- Department of Urology and Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Robert S. Svatek
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Neelam Mukherjee
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| |
Collapse
|
183
|
Zhang L, Wang Y, Wang L, Wang M, Li S, He J, Ji J, Li K, Cao L. Identifying survival of pan-cancer patients under immunotherapy using genomic mutation signature with large sample cohorts. J Mol Med (Berl) 2024; 102:69-79. [PMID: 37978056 DOI: 10.1007/s00109-023-02398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Although immune checkpoint inhibitors have led to durable clinical response in multiple cancers, only a small proportion of patients respond to this treatment. Therefore, we aim to develop a predictive model that utilizes gene mutation profiles to accurately identify the survival of pan-cancer patients with immunotherapy. Here, we develop and evaluate three different nomograms using two cohorts containing 1,594 cancer patients whose mutation profiles are obtained by MSK-IMPACT sequencing and 230 cancer patients receiving whole-exome sequencing, respectively. Using eighteen genes (SETD2, BRAF, NCOA3, LATS1, IL7R, CREBBP, TET1, EPHA7, KDM5C, MET, KMT2D, RET, PAK7, CSF1R, JAK2, FAT1, ASXL1 and SPEN), the first nomogram stratifies patients from both cohorts into High-Risk and Low-Risk groups. Pan-cancer patients in the High-Risk group exhibit significantly shorter overall survival and progression-free survival than patients in the Low-Risk group in both cohorts. Meanwhile, the first nomogram also accurately identifies the survival of patients with melanoma or lung cancer undergoing immunotherapy, or pan-cancer patients treated with anti-PD-1/PD-L1 inhibitor or anti-CTLA-4 inhibitor. The model proposed is not a prognostic model for the survival of pan-cancer patients without immunotherapy, but a simple, effective and robust predictive model for pan-cancer patients' survival under immunotherapy, and could provide valuable assistance for clinical practice.
Collapse
Affiliation(s)
- Liuchao Zhang
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Yuanyuan Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Liuying Wang
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Meng Wang
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Shuang Li
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Jia He
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Jianxin Ji
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China
| | - Kang Li
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China.
| | - Lei Cao
- Department of Epidemiology and Biostatistics, Public Health College, Harbin Medical University, Harbin, Hei Longjiang province, 150081, China.
| |
Collapse
|
184
|
Singh T, Bhattacharya M, Mavi AK, Gulati A, Rakesh, Sharma NK, Gaur S, Kumar U. Immunogenicity of cancer cells: An overview. Cell Signal 2024; 113:110952. [PMID: 38084844 DOI: 10.1016/j.cellsig.2023.110952] [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: 09/28/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023]
Abstract
The immune system assumes a pivotal role in the organism's capacity to discern and obliterate malignant cells. The immunogenicity of a cancer cell pertains to its proficiency in inciting an immunological response. The prowess of immunogenicity stands as a pivotal determinant in the triumph of formulating immunotherapeutic methodologies. Immunotherapeutic strategies include immune checkpoint inhibitors, chimeric antigen receptor (CAR) T-cell therapy, and on vaccines. Immunogenic cell death (ICD) epitomizes a form of cellular demise that incites an immune response against dying cells. ICD is characterized by the liberation of distinct specific molecules that activate the immune system, thereby leading to the identification and elimination of dying cells by immunocytes. One of the salient characteristics inherent to the ICD phenomenon resides in the vigorous liberation of adenosine triphosphate (ATP) by cellular entities dedicated to embarking upon the process of programmed cell death, yet refraining from complete apoptotic demise. ICD is initiated by a sequence of molecular events that occur during cell death. These occurrences encompass the unveiling or discharge of molecules such as calreticulin, high-mobility group box 1 (HMGB1), and adenosine triphosphate (ATP) from dying cells. These molecules act as "eat me" signals, which are recognized by immune cells, thereby prompting the engulfment and deterioration of expiring cells by phagocytes including various pathways such as Necroptosis, Apoptosis, and pyroptosis. Here, we review our current understanding of the pathophysiological importance of the immune responses against dying cells and the mechanisms underlying their activation. Overall, the ICD represents an important mechanism by which the immune system recognizes and eliminates dying cells, including cancer cells. Understanding the molecular events that underlie ICD bears the potential to engender innovative cancer therapeutics that harness the power of the immune system to combat cancer.
Collapse
Affiliation(s)
- Tanya Singh
- Department of Microbiology, Ram Lal Anand College, University of Delhi, Delhi 110021, India
| | - Madhuri Bhattacharya
- Department of Microbiology, Ram Lal Anand College, University of Delhi, Delhi 110021, India
| | - Anil Kumar Mavi
- Department of Botany, Sri Aurobindo College, University of Delhi, Delhi 110017, India.
| | - Anita Gulati
- Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi 110078, India
| | - Rakesh
- Janki Devi Memorial College, University of Delhi, Delhi 110060, India
| | - Naresh Kumar Sharma
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sonal Gaur
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH9, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh 201015, India.
| |
Collapse
|
185
|
Ye G, Tu L, Li Z, Li X, Zheng X, Song Y. SYNPO2 promotes the development of BLCA by upregulating the infiltration of resting mast cells and increasing the resistance to immunotherapy. Oncol Rep 2024; 51:14. [PMID: 38038167 PMCID: PMC10758676 DOI: 10.3892/or.2023.8673] [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: 05/11/2023] [Accepted: 09/27/2023] [Indexed: 12/02/2023] Open
Abstract
Synaptopodin 2 (SYNPO2) plays a pivotal role in regulating tumor growth, development and progression in bladder urothelial Carcinoma (BLCA). However, the precise biological functions and mechanisms of SYNPO2 in BLCA remain unclear. Based on TCGA database‑derived BLCA RNA sequencing data, survival analysis and prognosis analysis indicate that elevated SYNPO2 expression was associated with poor survival outcomes. Notably, exogenous SYNPO2 expression significantly promoted tumor invasion and migration by upregulating vimentin expression in BLCA cell lines. Enrichment analysis revealed the involvement of SYNPO2 in humoral immune responses and the PI3K/AKT signaling pathway. Moreover, increased SYNPO2 levels increased the sensitivity of BLCA to PI3K/AKT pathway‑targeted drugs while being resistant to conventional chemotherapy. In in vivo BLCA mouse models, SYNPO2 overexpression increased pulmonary metastasis of 5637 cells. High SYNPO2 expression led to increased infiltration of innate immune cells, particularly mast cells, in both nude mouse model and clinical BLCA samples. Furthermore, tumor immune dysfunction and exclusion score showed that patients with BLCA patients and high SYNPO2 expression exhibited worse clinical outcomes when treated with immune checkpoint inhibitors. Notably, in the IMvigor 210 cohort, SYNPO2 expression was significantly associated with the population of resting mast cells in BLCA tissue following PD1/PDL1 targeted therapy. In conclusion, SYNPO2 may be a promising prognostic factor in BLCA by modulating mast cell infiltration and exacerbating resistance to immune therapy and conventional chemotherapy.
Collapse
Affiliation(s)
- Gongjie Ye
- Department of Critical Care Medicine, Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Linglan Tu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310012, P.R. China
| | - Zhuduo Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310012, P.R. China
| | - Xiangyu Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310012, P.R. China
| | - Xiaoliang Zheng
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310012, P.R. China
| | - Yongfei Song
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, P.R. China
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310012, P.R. China
- Medical College, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
186
|
Xie Y, Pan X, Wang Z, Ma H, Xu W, Huang H, Zhang J, Wang X, Lian C. Multi-omics identification of GPCR gene features in lung adenocarcinoma based on multiple machine learning combinations. J Cancer 2024; 15:776-795. [PMID: 38213730 PMCID: PMC10777041 DOI: 10.7150/jca.90990] [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: 10/10/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024] Open
Abstract
Background: Lung adenocarcinoma is a common malignant tumor that ranks second in the world and has a high mortality rate. G protein-coupled receptors (GPCRs) have been reported to play an important role in cancer; however, G protein-coupled receptor-associated features have not been adequately investigated. Methods: In this study, GPCR-related genes were screened at single-cell and bulk transcriptome levels based on AUcell, single-sample gene set enrichment analysis (ssGSEA) and weighted gene co-expression network (WGCNA) analysis. And a new machine learning framework containing 10 machine learning algorithms and their multiple combinations was used to construct a consensus G protein-coupled receptor-related signature (GPCRRS). GPCRRS was validated in the training set and external validation set. We constructed GPCRRS-integrated nomogram clinical prognosis prediction tools. Multi-omics analyses included genomics, single-cell transcriptomics, and bulk transcriptomics to gain a more comprehensive understanding of prognostic features. We assessed the response of risk subgroups to immunotherapy and screened for personalized drugs targeting specific risk subgroups. Finally, the expression of key GPCRRS genes was verified by RT-qPCR. Results: In this study, we identified 10 GPCR-associated genes that were significantly associated with the prognosis of lung adenocarcinoma by single-cell transcriptome and bulk transcriptome. Univariate and multivariate showed that the survival rate was higher in low risk than in high risk, which also suggested that the model was an independent prognostic factor for LUAD. In addition, we observed significant differences in biological function, mutational landscape, and immune cell infiltration in the tumor microenvironment between high and low risk groups. Notably, immunotherapy was also relevant in the high and low risk groups. In addition, potential drugs targeting specific risk subgroups were identified. Conclusion: In this study, we constructed and validated a lung adenocarcinoma G protein-coupled receptor-related signature, which has an important role in predicting the prognosis of lung adenocarcinoma and the effect of immunotherapy. It is hypothesized that LDHA, GPX3 and DOCK4 are new potential targets for lung adenocarcinoma, which can achieve breakthroughs in prognosis prediction, targeted prevention and treatment of lung adenocarcinoma and provide important guidance for anti-tumor.
Collapse
Affiliation(s)
- Yiluo Xie
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center pulmonary critical care medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Xinyu Pan
- Department of Medical Imaging, Bengbu Medical College, Bengbu 233030, China
| | - Ziqiang Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China
| | - Hongyu Ma
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Wanjie Xu
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Hua Huang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu 233000, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center pulmonary critical care medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu 233030, China
| |
Collapse
|
187
|
Musella M, Manduca N, Maccafeo E, Ruggiero E, Sistigu A. In Vitro Evaluation of Cancer Cell Immunogenicity and Antigen-Specific T-Cell Cytotoxicity by Flow Cytometry. Methods Mol Biol 2024; 2748:13-28. [PMID: 38070104 DOI: 10.1007/978-1-0716-3593-3_2] [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] [Indexed: 12/18/2023]
Abstract
A cardinal principle of oncoimmunology is that cancer cells can be eliminated by tumor-infiltrating cytotoxic CD8 T lymphocytes. This has been widely demonstrated during the last 20 years and also recently harnessed for therapy. However, emerging evidence indicates that even neoplasms showing striking initial responses to conventional and targeted (immuno)therapies often acquire resistance, resulting in tumor relapse, increased aggressiveness, and metastatization. Indeed, tumors are complex ecosystems whose malignant and nonmalignant cells, constituting the tumor microenvironment, constantly interact and evolve in space and time. Together with patient's own genetic factors, such environmental interplays may curtail antitumor immune responses leading to cancer immune evasion and natural/acquired (immuno)therapy resistance. In this context, cancer stem cells (CSCs) are thought to be the roots of therapy failure. Flow cytometry is a powerful technology that finds extensive applications in cancer biology. It offers several unique advantages as it allows the rapid, quantitative, and multiparametric analysis of cell populations or functions at the single-cell level. In this chapter, we discuss a two-color flow cytometric protocol to evaluate cancer cell immunogenicity by analyzing the proliferative and tumor-killing potential of ovalbumin (OVA)-specific CD8 OT-1 T cells exposed to OVA-expressing MCA205 sarcoma cells and their CSC counterparts.
Collapse
Affiliation(s)
- Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
188
|
Li Z, Zheng C, Liu H, Lv J, Wang Y, Zhang K, Kong S, Chen F, Kong Y, Yang X, Cheng Y, Yang Z, Zhang C, Tian Y. A novel oxidative stress-related gene signature as an indicator of prognosis and immunotherapy responses in HNSCC. Aging (Albany NY) 2023; 15:14957-14984. [PMID: 38157249 PMCID: PMC10781479 DOI: 10.18632/aging.205323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE To identify molecular subtypes of oxidative stress-related genes in head and neck squamous cell carcinoma (HNSCC) and to construct a scoring model of oxidative stress-related genes. METHODS R language based scRNA-seq and bulk RNA-seq analyses were used to identify molecular isoforms of oxidative stress-related genes in HNSCC. An oxidative stress-related gene scoring (OSRS) model was constructed, which were verified through online data and immunohistochemical staining of clinical samples. RESULTS Using TCGA-HNSCC datasets, nine predictive genes for overall patient survival, rarely reported in previous similar studies, were screened. AREG and CES1 were identified as prognostic risk factors. CSTA, FDCSP, JCHAIN, IFFO2, PGLYRP4, SPOCK2 and SPINK6 were identified as prognostic factors. Collectively, all genes formed a prognostic risk signature model for oxidative stress in HNSCC, which were validated in GSE41613, GSE103322 and PRJEB23709 datasets. Immunohistochemical staining of SPINK6 in nasopharyngeal cancer samples validated the gene panel. Subsequent analysis indicated that subgroups of the oxidative stress prognostic signature played important roles during cellular communication, the immune microenvironment, the differential activation of transcription factors, oxidative stress and immunotherapeutic responses. CONCLUSIONS The risk model might predict HNSCC prognosis and immunotherapeutic responses.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
| | - Chunning Zheng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Hongtao Liu
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Clinical Pathology, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, Shandong 250014, P.R. China
| | - Jiling Lv
- Department of Respiratory and Critical Care Medicine, Shandong Second Provincial General Hospital, Jinan, Shandong 250299, P.R. China
| | - Yuanyuan Wang
- Department of Oncology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250299, P.R. China
| | - Kai Zhang
- Generalsurgery Department, Wenshang County People’s Hospital, Wenshang, Shandong 272500, P.R. China
| | - Shuai Kong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Feng Chen
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yongmei Kong
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
| | - Xiaowei Yang
- Department of Hepatobiliary Intervention, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P.R. China
| | - Yuxia Cheng
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Clinical Pathology, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, Shandong 250014, P.R. China
| | - Zhensong Yang
- Department of Gastrointestinal Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Chi Zhang
- Department of Cardiology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yuan Tian
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
- Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong University, Jinan, Shandong 250299, P.R. China
| |
Collapse
|
189
|
Wang Y, Huang Q, Zhong G, Lv J, Guo Q, Ma Y, Wang X, Zeng J. Sequential PET/CT and pathological biomarker crosstalk predict response to PD-1 blockers alone or combined with sunitinib in propensity score-matched cohorts of cancer of unknown primary treatment. Front Oncol 2023; 13:1191611. [PMID: 38205137 PMCID: PMC10777842 DOI: 10.3389/fonc.2023.1191611] [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: 03/22/2023] [Accepted: 10/26/2023] [Indexed: 01/12/2024] Open
Abstract
Introduction The efficacy of immune checkpoint inhibitors (ICIs), including toripalimab and pembrolizumab, has not been confirmed in the treatment of cancer of unknown primary (CUP), which has a very poor prognosis. Combined with anti-angiogenic therapies, ICIs are hypothesized to be effective in prolonging overall survival. The study aims to give evidence on the treatment effects of sunitinib combined with ICIs, find pathological biomarkers associated with changes in volumetric 18F FDG PET/CT parameters, and investigate inner associations among these markers associated with response on PET/CT. Methods The study recruited patients receiving combined treatment (ICIs + sunitinib), compared the effects of combined treatment with those of separate treatment and age-matched negative controls, and analyzed propensity score-matched (PSM) pairs. Markers associated with survival were identified, and their inner associations were tested using structural equation modeling. Results A total of 292 patients were enrolled in the final analysis, with 53 patients receiving combined treatment. Survival analysis demonstrated significantly prolonged survival in either combined or separate treatment, with the combined arm showing better response when PSM-paired using pre-treatment whole-body PET/CT parameters. The angiogenic markers KDR and VEGF mediate the PD-1 blockade impact on volumetric value changes in positive and negative manners. Conclusion The anti-angiogenic agent sunitinib may potentiate PD-1 blockade by diminishing angiogenesis or its downstream effects. The combined separate treatment increased the survival of CUP patients, and the responses could be evaluated using volumetric PET/CT parameters.
Collapse
Affiliation(s)
- Youlong Wang
- Hainan Hospital of PLA General Hospital, Department of General Surgery, Haitang District, Sanya, China
| | - Qi Huang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guanqing Zhong
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Lv
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qinzhi Guo
- Pancreas Center of Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Yifei Ma
- Department of Spine Surgery, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xinjia Wang
- Department of Spine Surgery, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Jiling Zeng
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Center, Guangzhou, China
| |
Collapse
|
190
|
Zhang J, Chen L, Wei W, Mao F. Long non-coding RNA signature for predicting gastric cancer survival based on genomic instability. Aging (Albany NY) 2023; 15:15114-15133. [PMID: 38127056 PMCID: PMC10781445 DOI: 10.18632/aging.205336] [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: 09/09/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Gastric cancer is a prevalent type of tumor with a poor prognosis. Given the high occurrence of genomic instability in gastric cancer, it is essential to investigate the prognostic significance of genes associated with genomic instability in this disease. METHODS We identified genomic instability-related lncRNAs (GInLncRNAs) by analyzing somatic mutation and transcriptome profiles. We evaluated co-expression and enrichment using various analyses, including univariate COX analysis and LASSO regression. Based on these findings, we established an lncRNA signature associated with genomic instability, which we subsequently assessed for prognostic value, immune cell and checkpoint analysis, drug sensitivity, and external validation. Finally, PCR assay was used to verify the expression of key lncRNAs. RESULTS Our study resulted in the establishment of a seven-lncRNA prognostic signature, including PTENP1-AS, LINC00163, RP11-169F17.1, C8ORF87, RP11-389G6.3, LINCO1210, and RP11-115H13.1. This signature exhibited independent prognostic value and was associated with specific immune cells and checkpoints in gastric cancer. Additionally, the model was correlated with somatic mutation and several chemotherapeutic drugs. We further confirmed the prognostic value of LINC00163, which was included in our model, in an independent dataset. Our model demonstrated superior performance compared to other models. PCR showed that LINC00163 was significantly up-regulated in 4 adjacent normal tissues compared with the GC tissues. CONCLUSIONS Our study resulted in the establishment of a seven-lncRNA signature associated with genomic instability, which demonstrated robust prognostic value in predicting the prognosis of gastric cancer. The signature also identified potential chemotherapeutic drugs, making it a valuable tool for clinical decision-making and medication use.
Collapse
Affiliation(s)
- Jialing Zhang
- Department of Gastroenterology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, People’s Republic of China
| | - Liang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Conversion Therapy Center for Hepatobiliary and Pancreatic Tumors, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, P.R. China
| | - Wei Wei
- Department of Anesthesiology and Pain Research Center, The First Hospital of Jiaxing or The Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Fei Mao
- Department of Urology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, People’s Republic of China
| |
Collapse
|
191
|
Raza F, Zafar H, Jiang L, Su J, Yuan W, Qiu M, Paiva-Santos AC. Progress of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. Biomater Sci 2023; 12:57-91. [PMID: 37902579 DOI: 10.1039/d3bm01170d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
In recent years, considerable attention has been given to phototherapy, including photothermal and photodynamic therapy to kill tumor cells by producing heat or reactive oxygen species (ROS). It has the high merits of noninvasiveness and limited drug resistance. To fully utilize this therapy, an extraordinary nanovehicle is required to target phototherapeutic agents in the tumor cells. Nanovesicles embody an ideal strategy for drug delivery applications. Cell membrane-derived biomimetic nanovesicles represent a developing type of nanocarrier. Combining this technique with cancer phototherapy could enable a novel strategy. Herein, efforts are made to describe a comprehensive overview of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. The description in this review is mainly based on representative examples of exosome-derived biomimetic nanomedicine research, ranging from their comparison with traditional nanocarriers to extensive applications in cancer phototherapy. Additionally, the challenges and future prospectives for translating these for clinical application are discussed.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Liangdi Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
192
|
Torres-Martínez S, Calabuig-Fariñas S, Gallach S, Mosqueda M, Munera-Maravilla E, Sirera R, Navarro L, Blasco A, Camps C, Jantus-Lewintre E. Circulating Immune Proteins: Improving the Diagnosis and Clinical Outcome in Advanced Non-Small Cell Lung Cancer. Int J Mol Sci 2023; 24:17587. [PMID: 38139416 PMCID: PMC10743468 DOI: 10.3390/ijms242417587] [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: 09/25/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Immunotherapy has been proven a viable treatment option for non-small cell lung cancer (NSCLC) treatment in patients. However, some patients still do not benefit. Finding new predictive biomarkers for immunocheckpoint inhibitor (ICI) response will improve treatment management in the clinical routine. In this regard, liquid biopsy is a useful and noninvasive alternative to surgical biopsies. In the present study, we evaluated the potential diagnostic, prognostic, and predictive value of seven different soluble mediators involved in immunoregulation. Fifty-two plasma samples from advanced NSCLC treated in first-line with pembrolizumab at baseline (PRE) and at first response assessment (FR) were analyzed. In terms of diagnostic value, our results revealed that sFGL1, sGAL-3, and sGAL-1 allowed for optimal diagnostic efficacy for cancer patients. Additionally, the combination of sFGL1 and sGAL-3 significantly improved diagnostic accuracy. Regarding the predictive value to assess patients' immune response, sCD276 levels at PRE were significantly higher in patients without tumor response (p = 0.035). Moreover, we observed that high levels of sMICB at PRE were associated with absence of clinical benefit (pembrolizumab treatment less than 6 months) (p = 0.049), and high levels of sMICB and sGAL-3 at FR are also related to a lack of clinical benefit (p = 0.027 and p = 0.03, respectively). Finally, in relation to prognosis significance, at PRE and FR, sMICB levels above the 75th percentile are related to poor progression-free survival (PFS) (p = 0.013 and p = 0.023, respectively) and overall survival (OS) (p = 0.001 and p = 0.011, respectively). An increase in sGAL3 levels at FR was associated with worse PFS (p = 0.037). Interestingly, high sGAL-3 at PRE was independently associated with PFS and OS with a hazard ratio (HR) of 2.45 (95% CI 1.14-5.25; p = 0.021) and 4.915 (95% CI 1.89-12.73; p = 0.001). In conclusion, plasma levels of sFGL1, sGAL-3, and sGAL-1 could serve as diagnostic indicators and sMICB, sCD276, and sGAL3 were linked to outcomes, suggesting their potential in assessing NSCLC under pembrolizumab treatment. Our results highlight the value of employing soluble immune biomarkers in advanced lung cancer patients treated with pembrolizumab at first-line.
Collapse
Affiliation(s)
- Susana Torres-Martínez
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Pathology, Universitat de València, 46010 Valencia, Spain
| | - Sandra Gallach
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Marais Mosqueda
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Ester Munera-Maravilla
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Rafael Sirera
- Department of Biotechnology, Universitat Politècnica de València, 46022 Valencia, Spain;
| | - Lara Navarro
- Department of Pathology, Hospital General Universitario de Valencia, 46014 Valencia, Spain;
| | - Ana Blasco
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Department of Medicine, Universitat de València, 46010 Valencia, Spain
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.T.-M.); (S.G.); (M.M.); (E.M.-M.); (C.C.); (E.J.-L.)
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe—Fundación Investigación Hospital General Universitario de Valencia, 46014 Valencia, Spain;
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Biotechnology, Universitat Politècnica de València, 46022 Valencia, Spain;
- Nanomedicine, Centro Investigación Príncipe Felipe—Universitat Politècnica de Valencia, 46022 Valencia, Spain
| |
Collapse
|
193
|
Yue M, Hu S, Sun H, Tuo B, Jia B, Chen C, Wang W, Liu J, Liu Y, Sun Z, Hu J. Extracellular vesicles remodel tumor environment for cancer immunotherapy. Mol Cancer 2023; 22:203. [PMID: 38087360 PMCID: PMC10717809 DOI: 10.1186/s12943-023-01898-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Tumor immunotherapy has transformed neoplastic disease management, yet low response rates and immune complications persist as major challenges. Extracellular vesicles including exosomes have emerged as therapeutic agents actively involved in a diverse range of pathological conditions. Mounting evidence suggests that alterations in the quantity and composition of extracellular vesicles (EVs) contribute to the remodeling of the immune-suppressive tumor microenvironment (TME), thereby influencing the efficacy of immunotherapy. This revelation has sparked clinical interest in utilizing EVs for immune sensitization. In this perspective article, we present a comprehensive overview of the origins, generation, and interplay among various components of EVs within the TME. Furthermore, we discuss the pivotal role of EVs in reshaping the TME during tumorigenesis and their specific cargo, such as PD-1 and non-coding RNA, which influence the phenotypes of critical immune cells within the TME. Additionally, we summarize the applications of EVs in different anti-tumor therapies, the latest advancements in engineering EVs for cancer immunotherapy, and the challenges encountered in clinical translation. In light of these findings, we advocate for a broader understanding of the impact of EVs on the TME, as this will unveil overlooked therapeutic vulnerabilities and potentially enhance the efficacy of existing cancer immunotherapies.
Collapse
Affiliation(s)
- Ming Yue
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Baojing Tuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chen Chen
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yang Liu
- Department of Radiotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450001, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
194
|
Akashi Y, Yamamoto Y, Hashimoto M, Adomi S, Fujita K, Kiba K, Minami T, Yoshimura K, Hirayama A, Uemura H. Prognostic Factors of Platinum-Refractory Advanced Urothelial Carcinoma Treated with Pembrolizumab. Cancers (Basel) 2023; 15:5780. [PMID: 38136326 PMCID: PMC10742147 DOI: 10.3390/cancers15245780] [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: 11/01/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
INTRODUCTION Immune checkpoint inhibitor (ICI) therapy has significantly improved the prognosis of some patients with advanced urothelial carcinoma (UC), but it does not provide high therapeutic efficacy in all patients. Therefore, identifying predictive biomarkers is crucial in determining which patients are candidates for ICI treatment. This study aimed to identify the predictors of ICI treatment response in patients with platinum-refractory advanced UC treated with pembrolizumab. METHODS Patients with platinum-refractory advanced UC who had received pembrolizumab at two hospitals in Japan were included. Univariate and multivariate analyses were performed to identify biomarkers for progression-free survival (PFS) and overall survival (OS). RESULTS Forty-one patients were evaluable for this analysis. Their median age was 75 years, and the vast majority of the patients were male (85.4%). The objective response rate was 29.3%, with a median overall survival (OS) of 17.8 months. On multivariate analysis, an Eastern Cooperative Oncology Group performance status (ECOG-PS) ≥ 2 (HR = 6.33, p = 0.03) and a baseline neutrophil-to-lymphocyte ratio (NLR) > 3 (HR = 2.79, p = 0.04) were significantly associated with poor OS. Antibiotic exposure did not have a significant impact on either PFS or OS. CONCLUSIONS ECOG-PS ≥ 2 and baseline NLR > 3 were independent risk factors for OS in patients with platinum-refractory advanced UC treated with pembrolizumab. Antibiotic exposure was not a predictor of ICI treatment response.
Collapse
Affiliation(s)
- Yasunori Akashi
- Department of Urology, Kindai University Nara Hospital, Ikoma 630-0293, Japan; (Y.A.)
| | - Yutaka Yamamoto
- Department of Urology, Kindai University Nara Hospital, Ikoma 630-0293, Japan; (Y.A.)
| | - Mamoru Hashimoto
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| | - Shogo Adomi
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| | - Keisuke Kiba
- Department of Urology, Kindai University Nara Hospital, Ikoma 630-0293, Japan; (Y.A.)
| | - Takafumi Minami
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| | - Kazuhiro Yoshimura
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| | - Akihide Hirayama
- Department of Urology, Kindai University Nara Hospital, Ikoma 630-0293, Japan; (Y.A.)
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Hospital, Osakasayama 589-8511, Japan
| |
Collapse
|
195
|
Chen B, Hong Y, Zhai X, Deng Y, Hu H, Tian S, Zhang Y, Ren X, Zhao J, Jiang C. m6A and m5C modification of GPX4 facilitates anticancer immunity via STING activation. Cell Death Dis 2023; 14:809. [PMID: 38065948 PMCID: PMC10709592 DOI: 10.1038/s41419-023-06241-w] [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: 07/11/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 12/18/2023]
Abstract
Cancer immunotherapy is arguably the most rapidly advancing realm of cancer treatment. Glutathione peroxidase 4 (GPX4) has emerged as the vital enzyme to prevent lipid peroxidation and maintain cellular redox homeostasis. However, the mechanism of GPX4 in the regulation of cancer immunotherapy of colon adenocarcinoma (COAD) are incompletely understood. In pan-cancer analysis, we found that GPX4 showed remarkably upregulated expression and exhibited significant association with overall survival in multiple cancer types, especially COAD. Furthermore, upregulated GPX4 expression was positively correlated with increased immune cells infiltration and enhanced expression of immunomodulators. Mechanistically, RBM15B- and IGFBP2-mediated N6-methyladenosine (m6A) modification and NSUN5-mediated 5-methylcytosine (m5C) modification of GPX4 facilitated anticancer immunity via activation of cyclic GMP-AMP synthase (cGAS)-stimulator of interferon (STING) signaling by maintaining redox homeostasis in COAD. The risk model and nomogram model constructed based on the GPX4-derived genes further confirmed the prognostic and treatment-guiding value of GPX4. In all, our study demonstrated that m6A and m5C modification of GPX4 may be a promising target for cancer immunotherapy via activating the cGAS-STING signaling pathway in COAD.
Collapse
Affiliation(s)
- Baoxiang Chen
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Yuntian Hong
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiang Zhai
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanrong Deng
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Heng Hu
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shunhua Tian
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yukang Zhang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xianghai Ren
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Jianhong Zhao
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Congqing Jiang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
196
|
Chen X, Hu G, Yu Q. Impact of disulfidptosis-associated clusters on breast cancer survival rates and guiding personalized treatment. Front Endocrinol (Lausanne) 2023; 14:1256132. [PMID: 38116315 PMCID: PMC10728640 DOI: 10.3389/fendo.2023.1256132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Background Breast cancer (BC) poses a serious threat to human health. Disulfidptosis is a recently discovered form of cell death associated with cancer prognosis and progression. However, the relationship between BC and disulfidptosis remains unclear. Methods We integrated single-cell sequencing and transcriptome sequencing in BC to assess the abundance and mutation status of disulfidptosis-associated genes (DAGs). Subsequently, we clustered the samples based on DAGs and constructed a prognostic model associated with disulfidptosis. Additionally, we performed pathway enrichment, immune response, and drug sensitivity analyses on the model. Finally, we validated the prognostic genes through Immunohistochemistry (IHC). Results The single-cell analysis identified 21 cell clusters and 8 cell types. By evaluating the abundance of DAGs in different cell types, we found specific expression of the disulfidoptosis core gene SLC7A11 in mesenchymal stem cells (MSCs). Through unsupervised clustering of DAGs, we identified two clusters. Utilizing differentially expressed genes from these clusters, we selected 7 genes (AFF4, SLC7A11, IGKC, IL6ST, LIMD2, MAT2B, and SCAND1) through Cox and Lasso regression to construct a prognostic model. External validation demonstrated good prognostic prediction of our model. BC patients were stratified into two groups based on riskscore, with the high-risk group corresponding to a worse prognosis. Immune response analysis revealed higher TMB and lower TIDE scores in the high-risk group, while the low-risk group exhibited higher CTLA4/PD-1 expression. This suggests that both groups may respond to immunotherapy, necessitating further research to elucidate potential mechanisms. Drug sensitivity analysis indicated that dasatinib, docetaxel, lapatinib, methotrexate, paclitaxel, and sunitinib may have better efficacy in the low-risk group. Finally, Immunohistochemistry (IHC) validated the expression of prognostic genes, demonstrating higher levels in tumor tissue compared to normal tissue. Conclusion Our study has developed an effective disulfidptosis-related prognostic prediction tool for BC and provides personalized guidance for the clinical management and immunotherapy selection of BC patients.
Collapse
Affiliation(s)
| | - Guohuang Hu
- Department of General Surgery, Affiliated Changsha Hospital of Hunan Normal University, Changsha, China
| | - Qianle Yu
- Department of General Surgery, Affiliated Changsha Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
197
|
Lu Y, Li Q, Xu L, Zheng Y, Wang Y, Liu Y, Zhang R, Liao L, Dong J. Thyroid dysfunction induced by anti-PD-1 therapy is associated with a better progression-free survival in patients with advanced carcinoma. J Cancer Res Clin Oncol 2023; 149:16501-16510. [PMID: 37715029 PMCID: PMC10645623 DOI: 10.1007/s00432-023-05364-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: 08/04/2023] [Accepted: 08/28/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE Thyroid dysfunction is the most common immune-related adverse event during anti-programmed cell death 1 (anti-PD-1) therapy. In this study, we monitored patients with advanced malignant tumors who received anti-PD-1 therapy to observe the characteristic of anti-PD-1 therapy-induced thyroid dysfunction and its correlation with prognosis. METHODS Patients with advanced carcinoma treated with anti-PD-1 therapy were evaluated for thyroid function at baseline and after treatment initiation from August 2020 to March 2022. Seventy-three patients were finally included in the study. RESULTS Among these patients, 19 (26.03%) developed thyroid dysfunction after receiving anti-PD-1 therapy. Primary hypothyroidism and thyrotoxicosis were the most common clinical manifestation. Anti-PD-1-induced thyroid dysfunction occurred 63 (26-131) days after administration; thyrotoxicosis appeared earlier than primary hypothyroidism. In Kaplan-Meier survival analysis, the progression-free survival (PFS) of the thyroid dysfunction group was better than that of the no thyroid dysfunction group (227 (95% confidence interval (CI) 50.85-403.15) days vs 164 (95% CI 77.76-250.24) days, p = 0.026). Male patients had better PFS than female patients (213 (95% CI 157.74-268.26) days vs 74 (95% CI 41.23-106.77) days, p = 0.031). In cox proportional hazards regression model, anti-PD-1-induced thyroid dysfunction remained an independent predictor of better PFS (hazard ratio (HR) = 0.339(0.136-0.848), p = 0.021). CONCLUSION Thyroid dysfunction is a common immune-related adverse events in advanced cancer patients treated with anti-PD-1 therapy and predicts a better prognosis. TRIAL REGISTRATION This study was retrospectively registered with Trial ClinicalTrials.gov (NCT05593744) on October 25, 2022.
Collapse
Affiliation(s)
- Yiran Lu
- Department of Endocrinology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Qingchen Li
- Department of Medical Oncology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Lusi Xu
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Shandong University, Ji-Nan, China
| | - Yanqing Zheng
- Department of Medical Oncology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Yanchao Wang
- Department of Medical Oncology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Ying Liu
- Department of Endocrinology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Rui Zhang
- Department of Endocrinology, Qilu Hospital, Shandong University, Ji-Nan, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Ji-Nan, China.
- Department of Endocrinology and Metabology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Institute of Nephrology, Ji-Nan, China.
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Shandong University, Ji-Nan, China.
| |
Collapse
|
198
|
Xu H, Jia Z, Liu F, Li J, Huang Y, Jiang Y, Pu P, Shang T, Tang P, Zhou Y, Yang Y, Su J, Liu J. Biomarkers and experimental models for cancer immunology investigation. MedComm (Beijing) 2023; 4:e437. [PMID: 38045830 PMCID: PMC10693314 DOI: 10.1002/mco2.437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 12/05/2023] Open
Abstract
The rapid advancement of tumor immunotherapies poses challenges for the tools used in cancer immunology research, highlighting the need for highly effective biomarkers and reproducible experimental models. Current immunotherapy biomarkers encompass surface protein markers such as PD-L1, genetic features such as microsatellite instability, tumor-infiltrating lymphocytes, and biomarkers in liquid biopsy such as circulating tumor DNAs. Experimental models, ranging from 3D in vitro cultures (spheroids, submerged models, air-liquid interface models, organ-on-a-chips) to advanced 3D bioprinting techniques, have emerged as valuable platforms for cancer immunology investigations and immunotherapy biomarker research. By preserving native immune components or coculturing with exogenous immune cells, these models replicate the tumor microenvironment in vitro. Animal models like syngeneic models, genetically engineered models, and patient-derived xenografts provide opportunities to study in vivo tumor-immune interactions. Humanized animal models further enable the simulation of the human-specific tumor microenvironment. Here, we provide a comprehensive overview of the advantages, limitations, and prospects of different biomarkers and experimental models, specifically focusing on the role of biomarkers in predicting immunotherapy outcomes and the ability of experimental models to replicate the tumor microenvironment. By integrating cutting-edge biomarkers and experimental models, this review serves as a valuable resource for accessing the forefront of cancer immunology investigation.
Collapse
Affiliation(s)
- Hengyi Xu
- State Key Laboratory of Molecular OncologyNational Cancer Center /National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ziqi Jia
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fengshuo Liu
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiayi Li
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yansong Huang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yiwen Jiang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pengming Pu
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tongxuan Shang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pengrui Tang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yongxin Zhou
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yufan Yang
- School of MedicineTsinghua UniversityBeijingChina
| | - Jianzhong Su
- Oujiang LaboratoryZhejiang Lab for Regenerative Medicine, Vision, and Brain HealthWenzhouZhejiangChina
| | - Jiaqi Liu
- State Key Laboratory of Molecular OncologyNational Cancer Center /National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
199
|
Ikeuchi Y, Someya M, Hasegawa T, Saito M, Mafune S, Tsuchiya T, Kitagawa M, Gocho T, Dehari H, Ogi K, Sasaki T, Hirohashi Y, Torigoe T, Hirokawa N, Miyazaki A, Sakata KI. Immunohistological evaluation of patients treated with intra-arterial chemoradiotherapy and surgery for oral cancer. Med Mol Morphol 2023; 56:288-296. [PMID: 37507576 DOI: 10.1007/s00795-023-00367-8] [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: 05/31/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Preoperative intra-arterial chemoradiotherapy (IACRT) can improve the outcome and reduce the extent of surgery in patients with advanced oral cancer. However, the response to this regimen varies among patients, which may be related to the immune status of the tumor. We investigated the effects of proteins involved in tumor immunity on the outcomes of combined IACRT and surgery for oral cancer. We examined CD8 + and FoxP3 + tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression on immune cells and tumor cells in pretreatment biopsy samples from 69 patients diagnosed with oral cancer treated with IACRT at our institution during 2000-2020. Patients with abundant CD8 + TILs had significantly better 5-year disease-specific survival (DSS) compared to that of patients with less infiltration of these cells (P = 0.016). Patients with higher FoxP3 + T-cells invasion had significantly better DSS compared to that of less FoxP3 (P = 0.005). Patients with high PD-L1 expression in tumor cells and immune cells had significantly better DSS than that of patients with low PD-L1 expression in these cells (P = 0.009 and P = 0.025, respectively). Collectively, these results suggest that the tumor immune microenvironment could affect outcomes of IACRT treatment in oral cancer.
Collapse
Affiliation(s)
- Yutaro Ikeuchi
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Masanori Someya
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan.
| | - Tomokazu Hasegawa
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Masato Saito
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Shoh Mafune
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Takaaki Tsuchiya
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Mio Kitagawa
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Toshio Gocho
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Hironari Dehari
- Department of Oral Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazuhiro Ogi
- Department of Oral Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takanori Sasaki
- Department of Oral Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naoki Hirokawa
- Department of Diagnostic Radiology, KKR Sapporo Medical Center, Sapporo, Japan
| | - Akihiro Miyazaki
- Department of Oral Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koh-Ichi Sakata
- Department of Radiology, Sapporo Medical University School of Medicine, S1W16, Chuo-Ku, Sapporo, Hokkaido, 060-8543, Japan
| |
Collapse
|
200
|
He W, Chen J, Zhou Y, Deng T, Feng Y, Luo X, Zhang C, Huang H, Liu J. Mitophagy genes in ovarian cancer: a comprehensive analysis for improved immunotherapy. Discov Oncol 2023; 14:221. [PMID: 38038814 PMCID: PMC10692064 DOI: 10.1007/s12672-023-00750-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Mitophagy is a process of selectively degrading damaged mitochondria, which has been found to be related to immunity, tumorigenesis, tumor progression, and metastasis. However, the role of mitophagy-related genes (MRGs) in the tumor microenvironment (TME) of ovarian cancer (OV) remains largely unexplored. METHODS We analyzed the expression, prognosis, and genetic alterations of 29 MRGs in 480 OV samples. Unsupervised clustering was used to classify OV into two subtypes (clusters A and B) based on MRG changes. We compared the clinical features, differential expressed genes (DEGs), pathways, and immune cell infiltration between the two clusters. We constructed a mitophagy scoring system (MRG_score) based on the DEGs and validated its ability to predict overall survival of OV patients. RESULTS We found that patients with high MRG_scores had better survival status and increased infiltration by immune cells. Further analysis showed that these patients may be more sensitive to immune checkpoint inhibitor (ICI) treatment. Additionally, the MRG_score significantly correlated with the sensitivity of chemotherapeutic drugs and targeted inhibitors. CONCLUSION Our comprehensive analysis of MRGs in the TME, clinical features, and patient prognosis revealed that the MRG_score is a potentially effective prognostic biomarker and predictor of treatment. This study provides new insights into the role of MRGs in OV and identifies patients who may benefit from ICI treatment, chemotherapy, or targeted treatment.
Collapse
Affiliation(s)
- Wenting He
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jieping Chen
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yun Zhou
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ting Deng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yanling Feng
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaolin Luo
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chuyao Zhang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - He Huang
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Jihong Liu
- Department of Gynecologic Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|