1
|
Zhang J, Zhan W, Hu H, Zhu H, Hao B, Wang S, Li Z, Zhang Z, Zhang T. Machine learning-based detoxification enzymes-related genes prognosis model in breast cancer: immune landscape and clinical significance. Discov Oncol 2025; 16:1178. [PMID: 40549224 PMCID: PMC12185839 DOI: 10.1007/s12672-025-02656-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 05/10/2025] [Indexed: 06/28/2025] Open
Abstract
BACKGROUND Breast cancer is one of the most common malignant tumors, threatening women's health and life globally. Despite significant treatment advances, its prognosis still faces great challenges. With the rapid development of molecular biology and genomics, the role of detoxification enzymes in breast cancer occurrence, development, and prognosis has gained increasing attention. This paper aims to establish a prognostic model based on detoxification enzymes-related genes to predict breast cancer patient survival. METHODS Unsupervised clustering was used to analyze breast cancer samples based on detoxification enzymes-related genes expression. Lasso cox regression analysis and univariate and multivariate Cox analysis were used to process the data, and machine learning algorithm was used to construct breast cancer prognosis model. The effect of detoxification enzymes-related genes on breast cancer was analyzed by single cell analysis. RESULTS The samples were classified into two subtypes, and a breast cancer prognosis model based on detoxification enzymes-related genes was constructed and validated using TCGA and GEO cohorts. Significant differences in pathways, immune infiltration, immunotherapy response, and drug sensitivity were observed between high- and low-risk groups. Single-cell analysis revealed that SQLE, a detoxification enzymes-related gene, was highly expressed in breast cancer epithelial cells (cancer cells), where SQLE + epithelial cells primarily influenced exhausted CD8 + T cells via the MIF signaling pathway. CONCLUSION In summary, the detoxification enzymes-related genes-based prognostic model developed in this study provides an effective tool for predicting breast cancer prognosis and offers new insights for diagnosis and treatment.
Collapse
Affiliation(s)
- Jingdi Zhang
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wendi Zhan
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Haihong Hu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongxia Zhu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Bo Hao
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Wang
- Department of Medical Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhuo Li
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhiming Zhang
- Health School of Nuclear Industry, 336 South Dongfeng Road, Hengyang, 421001, Hunan, China.
| | - Taolan Zhang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Research Center for Clinical Trial, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Li C, Tang Y, Zhang R, Shi L, Chen J, Zhang P, Zhang N, Li W. Inhibiting glycolysis facilitated checkpoint blockade therapy for triple-negative breast cancer. Discov Oncol 2025; 16:550. [PMID: 40244544 PMCID: PMC12006572 DOI: 10.1007/s12672-025-02320-w] [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: 11/23/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer cells are characterized by their altered energy metabolism. A hallmark of cancer metabolism is aerobic glycolysis, also called the Warburg effect. Hexokinase 2 (HK2), a crucial glycolytic enzyme converting glucose to glucose-6-phosphate, has been identified as a central player in the Warburg effect. Deletion of HK2 decreases cancer cell proliferation in animal models without explicit side effects, suggesting that targeting HK2 is a promising strategy for cancer therapy. In this study, we discovered a correlation between HK2 and the tumor immune response in triple-negative breast cancer. Inhibition of HK2 led to a reduction in G-CSF expression in 4T1 cells and a decrease in the development of myeloid-derived suppressor cells which, in turn, enhanced T cell immunity and prolonged the survival of 4T1 tumor-bearing mice. Furthermore, the HK2 inhibitor 3-BrPA improved the therapeutic efficacy of anti-PD-L1 therapy in 4T1 tumor-bearing mouse models. This study highlights the potential of glycolysis-targeting interventions as a novel treatment strategy, which can be combined with immunotherapy for the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Chong Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Tang
- Department of Gastrointestinal Surgery, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Ruizhi Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianying Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Zhang
- Department of Thyroid Breast Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Bao Q, Tang W, Tang W, Zhu X, Hong L, Zhang B. Identification of immune-related cervical cancer prognostic biomarkers and construction of prognostic model based on tumor microenvironment. Eur J Med Res 2025; 30:261. [PMID: 40205481 PMCID: PMC11984189 DOI: 10.1186/s40001-025-02515-5] [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: 08/18/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVE Tumor microenvironment (TME) and the expression of immune-related genes (IRGs) are closely related to the development of cervical cancer (CC). This study aims to explore some IRGs as prognostic biomarkers for CC patients based on TME. METHODS The abundance of tumor-infiltrating immune cells in CC samples was assessed using single-sample gene set enrichment analysis (ssGSEA). Thus, two immune-related groups are generated according to the immune status. The differentially expressed genes were discovered based on the grouping. Then, univariate Cox and LASSO regression analyses were performed using the R package glmnet. Five IRG prognostic signatures (HLA-DMA, DMBT1, CXCR6, CX3CL1, and SEMA3A) were established after that. The protein expression of some genes was verified by immunohistochemistry (IHC). RESULTS The signature of the five IRGs was identified to be an independent prognostic indicator for the overall survival in CC patients. A prognostic risk model was also constructed. CC patients were classified into high- and low-risk groups based on the median risk score. The survival time of patients in the high-risk group was shorter than that of those in the low-risk group. The five genes remarkably related to prognosis were screened, among which HLA-DMA, CXCR6, and CX3CL1 were the protective factors, whereas DMBT1 and SEMA3A were the risk factors. GO and KEGG enrichment analyses showed that the biomarkers of the five IRGs were enriched in the receptor-ligand interaction and chemokine signaling pathway. Moreover, CXCR6 expression was significantly correlated with immune cell infiltration among the five hub genes. IHC results demonstrated that the expression of SEMA3A protein level was increased, and CX3CL1 protein level was decreased in cervical cancer tissue. CONCLUSION Immune-related prognostic biomarkers in CC include HLA-DMA, DMBT1, CXCR6, CX3CL1, and SEMA3A. The risk score for the five genes is more accurate than that for other clinical risk factors in predicting prognosis at 3 and 5 years. The higher the risk score is, the worse the prognosis of CC patients is. Five prognostic biomarkers may participate in regulating TME through chemokine-mediated signaling pathways and receptor-ligand interactions. These findings provide new insights into the immunotherapy of CC.
Collapse
Affiliation(s)
- Qiufang Bao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China
- Department of Obstetrics and Gynecology, The First Clinical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fuzhou, Fujian, People's Republic of China
| | - Wenlu Tang
- Department of Anesthesiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, People's Republic of China
| | - Wenzhong Tang
- Department of Orthopaedics, Tingzhou Hospital of Fujian Province, Longyan, Fujian, People's Republic of China
| | - Xiaohong Zhu
- Department of Obstetrics and Gynecology, The First Clinical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Linliang Hong
- Department of Pediatrics, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| | - Bin Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
- Department of Obstetrics and Gynecology, The First Clinical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
4
|
Zhang Z, Liu B, Mei L, Chen R, Zhou H, Li Z. RREB1 could act as an immunological and prognostic biomarker: From comprehensive analysis to osteosarcoma validation. Int Immunopharmacol 2024; 143:113312. [PMID: 39405927 DOI: 10.1016/j.intimp.2024.113312] [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: 03/27/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND The Ras-responsive element binding protein 1 (RREB1) is a transcription factor involved in various biological processes. Notably, RREB1 plays a role in tumor immunity by regulating tumor-related gene expression, shaping the tumor microenvironment, and modulating immune checkpoints. Given these functions, RREB1 has emerged as a potential regulatory target in tumor immunotherapy. However, a comprehensive pan-cancer analysis evaluating RREB1's prognostic value and its role in modulating the immune microenvironment remains unexplored, warranting further investigation to better understand its mechanisms across different cancer types and its implications for personalized immunotherapy. METHODS We analyzed RREB1 expression across 33 cancer types using RNA sequencing data from the TCGA database. RREB1 alterations were further characterized using the cBioPortal database. Clinical and pathological features, along with prognostic significance, were assessed using TCGA clinical data. The involvement of RREB1 in the tumor microenvironment was evaluated using the CIBERSORT and ESTIMATE algorithms. Relationships between RREB1 expression and tumor mutation burden (TMB), as well as microsatellite instability (MSI), were investigated using Spearman's rank correlation coefficient. GSEA was applied to explore the biological functions of RREB1. Additionally, we assessed the link between RREB1 expression and the efficacy of PD-1/PD-L1 inhibitors. Finally, a series of in vitro experiments were performed to evaluate the impact of RREB1 expression on the malignant behavior of osteosarcoma (OS) and lung cancer cell lines. RESULTS RREB1 was overexpressed in several cancer types and correlated with patient prognosis. RREB1 expression was strongly associated with TMB, MSI, and immune cell infiltration, including regulatory T cells, CD8+ T cells, and macrophages. Furthermore, RREB1 expression was linked to immune responses and the efficacy of immunotherapy. In vitro experiments demonstrated that knockdown of RREB1 significantly inhibited the proliferation and migration of OS cells. CONCLUSIONS RREB1 shows potential as a prognostic marker for certain cancers and may predict the efficacy of immunotherapy. Additionally, RREB1 expression is related to immune-related markers, suggesting its role in prognosis and predicting responses to immune microenvironment therapies in specific tumors.
Collapse
Affiliation(s)
- Zhiming Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Binfeng Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lin Mei
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Ruiqi Chen
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Haoyang Zhou
- Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine of The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
5
|
Wu Y, Jiao J, Wu S, Jiang J. Strategies for the enhancement of IL-21 mediated antitumor activity in solid tumors. Cytokine 2024; 184:156787. [PMID: 39467483 DOI: 10.1016/j.cyto.2024.156787] [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: 07/25/2024] [Revised: 09/22/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Solid tumors significantly impact global health, necessitating enhanced prevention, early diagnosis, and treatment approaches. Tumor immunotherapy, notably through programmed cell death protein 1 (PD-1) and programmed cell death-ligand 1 (PD-L1), offers new hope to patients with advanced tumors, although many still do not benefit. Interleukin-21 (IL-21), a cytokine produced by certain immune cells, performs various biological functions by activating the JAK/STAT signaling pathway. Currently, recombinant IL-21 demonstrates promising antitumor activity and acceptable toxicity in several clinical trials. However, challenges such as side effects, off-target reactions, and a short half-life limit the effectiveness of cytokine-based immunotherapies. Therefore, researching enhanced IL-21 treatment strategies in solid tumors is crucial. Integrating IL-21 with various treatment modalities, including immune checkpoint inhibitors, additional cytokines, vaccines, or radiotherapy, is essential for improving response rates and prolonging patient survival. This review explores the specific mechanisms of IL-21 in prevalent high-incidence tumors, examines improved strategies for IL-21 in solid tumors, and aims to provide a theoretical basis for developing targeted treatment strategies.
Collapse
Affiliation(s)
- You Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Jing Jiao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| |
Collapse
|
6
|
Hang Y, Huang J, Ding M, Shen Y, Zhou Y, Cai W. Extracellular vesicles reshape the tumor microenvironment to improve cancer immunotherapy: Current knowledge and future prospects. Int Immunopharmacol 2024; 140:112820. [PMID: 39096874 DOI: 10.1016/j.intimp.2024.112820] [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: 06/03/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Tumor immunotherapy has revolutionized cancer treatment, but limitations remain, including low response rates and immune complications. Extracellular vesicles (EVs) are emerging as a new class of therapeutic agents for various diseases. Recent research shows that changes in the amount and composition of EVs can reshape the tumor microenvironment (TME), potentially improving the effectiveness of immunotherapy. This exciting discovery has sparked clinical interest in using EVs to enhance the immune system's response to cancer. In this Review, we delve into the world of EVs, exploring their origins, how they're generated, and their complex interactions within the TME. We also discuss the crucial role EVs play in reshaping the TME during tumor development. Specifically, we examine how their cargo, including molecules like PD-1 and non-coding RNA, influences the behavior of key immune cells within the TME. Additionally, we explore the current applications of EVs in various cancer therapies, the latest advancements in engineering EVs for improved immunotherapy, and the challenges faced in translating this research into clinical practice. By gaining a deeper understanding of how EVs impact the TME, we can potentially uncover new therapeutic vulnerabilities and significantly enhance the effectiveness of existing cancer immunotherapies.
Collapse
Affiliation(s)
- Yu Hang
- Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - JingYi Huang
- Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingming Ding
- Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanhua Shen
- Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - YaoZhong Zhou
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China.
| | - Wan Cai
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
7
|
Kang W, Cheng J, Pan L, Zhan P, Liu H, Lv T, Han H, Song Y. Heterogeneity between subgroups of first-line chemoimmunotherapy for extensive-stage small cell lung cancer patients: a meta-analysis and systematic review. Front Oncol 2024; 14:1334957. [PMID: 39493446 PMCID: PMC11527596 DOI: 10.3389/fonc.2024.1334957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Objectives Differences in clinicopathological characteristics of extensive-stage small cell lung cancer (ES-SCLC) patients may influence the immune response. This study aims to evaluate the heterogeneity of response to first-line chemoimmunotherapy between subgroups in ES-SCLC to screen out suitable populations. Materials and methods We searched the PubMed, EMBASE, and Cochrane Library databases from inception to December 3, 2022 for randomized controlled trials (RCTs) of ES-SCLC chemoimmunotherapy. We also reviewed main conferences from January 1, 2021 to October 1, 2023. A trial-specific hazard ratio (HR) ratio for each subgroup was calculated, and these ratios were then pooled using the deft approach. Results A total of 9 RCTs with 4099 patients were finally included. The pooled ratios were 0.92 (95% CI = 0.77 to 1.09) for OS-HRs and 0.79 (95% CI = 0.55 to 1.13) for PFS-HRs in women versus men. The pooled ratios of OS-HRs and PFS-HRs in patients with positive versus negative PD-L1 expression were 1.26 (95% CI = 0.91 to 1.73) and 1.08 (95% CI = 0.77 to 1.52), respectively. The pooled ratios of OS-HRs and PFS-HRs in patients without versus with brain metastasis were 0.77 (95% CI = 0.59 to 1.01) and 0.71 (95% CI = 0.44 to 1.12). No statistically significant differences were also found in terms of subgroups for age, liver metastasis, smoking status, ECOG PS, LDH level, type of platinum salt and race. Conclusion Women or patients with negative PD-L1 expression or with LDH ≤ ULN or without brain metastasis tend to benefit more from first-line chemoimmunotherapy in ES-SCLC. More trials are needed to prospectively validate the therapeutic heterogeneity among clinicopathological characteristics. Systematic review registration https://inplasy.com/inplasy-2023-3-0064/ identifier, INPLASY202330064.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hedong Han
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing
University, Nanjing, China
| | - Yong Song
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing
University, Nanjing, China
| |
Collapse
|
8
|
Matsueda S, Chen L, Li H, Yao H, Yu F. Recent clinical researches and technological development in TIL therapy. Cancer Immunol Immunother 2024; 73:232. [PMID: 39264449 PMCID: PMC11393248 DOI: 10.1007/s00262-024-03793-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/29/2024] [Indexed: 09/13/2024]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy represents a groundbreaking advancement in the solid cancer treatment, offering new hope to patients and their families with high response rates and long overall survival. TIL therapy involves extracting immune cells from a patient's tumor tissue, expanding them ex vivo, and infusing them back into the patient to target and eliminate cancer cells. This revolutionary approach harnesses the power of the immune system to combat cancers, ushering in a new era of T cell-based therapies along with CAR-T and TCR-therapies. In this comprehensive review, we aim to elucidate the remarkable potential of TIL therapy by delving into recent advancements in basic and clinical researches. We highlight on the evolving landscape of TIL therapy as a prominent immunotherapeutic strategy, its multifaceted applications, and the promising outcomes. Additionally, we explore the future horizons of TIL therapy, next-generation TILs, and combination therapy, to overcome the limitations and improve clinical efficacy of TIL therapy.
Collapse
Affiliation(s)
- Satoko Matsueda
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA.
| | - Lei Chen
- Department of Neurosurgery, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Hongmei Li
- Department of Oncology, Qingdao University Medical School, Qinddao, 266003, China
| | - Hui Yao
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA
| | - Fuli Yu
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA
| |
Collapse
|
9
|
Liu L, Chen J, Ye F, Chu F, Rao C, Wang Y, Yan Y, Wu J. Prognostic value of oxidative phosphorylation-related genes in hepatocellular carcinoma. Discov Oncol 2024; 15:258. [PMID: 38960931 PMCID: PMC11222354 DOI: 10.1007/s12672-024-01129-3] [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: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is the most prevalent malignancies worldwide. Recently, oxidative phosphorylation (OXPHOS) has received extensive concern as an emerging target in antitumor therapy. However, the OXPHOS-involved underlying genes and clinical utilization in HCC remain worth exploring. The present research aimed to create an OXPHOS-relevant signature in HCC. PATIENTS AND METHODS In this study, the prognostic signature genes linked with OXPHOS were identified, and prognostic models were built using least absolute shrinkage and selection operator (LASSO) cox regression analysis. Furthermore, the combination study of immune microenvironment and signature genes looked into the involvement of immune cells in signature-based genes in HCC. Following that, chemotherapeutic drug sensitivity and immunotherapy analysis was implemented to predict clinical efficacy in HCC patients. Finally, clinical samples were collected to measure the expression of OXPHOS-related signature genes. RESULTS Following a series of screens, six prognostic signature genes related with OXPHOS were identified: MRPS23, MPV17, MAPK3, IGF2BP2, CDK5, and IDH2, on which a risk model was built. The findings revealed a significant drop in the survival rate of HCC patients as their risk score increased. Meanwhile, independent prognostic study demonstrated that the risk score could accurately identify HCC patients. Immuno-microenvironmental correlation research suggested that the prognostic characteristics could serve as a reference index for both immunotherapy and chemotherapy. Finally, RT-qPCR exhibited a trend in signature gene expression that was consistent with the results. CONCLUSION In this study, a total of six prognostic genes associated with OXPHOS were selected and a prognostic model was constructed, providing an essential reference for the study of OXPHOS in HCC.
Collapse
Affiliation(s)
- Luzheng Liu
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Jiacheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Fei Ye
- Department of Blood Cell Therapy, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Fengran Chu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China
| | - Chaoluan Rao
- Department of Nursing, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Yong Wang
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China
| | - Yanggang Yan
- Department of Interventional Radiology and Vascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570100, Hainan Province, China.
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
10
|
Shen J, Su X, Wang Q, Ke Y, Zheng T, Mao Y, Wang Z, Dong J, Duan S. Current and future perspectives on the regulation and functions of miR-545 in cancer development. CANCER PATHOGENESIS AND THERAPY 2024; 2:142-154. [PMID: 39027151 PMCID: PMC11252520 DOI: 10.1016/j.cpt.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 07/20/2024]
Abstract
Micro ribonucleic acids (miRNAs) are a highly conserved class of single-stranded non-coding RNAs. Within the miR-545/374a cluster, miR-545 resides in the intron of the long non-coding RNA (lncRNA) FTX on Xq13.2. The precursor form, pre-miR-545, is cleaved to generate two mature miRNAs, miR-545-3p and miR-545-5p. Remarkably, these two miRNAs exhibit distinct aberrant expression patterns in different cancers; however, their expression in colorectal cancer remains controversial. Notably, miR-545-3p is affected by 15 circular RNAs (circRNAs) and 10 long non-coding RNAs (lncRNAs), and it targets 27 protein-coding genes (PCGs) that participate in the regulation of four signaling pathways. In contrast, miR-545-5p is regulated by one circRNA and five lncRNAs, it targets six PCGs and contributes to the regulation of one signaling pathway. Both miR-545-3p and miR-545-5p affect crucial cellular behaviors, including cell cycle, proliferation, apoptosis, epithelial-mesenchymal transition, invasion, and migration. Although low miR-545-3p expression is associated with poor prognosis in three cancer types, studies on miR-545-5p are yet to be reported. miR-545-3p operates within a diverse range of regulatory networks, thereby augmenting the efficacy of cancer chemotherapy, radiotherapy, and immunotherapy. Conversely, miR-545-5p enhances immunotherapy efficacy by inhibiting T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) expression. In summary, miR-545 holds immense potential as a cancer biomarker and therapeutic target. The aberrant expression and regulatory mechanisms of miR-545 in cancer warrant further investigation.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Xinming Su
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Qurui Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yufei Ke
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Tianyu Zheng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yunan Mao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Jingyin Dong
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| |
Collapse
|
11
|
Tovar Perez JE, Zhang S, Hodgeman W, Kapoor S, Rajendran P, Kobayashi KS, Dashwood RH. Epigenetic regulation of major histocompatibility complexes in gastrointestinal malignancies and the potential for clinical interception. Clin Epigenetics 2024; 16:83. [PMID: 38915093 PMCID: PMC11197381 DOI: 10.1186/s13148-024-01698-8] [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: 11/10/2023] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Gastrointestinal malignancies encompass a diverse group of cancers that pose significant challenges to global health. The major histocompatibility complex (MHC) plays a pivotal role in immune surveillance, orchestrating the recognition and elimination of tumor cells by the immune system. However, the intricate regulation of MHC gene expression is susceptible to dynamic epigenetic modification, which can influence functionality and pathological outcomes. MAIN BODY By understanding the epigenetic alterations that drive MHC downregulation, insights are gained into the molecular mechanisms underlying immune escape, tumor progression, and immunotherapy resistance. This systematic review examines the current literature on epigenetic mechanisms that contribute to MHC deregulation in esophageal, gastric, pancreatic, hepatic and colorectal malignancies. Potential clinical implications are discussed of targeting aberrant epigenetic modifications to restore MHC expression and 0 the effectiveness of immunotherapeutic interventions. CONCLUSION The integration of epigenetic-targeted therapies with immunotherapies holds great potential for improving clinical outcomes in patients with gastrointestinal malignancies and represents a compelling avenue for future research and therapeutic development.
Collapse
Affiliation(s)
| | - Shilan Zhang
- Department of Cardiovascular Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200070, China
| | - William Hodgeman
- Wolfson Medical School, The University of Glasgow, Glasgow, G12 8QQ, UK
| | - Sabeeta Kapoor
- Center for Epigenetics and Disease Prevention, Texas A&M Health, Houston, TX, 77030, USA
| | - Praveen Rajendran
- Center for Epigenetics and Disease Prevention, Texas A&M Health, Houston, TX, 77030, USA
- Department of Translational Medical Sciences, and Antibody & Biopharmaceuticals Core, Texas A&M Medicine, Houston, TX, 77030, USA
| | - Koichi S Kobayashi
- Department of Immunology, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
- Hokkaido University Institute for Vaccine Research and Development, Sapporo, 060-8638, Japan
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, Bryan, TX, 77087, USA
| | - Roderick H Dashwood
- Center for Epigenetics and Disease Prevention, Texas A&M Health, Houston, TX, 77030, USA.
- Department of Translational Medical Sciences, and Antibody & Biopharmaceuticals Core, Texas A&M Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Dong X, Shao C, Xu S, Tu J, Xu W, Chen D, Tang Y. Construction and validation of a prognostic signature based on anoikis-related lncRNAs in lung adenocarcinoma. Aging (Albany NY) 2024; 16:9899-9917. [PMID: 38850527 PMCID: PMC11210241 DOI: 10.18632/aging.205905] [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: 08/11/2023] [Accepted: 05/02/2024] [Indexed: 06/10/2024]
Abstract
Lung adenocarcinoma (LUAD) is the most common type of lung cancer and is characterized by a high death rate and a poor prospect for survival. Anoikis, which is a kind of programmed cell apoptosis, is an important factor in the advancement of tumors. Nonetheless, the function of anoikis-related lncRNAs (ARLRs) in LUAD is still not well understood. The TCGA database was queried for genomic and clinical information. A prognostic signature for ARLRs was established via the use of coexpression analysis and Cox regression. Validation of the model's accuracy was conducted utilizing K-M curves and receiver operating characteristic (ROC) curves, and the signature was utilized to develop a nomogram. LncRNAs were implicated in the progression of tumors, as determined by functional enrichment analysis. There was an improvement in prognosis, increased immune cell infiltration, and higher immune scores among the low-risk patients. Additionally, we found that the two groups had varied anticancer drug sensitivities, which could help guide treatment. The impact of one ARLR, AC026355.2, on migration and invasion was validated by in vitro experiments in LUAD cells. Herein, a new lncRNA signature associated with anoikis was identified and estimated, potentially serving as a prognostic indicator for LUAD patients.
Collapse
Affiliation(s)
- Xiaoqi Dong
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Chuan Shao
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Shuguang Xu
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Jinjing Tu
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Wenjing Xu
- Ningbo University Health Science Center, Ningbo, China
| | - Dahua Chen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Yaodong Tang
- Department of Pulmonary and Critical Care Medicine, Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| |
Collapse
|
13
|
Liu Z, Han S, Luo Y, Zhao Z, Ni L, Chai L, Tang H. PERP May Affect the Prognosis of Lung Adenocarcinoma by Inhibiting Apoptosis. Cancer Manag Res 2024; 16:199-214. [PMID: 38525370 PMCID: PMC10961073 DOI: 10.2147/cmar.s443490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Background PERP, a member of the peripheral myelin protein gene family, is a new therapeutic target in cancer. The relationships between PERP and immune cell infiltration in lung cancer have not been studied. Therefore, the role of PERP in the tumour microenvironment (TME) of lung cancer needs to be further explored. Methods In this study, we explored the association between PERP expression and clinical characteristics by analysing data from the TCGA database. Cox regression and Kaplan‒Meier methods were used to investigate the relationship between the expression of PERP and overall survival in patients with lung adenocarcinoma (LUAD). The relationship between PERP expression and the degree of infiltration of specific immune cell subsets in LUAD was evaluated using the TIMER database and GEPIA. We also performed GO enrichment analysis and KEGG enrichment analysis to reveal genes coexpressed with PERP using the Coexpedia database. Finally, we verified the expression and function of PERP in LUAD tissues and the A549 cell line by RT‒PCR, Western blot, CCK-8, IHC, and wound healing assays. The mouse model was used to study the in vivo effects of PERP. Results According to our results, PERP expression was significantly higher in LUAD tissues and associated with the clinical characteristics of the disease. Survival was independently associated with PERP in LUAD patients. We further verified that PERP might regulate B-cell infiltration in LUAD to affect the prognosis of LUAD. To identify PERP-related signalling pathways in LUAD, we performed a genome-aggregation analysis (GSEA) between low and high PERP expression datasets. LUAD cells express higher levels of PERP than paracarcinoma cells, and PERP inhibits the proliferation and metastasis of A549 cells through apoptosis. Conclusion PERP may affect the prognosis of lung adenocarcinoma by inhibiting apoptosis and is associated with immune cell infiltration.
Collapse
Affiliation(s)
- Zhongxiang Liu
- Department of Pulmonary and Critical Care Medicine, the Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, the First Affiliated Hospital of Jiangsu Vocational College of Medicine, Yancheng, 224000, People’s Republic of China
| | - Shuhua Han
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Yuhong Luo
- College of Life Science and Technology, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Zhangyan Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, People’s Republic of China
| | - Lingyu Ni
- China School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210046, People’s Republic of China
| | - Linlin Chai
- Department of Pathology, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, The First Affiliated Hospital of Jiangsu Vocational College of Medicine, Yancheng, 224000, People’s Republic of China
| | - Haicheng Tang
- Department of Respiratory and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, People’s Republic of China
| |
Collapse
|
14
|
Bao X, Li Q, Chen D, Dai X, Liu C, Tian W, Zhang H, Jin Y, Wang Y, Cheng J, Lai C, Ye C, Xin S, Li X, Su G, Ding Y, Xiong Y, Xie J, Tano V, Wang Y, Fu W, Deng S, Fang W, Sheng J, Ruan J, Zhao P. A multiomics analysis-assisted deep learning model identifies a macrophage-oriented module as a potential therapeutic target in colorectal cancer. Cell Rep Med 2024; 5:101399. [PMID: 38307032 PMCID: PMC10897549 DOI: 10.1016/j.xcrm.2024.101399] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 02/04/2024]
Abstract
Colorectal cancer (CRC) is a common malignancy involving multiple cellular components. The CRC tumor microenvironment (TME) has been characterized well at single-cell resolution. However, a spatial interaction map of the CRC TME is still elusive. Here, we integrate multiomics analyses and establish a spatial interaction map to improve the prognosis, prediction, and therapeutic development for CRC. We construct a CRC immune module (CCIM) that comprises FOLR2+ macrophages, exhausted CD8+ T cells, tolerant CD8+ T cells, exhausted CD4+ T cells, and regulatory T cells. Multiplex immunohistochemistry is performed to depict the CCIM. Based on this, we utilize advanced deep learning technology to establish a spatial interaction map and predict chemotherapy response. CCIM-Net is constructed, which demonstrates good predictive performance for chemotherapy response in both the training and testing cohorts. Lastly, targeting FOLR2+ macrophage therapeutics is used to disrupt the immunosuppressive CCIM and enhance the chemotherapy response in vivo.
Collapse
Affiliation(s)
- Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| | - Qiong Li
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Weihong Tian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yin Wang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jinlin Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chunyu Lai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Chanqi Ye
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Shan Xin
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xin Li
- Department of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ge Su
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yongfeng Ding
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Yangyang Xiong
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jindong Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Vincent Tano
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637551, Republic of Singapore
| | - Yanfang Wang
- Ludwig-Maximilians-Universität München (LMU), 80539 Munich, Germany
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Shuiguang Deng
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jianpeng Sheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| |
Collapse
|
15
|
Tian J, Fu W, Xie Z, Zhao Y, Yang H, Zhao J. Methionine enkephalin (MENK) protected macrophages from ferroptosis by downregulating HMOX1 and ferritin. Proteome Sci 2024; 22:2. [PMID: 38245706 PMCID: PMC10799539 DOI: 10.1186/s12953-024-00228-x] [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: 05/02/2023] [Accepted: 01/06/2024] [Indexed: 01/22/2024] Open
Abstract
OBJECTIVE The aim of this work was to investigate the immunological effect of MENK by analyzing the protein spectrum and bioinformatics of macrophage RAW264.7, and to explore the relationship between macrophage and ferroptosis. RESULT We employed proteomic analysis to identify differentially expressed proteins (DEPs) between macrophages and macrophages intervened by MENK. A total of 208 DEPs were identified. Among these, 96 proteins had upregulated expression and 112 proteins had downregulated expression. Proteomic analysis revealed a significant enrichment of DEPs associated with iron metabolism. The identification of hub genes was conducted using KEGG pathway diagrams and protein-protein interaction (PPI) analysis. The hub genes identified in this study include HMOX1 and Ferritin (FTH and FTL). A correlation was established between HMOX1, FTH, and FTL in the GO and KEGG databases. The results of PCR, WB and immunofluorescence showed that MENK downregulated the level of HMOX1 and FTH. CONCLUSION MENK had the potential to become an adjuvant chemotherapy drug by regulating iron metabolism in macrophages, reducing levels of HMOX1 and ferritin. We proposed an innovative research direction on the therapeutic potential of MENK, focusing on the relationship between ferroptosis and macrophage activity.
Collapse
Affiliation(s)
- Jing Tian
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China.
| | - Wenrui Fu
- Graduate College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Zifeng Xie
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Yuanlong Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Haochen Yang
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| | - Jiafan Zhao
- First Clinical Medical College, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China
| |
Collapse
|
16
|
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: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
17
|
Zhang Y, Guo J, Zhang L, Li Y, Sheng K, Zhang Y, Liu L, Gong W, Guo K. CircASPH Enhances Exosomal STING to Facilitate M2 Macrophage Polarization in Colorectal Cancer. Inflamm Bowel Dis 2023; 29:1941-1956. [PMID: 37624989 DOI: 10.1093/ibd/izad113] [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: 09/01/2022] [Indexed: 08/27/2023]
Abstract
Exosomes are considered a mediator of communication within the tumor microenvironment (TME), which modulates cancer progression through transmitting cargos between cancer cells and other cancer-related cells in TME. Circular RNAs (circRNAs) have emerged to be regulators in colorectal cancer (CRC) progression, but most of them have not been discussed in CRC. This study aims to investigate the role of circRNA aspartate beta-hydroxylase (circASPH) in CRC progression and its correlation with exosome-mediated TME. At first, we determined that circASPH was upregulated in CRC samples and cell lines. Functionally, the circASPH deficiency suppressed the malignant processes of CRC cells and also inhibited in vivo tumor growth via enhancing antitumor immunity. Mechanically, circASPH facilitated macrophage M2 polarization by upregulating exosomal stimulator of interferon genes (STING). CircASPH interacted with insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) to stabilize IGF2BP2 protein, therefore enhancing the stability of m6A-modified STING mRNA. In turn, coculture of STING-overexpressed macrophages recovered the suppression of silenced circASPH on the malignancy of CRC cells both in vitro and in vivo. Our study demonstrated that circASPH enhances exosomal STING to facilitate M2 macrophage polarization, which further accelerates CRC progression. The findings support circASPH as a promising therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Yuting Zhang
- Department of Gastroenterology, the First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jiakun Guo
- College of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Liyin Zhang
- Department of Dermatology, Wuxi's Second People Hospital Affiliated With Nanjing Medical University, 68 Zhongshan Road, Wuxi, Jiangsu 214000, China
| | - Ying Li
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230001, Anhui, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, 230001, Anhui, China
| | - Yawei Zhang
- Department of General Surgery, the First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Liu Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wenbin Gong
- Department of General Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Kun Guo
- Department of General Surgery, the First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| |
Collapse
|
18
|
Cao Y, Wo M, Xu C, Fei X, Jin J, Shan Z. An AMPK agonist suppresses the progress of colorectal cancer by regulating the polarization of TAM to M1 through inhibition of HIF-1α and mTOR signal pathway. J Cancer Res Ther 2023; 19:1560-1567. [PMID: 38156922 DOI: 10.4103/jcrt.jcrt_2670_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/17/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE This study aimed to evaluate the impact of an adenosine monophosphate-activated protein kinase (AMPK) agonist, metformin (MET), on the antitumor effects of macrophages and to determine the underlying mechanism involved in the process. MATERIALS AND METHODS M0 macrophages were derived from phorbol-12-myristate-13-acetate-stimulated THP-1 cells. RESULTS The levels of tumor necrosis factor-alpha (TNF-α) and human leukocyte antigen-DR (HLA-DR) were decreased in macrophages incubated with HCT116 cells, whereas those of arginase-1 (Arg-1), CD163, and CD206 were elevated; these effects were reversed by MET. The transfection of small interfering (si) RNA abrogated the influence of MET on the expression of the M1/M2 macrophage biomarkers. MET significantly suppressed the proliferation and migration abilities of HCT116 cells incubated with M0 macrophages; these actions were reversed by siRNA transfection against AMPK. The hypoxia-inducible factor 1-alpha (HIF-1α), phosphorylated protein kinase B (p-AKT), and phosphorylated mammalian target of rapamycin (p-mTOR) levels were reduced by the introduction of MET and promoted by siRNA transfection against AMPK. In addition, the levels of HIF-1α, p-AKT, and p-mTOR suppressed by MET were markedly increased following the transfection of siRNA against AMPK. CONCLUSION These findings indicate that MET can repress the progression of colorectal cancer by transforming tumor-associated macrophages to the M1phenotype via inhibition of the HIF-1α and mTOR signaling pathways.
Collapse
Affiliation(s)
- Yuanyuan Cao
- Department of Laboratory Medicine, Hangzhou Cancer Hospital, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Shangcheng, China
| | - Mingyi Wo
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Chan Xu
- Department of Laboratory Medicine, Affiliated Third Hospital of Zhejiang Traditional Chinese Medicine University, Xihu, Hangzhou, Zhejiang, China
| | - Xianming Fei
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Juan Jin
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Zhiming Shan
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| |
Collapse
|
19
|
Zuo B, Wang L, Li X, Li X, Wang J, Xiong Y, Lei J, Zhang X, Chen Y, Liu Q, Jiao J, Sui M, Fan J, Wu N, Song Z, Li G. Abnormal low expression of SFTPC promotes the proliferation of lung adenocarcinoma by enhancing PI3K/AKT/mTOR signaling transduction. Aging (Albany NY) 2023; 15:12451-12475. [PMID: 37955668 PMCID: PMC10683597 DOI: 10.18632/aging.205191] [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: 06/23/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023]
Abstract
The abnormality of surfactant protein C (SFTPC) has been linked to the development of a number of interstitial lung diseases, according to mounting evidence. Nonetheless, the function and mechanism of SFTPC in the biological progression of lung adenocarcinoma (LUAD) remain unclear. Analysis of public datasets and testing of clinical samples suggested that SFTPC expression was abnormally low in LUAD, which was associated with the onset and poor prognosis of LUAD. The SFTPC-related risk score was derived using least absolute shrinkage and selection operator Cox regression as well as multivariate Cox regression. The risk score was highly correlated with tumor purity and tumor mutation burden, and it could serve as an independent prognostic indicator for LUAD. Low-risk LUAD patients may benefit more from CTLA-4 or/and PD-1 inhibitors. Overall, the risk score is useful for LUAD patient prognostication and treatment guidance. Moreover, in vitro and in vivo experiments demonstrated that SFTPC inhibits the proliferation of LUAD by inhibiting PI3K/AKT/mTOR signaling transduction. These results reveal the molecular mechanism by which SFTPC inhibits the proliferation of LUAD and suggest that SFTPC could be a new therapeutic target for LUAD.
Collapse
Affiliation(s)
- Baile Zuo
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Lin Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi’an, Shaanxi, China
| | - Xiaoyan Li
- Department of Blood Transfusion, Shanxi Provincial People’s Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xin Li
- Department of Geriatric Medicine, Shanxi Provincial People’s Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinping Wang
- Department of Ultrasound, Shanxi Provincial People’s Hospital, Affiliate of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xi Zhang
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yifan Chen
- College of Management, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Qiongwen Liu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Jinke Jiao
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Mengru Sui
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Jinhan Fan
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Ningxue Wu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Zewen Song
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Guoyin Li
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi’an, Shaanxi, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Yang X, Luo Y, Li M, Jin Z, Chen G, Gan C. Long non-coding RNA NBR2 suppresses the progression of colorectal cancer by downregulating miR-19a to regulate M2 macrophage polarization. CHINESE J PHYSIOL 2023; 66:546-557. [PMID: 38149567 DOI: 10.4103/cjop.cjop-d-23-00064] [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: 12/28/2023] Open
Abstract
Colorectal cancer (CRC) is a malignant tumor of the gastrointestinal tract that significantly impacts the health of patients and lacks promising methods of diagnosis. Tumor-associated macrophages (TAMs) are involved in CRC progression, and their function is regulated by long non-coding RNAs (lncRNAs). The lncRNA NBR2 was recently reported as an oncogene, whose function in CRC remains uncertain. The present study aimed to investigate the biological function of lncRNA NBR2 in the progression of CRC and its underlying molecular mechanisms. Ten pairs of clinical CRC and para-carcinoma tissues were collected to determine the expression levels of lncRNA NBR2 and miR-19a, and the polarization state of TAMs. Quantitative reverse transcriptase-polymerase chain reaction was used to evaluate the expression of miR-19a, and western blotting was used to determine the expression levels of tumor necrosis factor-α, human leukocyte antigen-DR, arginase-1, CD163, CD206, interleukin-4, AMP-activated protein kinase (AMPK), p-AMPK, hypoxia-inducible factor-1α (HIF-1α), protein kinase B (AKT), p-AKT, mechanistic target of rapamycin (mTOR), and p-mTOR in TAMs. The proliferative ability of HCT-116 cells was detected using the CCK8 assay, and the migratory ability of HCT-116 cells was evaluated using the Transwell assay. The interaction between lncRNA NBR2 and miR-19a was determined using the luciferase assay. The lncRNA NBR2 was downregulated and miR-19a was highly expressed in CRC cells, accompanied by a high M2 polarization. Downregulated miR-19a promoted M1 polarization, activated AMPK, suppressed HIF-1α and AKT/mTOR signaling pathways, and promoted antitumor properties in NBR2-overexpressed TAMs, which were all reversed by the introduction of the miR-19a mimic. LncRNA NBR2 was verified to target miR-19a in macrophages according to the results of the luciferase assay. Collectively, lncRNA NBR2 may suppress the progression of CRC by downregulating miR-19a to regulate M2 macrophage polarization.
Collapse
Affiliation(s)
- Xiaoting Yang
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Ye Luo
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Mengying Li
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Zhan Jin
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Gao Chen
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Chunchun Gan
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| |
Collapse
|
21
|
Huang J, Zhang J, Zhang F, Lu S, Guo S, Shi R, Zhai Y, Gao Y, Tao X, Jin Z, You L, Wu J. Identification of a disulfidptosis-related genes signature for prognostic implication in lung adenocarcinoma. Comput Biol Med 2023; 165:107402. [PMID: 37657358 DOI: 10.1016/j.compbiomed.2023.107402] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most prevalent subtype of non-small cell lung cancer. Additionally, disulfidptosis, a newly discovered type of cell death, has been found to be closely associated with the onset and progression of tumors. METHODS The study first identified genes related to disulfidptosis through correlation analysis. These genes were then screened using univariate cox regression and LASSO regression, and a prognostic model was constructed through multivariate cox regression. A nomogram was also created to predict the prognosis of LUAD. The model was validated in three independent data sets: GSE72094, GSE31210, and GSE37745. Next, patients were grouped based on their median risk score, and differentially expressed genes between the two groups were analyzed. Enrichment analysis, immune infiltration analysis, and drug sensitivity evaluation were also conducted. RESULTS In this study, we examined 21 genes related to disulfidptosis and developed a gene signature that was found to be associated with a poorer prognosis in LUAD. Our model was validated using three independent datasets and showed AUC values greater than 0.5 at 1, 3, and 5 years. Enrichment analysis revealed that the disulfidptosis-related genes signature had a multifaceted impact on LUAD, particularly in relation to tumor development, proliferation, and metastasis. Patients in the high-risk group exhibited higher tumor purity and lower stromal score, ESTIMATE score, and Immune score. CONCLUSION This study constructed a gene signature related to disulfidptosis in lung adenocarcinoma and analyzed its impact on the disease and its association with the tumor microenvironment. The findings of this research provide valuable insights into the understanding of lung adenocarcinoma and could potentially lead to the development of new treatment strategies.
Collapse
Affiliation(s)
- Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rui Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yiyan Zhai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yifei Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoyu Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengsen Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Leiming You
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
22
|
Wei Z, Zhou C, Shen Y, Deng H, Shen Z. Identification of a new anoikis-related gene signature for prognostic significance in head and neck squamous carcinomas. Medicine (Baltimore) 2023; 102:e34790. [PMID: 37682196 PMCID: PMC10489427 DOI: 10.1097/md.0000000000034790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
Anoikis, a mode of programmed cell death, is essential for normal development and homeostasis in the organism and plays an important role in the onset and progression of cancers. The authors of this research sought to establish a gene signature associated with anoikis to predict therapy outcomes and patient prognosis for individuals with head and neck squamous cell carcinoma (HNSCC). Transcriptome data of anoikis-related genes (ARGs) in individuals with HNSCC were retrieved from public databases to aid in the formulation of the gene signature. A novel ARG signature was then created using a combination of the Least Absolute Shrinkage and Selection Operator regression and Cox regression analysis. The relationship between ARGs and tumor immune microenvironment in HNSCC was explored using single-cell analysis. HNSCC individuals were classified into high-risk and low-risk groups as per the median value of risk score. The study also investigated the variations in the infiltration status of immune cells, tumor microenvironment, sensitivity to immunotherapy and chemotherapeutics, as well as functional enrichment between the low-risk and high-risk categories. A total of 18 ARGs were incorporated in the formulation of the signature. Our signature's validity as a standalone predictive predictor was validated by multivariate Cox regression analysis and Kaplan-Meier survival analysis. Generally, the prognosis was worse for high-risk individuals. Subjects in the low-risk groups had a better prognosis and responded in a better way to combination immunotherapy, had higher immunological ratings and activity levels, and had more immune cell infiltration. In addition, gene set enrichment analysis findings showed that the low-risk subjects exhibited heightened activity in several immune-related pathways. However, the high-risk patients responded better to chemotherapy. The aim of this research was to develop a new ARG signature to predict the prognosis and sensitivity to immunotherapeutic and chemotherapeutic schemes for HNSCC patient. As a result, this could help spur the creation of new chemotherapeutics and immunotherapeutic approaches for patients with HNSCC.
Collapse
Affiliation(s)
- Zhengyu Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Yi Shen
- Health Science Center, Ningbo University, Ningbo, China
| | - Hongxia Deng
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
23
|
Han Z, Yi X, Li J, Zhang T, Liao D, You J, Ai J. RNA m 6A modification in prostate cancer: A new weapon for its diagnosis and therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188961. [PMID: 37507057 DOI: 10.1016/j.bbcan.2023.188961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Prostate cancer (PCa) is the most common malignant tumor and the second leading cause of cancer-related mortality in men worldwide. Despite significant advances in PCa therapy, the underlying molecular mechanisms have yet to be fully elucidated. Recently, epigenetic modification has emerged as a key player in tumor progression, and RNA-based N6-methyladenosine (m6A) epigenetic modification was found to be crucial. This review summarizes comprehensive state-of-art mechanisms underlying m6A modification, its implication in the pathogenesis, and advancement of PCa in protein-coding and non-coding RNA contexts, its relevance to PCa immunotherapy, and the ongoing clinical trials for PCa treatment. This review presents potential m6A-based targets and paves a new avenue for diagnosing and treating PCa, providing new guidelines for future related research through a systematic review of previous results.
Collapse
Affiliation(s)
- Zeyu Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Tianyi Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Dazhou Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China.
| |
Collapse
|
24
|
Zhang W, Xu L, Zhang X, Xu J, Jin JO. Escherichia coli adhesion portion FimH polarizes M2 macrophages to M1 macrophages in tumor microenvironment via toll-like receptor 4. Front Immunol 2023; 14:1213467. [PMID: 37720226 PMCID: PMC10502728 DOI: 10.3389/fimmu.2023.1213467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Background Macrophages are key effector cells of innate immunity and play a critical role in the immune balance of disease pathogenesis, especially in the tumor microenvironment. In previous studies, we showed that FimH, an Escherichia coli adhesion portion, promoted dendritic cell activation. However, the effect of FimH in macrophage polarization has yet to be fully examined. In this study, we investigated the potential effect of FimH on macrophages, as well as the polarization from M2 to M1 macrophages, contributing to the overall antitumor effect. Methods Mouse bone marrow derived macrophages and peritoneal macrophages were generated to test the effect of FimH in vitro. The expression of costimulatory molecules and production of cytokines were analyzed. The effect of FimH in the tumor-associated macrophages was examine in the B16F10-tumor bearing C57BL/6. Results FimH was found to promote M1 macrophage activation. In addition, FimH polarized M2 macrophages, which were induced by interleukin (IL)-4 and IL-13 into M1 macrophages were dependent on toll-like receptor 4 and myeloid differentiation factor 2. Moreover, FimH reprogramed the tumor-associated macrophage (TAM) into M1 macrophages in B16 melanoma tumor-bearing mice and promoted an inflammatory reaction in the tumor microenvironment (TME). Furthermore, FimH promoted M1 macrophage activation, as well as the reversion of M2 macrophages into M1 macrophages in humans. Finally, FimH treatment was found to enhance the anti-cancer immunity of anti-PD-L1 antibody by the induction of M1 polarization from TAM. Conclusion This study demonstrated the potential effect of FimH on the activation of macrophages, responsible for the repolarization of M2 macrophages into the M1 phenotype via the TLR4 signaling pathway. Moreover, FimH could also reprogram TAM polarization to the M1 status in the TME, as well as enhance the anti-tumor activity of immune checkpoint blockade.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Xu
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- The Laboratory for Immunotherapy, Clinical Center for BioTherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianqing Xu
- The Laboratory for Immunotherapy, Clinical Center for BioTherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- Dpartment of Microbiology, University of Ulsan College of Medicine, ASAN Medical Center, Seoul, Republic of Korea
| |
Collapse
|
25
|
Shu J, Jiang J, Zhao G. Identification of novel gene signature for lung adenocarcinoma by machine learning to predict immunotherapy and prognosis. Front Immunol 2023; 14:1177847. [PMID: 37583701 PMCID: PMC10424935 DOI: 10.3389/fimmu.2023.1177847] [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: 03/02/2023] [Accepted: 07/13/2023] [Indexed: 08/17/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) as a frequent type of lung cancer has a 5-year overall survival rate of lower than 20% among patients with advanced lung cancer. This study aims to construct a risk model to guide immunotherapy in LUAD patients effectively. Materials and methods LUAD Bulk RNA-seq data for the construction of a model, single-cell RNA sequencing (scRNA-seq) data (GSE203360) for cell cluster analysis, and microarray data (GSE31210) for validation were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. We used the Seurat R package to filter and process scRNA-seq data. Sample clustering was performed in the ConsensusClusterPlus R package. Differentially expressed genes (DEGs) between two groups were mined by the Limma R package. MCP-counter, CIBERSORT, ssGSEA, and ESTIMATE were employed to evaluate immune characteristics. Stepwise multivariate analysis, Univariate Cox analysis, and Lasso regression analysis were conducted to identify key prognostic genes and were used to construct the risk model. Key prognostic gene expressions were explored by RT-qPCR and Western blot assay. Results A total of 27 immune cell marker genes associated with prognosis were identified for subtyping LUAD samples into clusters C3, C2, and C1. C1 had the longest overall survival and highest immune infiltration among them, followed by C2 and C3. Oncogenic pathways such as VEGF, EFGR, and MAPK were more activated in C3 compared to the other two clusters. Based on the DEGs among clusters, we confirmed seven key prognostic genes including CPA3, S100P, PTTG1, LOXL2, MELTF, PKP2, and TMPRSS11E. Two risk groups defined by the seven-gene risk model presented distinct responses to immunotherapy and chemotherapy, immune infiltration, and prognosis. The mRNA and protein level of CPA3 was decreased, while the remaining six gene levels were increased in clinical tumor tissues. Conclusion Immune cell markers are effective in clustering LUAD samples into different subtypes, and they play important roles in regulating the immune microenvironment and cancer development. In addition, the seven-gene risk model may serve as a guide for assisting in personalized treatment in LUAD patients.
Collapse
Affiliation(s)
- Jianfeng Shu
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Jinni Jiang
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Guofang Zhao
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo, China
| |
Collapse
|
26
|
Feng X, Shan R, Hu X. The linkage of NF-κB signaling pathway-associated long non-coding RNAs with tumor microenvironment and prognosis in cervical cancer. BMC Med Genomics 2023; 16:169. [PMID: 37461017 PMCID: PMC10351132 DOI: 10.1186/s12920-023-01605-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND NF-κB signaling pathway participate closely in regulating inflammation and immune response in many cancers. Long non-coding RNAs (lncRNAs) associated with NF-κB signaling have not been characterized in cervical cancer. This study revealed the linkage between tumor microenvironment and NF-κB signaling-associated lncRNAs in cervical cancer. MATERIALS AND METHODS The expression profiles of cervical cancer samples from The Cancer Genome Atlas (TCGA) database were downloaded. NF-κB signaling-associated lncRNAs were screened as a basis to perform molecular subtyping. Immune cell infiltration was assessed by ESTIMATE, Microenvironment Cell Populations (MCP)-counter and single sample gene set enrichment analysis (ssGSEA). The key NF-κB signaling-associated lncRNAs were identified by univariate analysis, least absolute shrinkage and selection operator, and stepAIC. RESULTS Three molecular subtypes or clusters (cluster 3, cluster 2, and cluster 1) were categorized based on 27 prognostic NF-κB signaling-associated lncRNAs. Cluster 2 had the worst prognosis, highest immune infiltration, as well as the highest expression of most of immune checkpoints. Three clusters showed different sensitivities to immunotherapy and chemotherapy. Six key NF-κB signaling-associated lncRNAs were screened to establish a six-lncRNA risk model for predicting cervical cancer prognosis. CONCLUSIONS NF-κB signaling-associated lncRNAs played an important role in regulating immune microenvironment. The subtyping based on NF-κB signaling-associated lncRNAs may assist in the selection of optimal treatments. The six key NF-κB signaling-associated lncRNAs could act as prognostic biomarkers in prognostic prediction for cervical cancer.
Collapse
Affiliation(s)
- Xue Feng
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150010, China
| | - Ru Shan
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, 150010, China
| | - Xiaomeng Hu
- Department of Medical Psychology, Harbin Medical University, Harbin, 150010, China.
| |
Collapse
|
27
|
Duan X, Du H, Yuan M, Liu L, Liu R, Shi J. Bioinformatics analysis of necroptosis‑related lncRNAs and immune infiltration, and prediction of the prognosis of patients with esophageal carcinoma. Exp Ther Med 2023; 26:331. [PMID: 37346407 PMCID: PMC10280318 DOI: 10.3892/etm.2023.12030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/21/2023] [Indexed: 06/23/2023] Open
Abstract
Esophageal carcinoma (ESCA) is one of the most common malignancies in the world, and has high morbidity and mortality rates. Necrosis and long noncoding RNAs (lncRNAs) are involved in the progression of ESCA; however, the specific mechanism has not been clarified. The aim of the present study was to investigate the role of necrosis-related lncRNAs (nrlncRNAs) in patients with ESCA by bioinformatics analysis, and to establish a nrlncRNA model to predict ESCA immune infiltration and prognosis. To form synthetic matrices, ESCA transcriptome data and related information were obtained from The Cancer Genome Atlas. A nrlncRNA model was established by coexpression, univariate Cox (Uni-Cox), and least absolute shrinkage and selection operator analyses. The predictive ability of this model was evaluated by Kaplan-Meier, receiver operating characteristic (ROC) curve, Uni-Cox, multivariate Cox regression, nomogram and calibration curve analyses. A model containing eight nrlncRNAs was generated. The areas under the ROC curves for 1-, 3- and 5-year overall survival were 0.746, 0.671 and 0.812, respectively. A high-risk score according to this model could be used as an indicator for systemic therapy use, since the half-maximum inhibitory concentration values varied significantly between the high-risk and low-risk groups. Based on the expression of eight prognosis-related nrlncRNAs, the patients with ESCA were regrouped using the 'ConsensusClusterPlus' package to explore potential molecular subgroups responding to immunotherapy. The patients with ESCA were divided into three clusters based on the eight nrlncRNAs that constituted the risk model: The most low-risk group patients were classified into cluster 1, and the high-risk group patients were mainly concentrated in clusters 2 and 3. Survival analysis showed that Cluster 1 had a better survival than the other groups (P=0.016). This classification system could contribute to precision treatment. Furthermore, two nrlncRNAs (LINC02811 and LINC00299) were assessed in the esophageal epithelial cell line HET-1A, and in the human esophageal cancer cell lines KYSE150 and TE1. There were significant differences in the expression levels of these lncRNAs between tumor and normal cells. In conclusion, the present study suggested that nrlncRNA models may predict the prognosis of patients with ESCA, and provide guidance for immunotherapy and chemotherapy decision making. Furthermore, the present study provided strategies to promote the development of individualized and precise treatment for patients with ESCA.
Collapse
Affiliation(s)
- Xiaoyang Duan
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Huazhen Du
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Meng Yuan
- Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 804-8550, Japan
| | - Lie Liu
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Rongfeng Liu
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jian Shi
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
28
|
Zhang Q, Feng J, Liu K, Yang X, Huang Y, Tang B. STK11 mutation impacts CD1E expression to regulate the differentiation of macrophages in lung adenocarcinoma. Immun Inflamm Dis 2023; 11:e958. [PMID: 37506141 PMCID: PMC10373563 DOI: 10.1002/iid3.958] [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: 11/02/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The deficiency of serine/threonine protein kinase 11 (STK11), one of the most common tumor suppressor genes in non-small-cell lung cancer, is a crucial player in tumor immune microenvironment regulation. This study attempted to unveil how mutated STK11 impact the differentiation of macrophages in lung adenocarcinoma (LUAD). METHODS STK11 and CD1E expression levels in different cell models were assessed by quantitative reverse transcription polymerase chain reaction. Western blot was utilized to detect the protein expression levels of STK11, CD1E, apoptosis markers, and AMPK signaling pathway markers after transfection treatment. Cell viability and macrophage differentiation were detected by CCK-8 and flow cytometry. Immunohistochemistry and immunofluorescence were employed to detect the expression of related genes and macrophage markers, respectively. RESULTS This study found that STK11 mutations promoted the proliferation of LUAD cells and inhibited the differentiation of M1 macrophages, apoptosis, and the AMPK signaling pathway. Mutated STK11 led to CD1E downregulation, which curbed the differentiation of M1 macrophages and hence promoted LUAD progression. It was further validated by the in vivo experimental results that STK11 mutation significantly decreased the immune infiltration of M1 macrophages and promoted LUAD progression. CONCLUSION It was revealed that STK11 mutation affected CD1E expression to regulate macrophage differentiation in LUAD and then promote tumor progression. In this way, CD1E could be a potential biological target for the therapeutic interventions of STK11-mutant LUAD patients. These findings also threw new light on a new therapeutic strategy for STK11-mutant tumor patients that assisted the macrophage polarization pathway.
Collapse
Affiliation(s)
- Qingfeng Zhang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Juan Feng
- Department of Operating Room, Zigong Fourth People's Hospital, Zigong, China
| | - Kui Liu
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Xiaoyan Yang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Yun Huang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Bo Tang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| |
Collapse
|
29
|
Kumar V, Bauer C, Stewart JH. TIME Is Ticking for Cervical Cancer. BIOLOGY 2023; 12:941. [PMID: 37508372 PMCID: PMC10376148 DOI: 10.3390/biology12070941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
Cervical cancer (CC) is a major health problem among reproductive-age females and comprises a leading cause of cancer-related deaths. Human papillomavirus (HPV) is the major risk factor associated with CC incidence. However, lifestyle is also a critical factor in CC pathogenesis. Despite HPV vaccination introduction, the incidence of CC is increasing worldwide. Therefore, it becomes critical to understand the CC tumor immune microenvironment (TIME) to develop immune cell-based vaccination and immunotherapeutic approaches. The current article discusses the immune environment in the normal cervix of adult females and its role in HPV infection. The subsequent sections discuss the alteration of different immune cells comprising CC TIME and their targeting as future therapeutic approaches.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - Caitlin Bauer
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| | - John H Stewart
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
- Louisiana Children's Medical Center Cancer Center, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Science Center (LSUHSC), 1700 Tulane Avenue, New Orleans, LA 70012, USA
| |
Collapse
|
30
|
Shi Y, Huang J, Hu Y, Shen Y. Multiomics data analyses to identify SLC25A17 as a novel biomarker to predict the prognosis and immune microenvironment in head and neck squamous cell carcinoma. BMC Bioinformatics 2023; 24:269. [PMID: 37386359 DOI: 10.1186/s12859-023-05399-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023] Open
Abstract
OBJECTIVE This study aims to explore the predictive value of SLC25A17 in the prognosis and tumor microenvironment (TME) of patients with head and neck squamous cell carcinoma (HNSCC) and to provide ideas for individual clinical treatment. METHODS A pancancer analysis of the differential expression of SLC25A17 among different tumors was first conducted via the TIMER 2.0 database. Subsequently, the expression of SLC25A17 and related clinical information of HNSCC patients were obtained from the TCGA database, and patients were divided into two groups according to the median value of SLC25A17 expression. K‒M survival analysis was conducted to compare the overall survival (OS) and progression-free survival (PFS) between the groups. The Wilcoxon test was used to compare the distribution of SLC25A17 in different clinical characteristics, and univariate Cox and multivariate Cox analyses were performed to analyze independent prognostic factors to establish a predictive nomogram. Calibration curves were generated to verify the reliability of predicting 1-year, 3-year and 5-year survival rates and another cohort (GSE65858) was used for external validation. Gene set enrichment analysis was conducted to compare the enriched pathways, and the immune microenvironment was assessed using the CIBERSORT and estimate packages. Furthermore, the expression levels of SLC25A17 in immune cells were also analyzed with single-cell RNA-seq via the TISCH. Moreover, the immunotherapeutic response and chemotherapy drug sensitivity were compared between the two groups to guide precise treatment. The TIDE database was applied to predict the possibility of immune escape in the TCGA-HNSC cohort. RESULTS Compared with normal samples, the expression of SLC25A17 was much higher in HNSCC tumor samples. For patients with high SLC25A17 expression, the OS and PFS were shorter than those with low SLC25A17 expression, indicating a worse prognosis. The expression of SLC25A17 varied in different clinical features. Univariate Cox and multivariate COX analyses showed that SLC25A17, age, and lymph node metastasis are independent prognostic risk factors for HNSCC, and the survival prediction model based on these factors had reliable predictive value. Patients in the low-expression group exhibited more immune cell infiltration, higher TME scores, higher IPS scores and lower TIDE scores than those in the high-expression groups, suggesting better immunotherapeutic response with lower SLC25A17 expression. Moreover, patients in the high-expression group were more sensitive to chemotherapy. CONCLUSIONS SLC25A17 can effectively predict the prognosis of HNSCC patients and could be a precise individual-targeted indicator for the treatment of HNSCC patients.
Collapse
Affiliation(s)
- Yunbin Shi
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Juntao Huang
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Yi Hu
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yi Shen
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China.
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China.
- Department of Otolaryngology Head and Neck Surgery, Ningbo No.2 Hospital, Ningbo, China.
| |
Collapse
|
31
|
Li X, Yan X, Wang Y, Kaur B, Han H, Yu J. The Notch signaling pathway: a potential target for cancer immunotherapy. J Hematol Oncol 2023; 16:45. [PMID: 37131214 PMCID: PMC10155406 DOI: 10.1186/s13045-023-01439-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/13/2023] [Indexed: 05/04/2023] Open
Abstract
Dysregulation of the Notch signaling pathway, which is highly conserved across species, can drive aberrant epigenetic modification, transcription, and translation. Defective gene regulation caused by dysregulated Notch signaling often affects networks controlling oncogenesis and tumor progression. Meanwhile, Notch signaling can modulate immune cells involved in anti- or pro-tumor responses and tumor immunogenicity. A comprehensive understanding of these processes can help with designing new drugs that target Notch signaling, thereby enhancing the effects of cancer immunotherapy. Here, we provide an up-to-date and comprehensive overview of how Notch signaling intrinsically regulates immune cells and how alterations in Notch signaling in tumor cells or stromal cells extrinsically regulate immune responses in the tumor microenvironment (TME). We also discuss the potential role of Notch signaling in tumor immunity mediated by gut microbiota. Finally, we propose strategies for targeting Notch signaling in cancer immunotherapy. These include oncolytic virotherapy combined with inhibition of Notch signaling, nanoparticles (NPs) loaded with Notch signaling regulators to specifically target tumor-associated macrophages (TAMs) to repolarize their functions and remodel the TME, combining specific and efficient inhibitors or activators of Notch signaling with immune checkpoint blockers (ICBs) for synergistic anti-tumor therapy, and implementing a customized and effective synNotch circuit system to enhance safety of chimeric antigen receptor (CAR) immune cells. Collectively, this review aims to summarize how Notch signaling intrinsically and extrinsically shapes immune responses to improve immunotherapy.
Collapse
Affiliation(s)
- Xinxin Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, People's Republic of China
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Yufeng Wang
- Cancer Institute, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77225, USA
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China.
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA.
| |
Collapse
|
32
|
Naletova I, Tomasello B, Attanasio F, Pleshkan VV. Prospects for the Use of Metal-Based Nanoparticles as Adjuvants for Local Cancer Immunotherapy. Pharmaceutics 2023; 15:1346. [PMID: 37242588 PMCID: PMC10222518 DOI: 10.3390/pharmaceutics15051346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Immunotherapy is among the most effective approaches for treating cancer. One of the key aspects for successful immunotherapy is to achieve a strong and stable antitumor immune response. Modern immune checkpoint therapy demonstrates that cancer can be defeated. However, it also points out the weaknesses of immunotherapy, as not all tumors respond to therapy and the co-administration of different immunomodulators may be severely limited due to their systemic toxicity. Nevertheless, there is an established way through which to increase the immunogenicity of immunotherapy-by the use of adjuvants. These enhance the immune response without inducing such severe adverse effects. One of the most well-known and studied adjuvant strategies to improve immunotherapy efficacy is the use of metal-based compounds, in more modern implementation-metal-based nanoparticles (MNPs), which are exogenous agents that act as danger signals. Adding innate immune activation to the main action of an immunomodulator makes it capable of eliciting a robust anti-cancer immune response. The use of an adjuvant has the peculiarity of a local administration of the drug, which positively affects its safety. In this review, we will consider the use of MNPs as low-toxicity adjuvants for cancer immunotherapy, which could provide an abscopal effect when administered locally.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, 95125 Catania, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
33
|
He Z, Song B, Zhu M, Liu J. Comprehensive pan-cancer analysis of STAT3 as a prognostic and immunological biomarker. Sci Rep 2023; 13:5069. [PMID: 36977736 PMCID: PMC10050087 DOI: 10.1038/s41598-023-31226-2] [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] [Received: 10/13/2022] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Numerous studies have indicated that STAT3 plays a key role in promoting oncogenesis and it is considered a potential therapeutic target for cancer treatment; however, there are no reports on STAT3 using pan-cancer analysis. Therefore, it is important to investigate the role of STAT3 in different types of tumors using pan-cancer analysis. In the present study, we used multiple databases to comprehensively analyze the relationship between STAT3 expression and prognosis, different stages of patients with cancer, investigate the clinical value of STAT3 in predicting prognosis, and the relationship between STAT3 genetic alteration and prognosis, drug sensitivity, and STAT3 expression, to determine whether STAT3 participates in tumor immunity, to provide a rationale for STAT3 as a treatment target for a broad-spectrum malignancies. Our results indicate that STAT3 can serve as a prognostic, sensitivity prediction biomarker and a target for immunotherapy, which has been of great value for pan-cancer treatment. Overall, we found that STAT3 significantly predicted cancer prognosis, drug resistance, and immunotherapy, providing a rationale for further experimental studies.
Collapse
Affiliation(s)
- Zhibo He
- The School of Foreign Languages, Jiujiang University, Jiujiang, China
| | - Biao Song
- Medical School, Jiujiang University, Jiujiang, China
| | - Manling Zhu
- Medical School, Jiujiang University, Jiujiang, China
| | - Jun Liu
- Medical School, Jiujiang University, Jiujiang, China.
- Laboratory of Precision Preventive Medicine, Jiujiang University, Jiujiang, China.
| |
Collapse
|
34
|
Zhang X, Deng Q, Wan X, Zhao J, Zheng X, Wang H, Wang HQ, Yang W. Pan-cancer analysis reveals the associations between MMP13 high expression and carcinogenesis and its value as a serum diagnostic marker. Aging (Albany NY) 2023; 15:2115-2135. [PMID: 37000142 PMCID: PMC10085597 DOI: 10.18632/aging.204599] [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: 10/29/2022] [Accepted: 03/08/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND Matrix metalloproteinase-13 (MMP13) is a member of the endopeptidase matrix metalloproteinase family, which is involved in many normal physiological processes and even tumorigenesis. However, its co-carcinogenic signature in different cancers is not fully understood. METHODS In this study, we first analyzed the expression of MMP13 in pan-cancer and its association with prognosis, immune infiltration, and cancer-related signaling pathways through integrated bioinformatics. Furthermore, western blotting (WB) was used to verify the expression of MMP13 and epithelial-mesenchymal transition (EMT) factors in cancer tissues. Finally, the value of MMP13 as a serum diagnostic marker was analyzed by enzyme-linked immunosorbent assay (ELISA). RESULTS MMP13 expression is frequently upregulated in multiple cancers that always indicate an adverse prognosis. MMP13 undergoes comprehensive genetic alterations and promoter methylation reduction in various tumors. Additionally, immune correlation analysis showed that MMP13 expression was significantly associated with TMB, MSI, and tumor immune infiltration. Pathway enrichment analysis showed that MMP13 upregulation was correlated with activation of the EMT signaling pathway, which was verified by WB in lung adenocarcinoma tissues. Most importantly, ELISA results showed that serum MMP13 levels could be used for the diagnosis of multiple tumors, including BRCA, HNSC, LUAD, and LUSC, with the area under the curve (AUC) values of 0.8494, 0.9259, 0.7144, and 0.8575, respectively. CONCLUSIONS MMP13 is often overexpressed across cancers and predicts poor prognosis, with the potential as a therapeutic target. Furthermore, the up-regulation of its expression can be effectively reflected in the serum protein level, thus serving as a valuable diagnostic marker.
Collapse
Affiliation(s)
- Xinhui Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Qingmei Deng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiaofeng Wan
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Jingyu Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Xin Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Hong-Qiang Wang
- Biological Molecular Information System Laboratory, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Wulin Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Medical Pathology Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
35
|
Tu Z, Wang X, Cai H, Sheng Y, Wu L, Huang K, Zhu X. The cell senescence regulator p16 is a promising cancer prognostic and immune check-point inhibitor (ICI) therapy biomarker. Aging (Albany NY) 2023; 15:2136-2157. [PMID: 36961395 PMCID: PMC10085592 DOI: 10.18632/aging.204601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/01/2023] [Indexed: 03/25/2023]
Abstract
Cyclin-dependent kinase inhibitor 2A (CDKN2A) encodes the cell senescence regulator protein p16. The expression of p16 raises in cell senescence and has a nuclear regulation in cell aging. Meanwhile, it's also reported to inhibit the aggression of several cancers. But its clinical application and role in cancer immunotherapy needs further investigation. We collected the transcriptional data of pan-cancer and normal human tissues from The Cancer Genome Atlas and the Genotype-Tissue Expression databases. CBioPortal webtool was employed to mine the genomic alteration status of CDKN2A across cancers. Kaplan-Meier method and univariate Cox regression were performed for prognostic assessments across cancers, respectively. Gene Set Enrichment Analysis is the main method used to search the associated cancer hallmarks associated with CDKN2A. TIMER2.0 was used to analyze the immune cell infiltration relevance with CDKN2A in pan-cancer. The associations between CDKN2A and immunotherapy biomarkers or regulators were performed by spearman correlation analysis. We found CDKN2A is overexpressed in most cancers and exhibits prognosis predictive ability in various cancers. In addition, it is significantly correlated with immune-activated hallmarks, cancer immune cell infiltrations and immunoregulators. The most interesting finding is that CDKN2A can significantly predict anti-PDL1 therapy response. Finally, specific inhibitors which correlated with CDKN2A expression in different cancer types were also screened by using Connectivity Map (CMap) tool. The results revealed that CDKN2A acts as a robust cancer prognostic and immunotherapy biomarker. Its function in the regulation of cancer cell senescence might shape the tumor microenvironment and contribute to its predictive ability of immunotherapy.
Collapse
Affiliation(s)
- 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
| | - Xiaolin Wang
- The Second Clinical Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Huan Cai
- Department of Medical Ultrasonics, Integrated Chinese and Western Medicine Hospital of Jiangxi Province, Nanchang 330006, Jiangxi, P.R. China
| | - Yilei Sheng
- The HuanKui Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Lei Wu
- 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
| | - 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
| |
Collapse
|
36
|
Vitale C, Bottino C, Castriconi R. Monocyte and Macrophage in Neuroblastoma: Blocking Their Pro-Tumoral Functions and Strengthening Their Crosstalk with Natural Killer Cells. Cells 2023; 12:885. [PMID: 36980226 PMCID: PMC10047506 DOI: 10.3390/cells12060885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Over the past decade, immunotherapy has represented an enormous step forward in the fight against cancer. Immunotherapeutic approaches have increasingly become a fundamental part of the combined therapies currently adopted in the treatment of patients with high-risk (HR) neuroblastoma (NB). An increasing number of studies focus on the understanding of the immune landscape in NB and, since this tumor expresses low or null levels of MHC class I, on the development of new strategies aimed at enhancing innate immunity, especially Natural Killer (NK) cells and macrophages. There is growing evidence that, within the NB tumor microenvironment (TME), tumor-associated macrophages (TAMs), which mainly present an M2-like phenotype, have a crucial role in mediating NB development and immune evasion, and they have been correlated to poor clinical outcomes. Importantly, TAM can also impair the antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells upon the administration of anti-GD2 monoclonal antibodies (mAbs), the current standard immunotherapy for HR-NB patients. This review deals with the main mechanisms regulating the crosstalk among NB cells and TAMs or other cellular components of the TME, which support tumor development and induce drug resistance. Furthermore, we will address the most recent strategies aimed at limiting the number of pro-tumoral macrophages within the TME, reprogramming the TAMs functional state, thus enhancing NK cell functions. We also prospectively discuss new or unexplored aspects of human macrophage heterogeneity.
Collapse
Affiliation(s)
- Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
37
|
Gargiulo E, Giordano M, Niemann CU, Moussay E, Paggetti J, Morande PE. The protective role of the microenvironment in hairy cell leukemia treatment: Facts and perspectives. Front Oncol 2023; 13:1122699. [PMID: 36968995 PMCID: PMC10031020 DOI: 10.3389/fonc.2023.1122699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Hairy cell leukemia (HCL) is an incurable, rare lymphoproliferative hematological malignancy of mature B cAlthough first line therapy with purine analogues leads to positive results, almost half of HCL patients relapse after 5-10 years, and standard treatment may not be an option due to intolerance or refractoriness. Proliferation and survival of HCL cells is regulated by surrounding accessory cells and soluble signals present in the tumor microenvironment, which actively contributes to disease progression. In vitro studies show that different therapeutic approaches tested in HCL impact the tumor microenvironment, and that this milieu offers a protection affecting treatment efficacy. Herein we explore the effects of the tumor microenvironment to different approved and experimental therapeutic options for HCL. Dissecting the complex interactions between leukemia cells and their milieu will be essential to develop new targeted therapies for HCL patients.
Collapse
Affiliation(s)
- Ernesto Gargiulo
- Tumor Stroma Interactions – Department of Cancer Research, Luxembourg Institute of HealthLuxembourg, Luxembourg
- Chronic Lymphocytic Leukemia Laboratory, Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- PERSIMUNE, Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Mirta Giordano
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carsten U. Niemann
- Chronic Lymphocytic Leukemia Laboratory, Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Etienne Moussay
- Tumor Stroma Interactions – Department of Cancer Research, Luxembourg Institute of HealthLuxembourg, Luxembourg
| | - Jérôme Paggetti
- Tumor Stroma Interactions – Department of Cancer Research, Luxembourg Institute of HealthLuxembourg, Luxembourg
| | - Pablo Elías Morande
- Tumor Stroma Interactions – Department of Cancer Research, Luxembourg Institute of HealthLuxembourg, Luxembourg
- Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|
38
|
Ultrasound-targeted microbubble destruction remodels tumour microenvironment to improve immunotherapeutic effect. Br J Cancer 2023; 128:715-725. [PMID: 36463323 PMCID: PMC9977958 DOI: 10.1038/s41416-022-02076-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer immunotherapy (CIT) has gained increasing attention and made promising progress in recent years, especially immune checkpoint inhibitors such as antibodies blocking programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). However, its therapeutic efficacy is only 10-30% in solid tumours and treatment sensitivity needs to be improved. The complex tissue environment in which cancers originate is known as the tumour microenvironment (TME) and the complicated and dynamic TME is correlated with the efficacy of immunotherapy. Ultrasound-targeted microbubble destruction (UTMD) is an emerging technology that integrates diagnosis and therapy, which has garnered much traction due to non-invasive, targeted drug delivery and gene transfection characteristics. UTMD has also been studied to remodel TME and improve the efficacy of CIT. In this review, we analyse the effects of UTMD on various components of TME, including CD8+ T cells, tumour-infiltrating myeloid cells, regulatory T cells, natural killer cells and tumour vasculature. Moreover, UTMD enhances the permeability of the blood-brain barrier to facilitate drug delivery, thus improving CIT efficacy in vivo animal experiments. Based on this, we highlight the potential of immunotherapy against various cancer species and the clinical application prospects of UTMD.
Collapse
|
39
|
Sun P, Li Z, Zhang D, Zeng W, Zheng Y, Mei L, Chen H, Gao N, Zeng X. Multifunctional biodegradable nanoplatform based on oxaliplatin prodrug cross-linked mesoporous polydopamine for enhancing cancer synergetic therapy. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
40
|
Pilavaki P, Panagi M, Arifi S, Jones RL, Stylianopoulos T, Constantinidou A. Exploring the landscape of immunotherapy approaches in sarcomas. Front Oncol 2023; 12:1069963. [PMID: 36686827 PMCID: PMC9853527 DOI: 10.3389/fonc.2022.1069963] [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: 10/14/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Sarcomas comprise a heterogenous group of malignancies, of more than 100 different entities, arising from mesenchymal tissue, and accounting for 1% of adult malignancies. Surgery, radiotherapy and systemic therapy constitute the therapeutic armamentarium against sarcomas, with surgical excision and conventional chemotherapy, remaining the mainstay of treatment for local and advanced disease, respectively. The prognosis for patients with metastatic disease is dismal and novel therapeutic approaches are urgently required to improve survival outcomes. Immunotherapy, is a rapidly evolving field in oncology, which has been successfully applied in multiple cancers to date. Immunomodulating antibodies, adoptive cellular therapy, cancer vaccines, and cytokines have been tested in patients with different types of sarcomas through clinical trials, pilot studies, retrospective and prospective studies. The results of these studies regarding the efficacy of different types of immunotherapies in sarcomas are conflicting, and the application of immunotherapy in daily clinical practice remains limited. Additional clinical studies are ongoing in an effort to delineate the role of immunotherapy in patients with specific sarcoma subtypes.
Collapse
Affiliation(s)
- Pampina Pilavaki
- Medical School, University of Cyprus, Nicosia, Cyprus
- Medical Oncology, Bank of Cyprus Oncology Center, Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering University of Cyprus, Nicosia, Cyprus
| | - Samia Arifi
- Medical Oncology Department, Hassan II University Hospital/Faculty of Medicine and Pharmacy University of Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Robin L. Jones
- Sarcoma Unit, Royal Marsden National Health Service (NHS) Foundation Trust, London, London, United Kingdom
- Sarcoma Clinical Trial Unit, Institute of Cancer Research, London, United Kingdom
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering University of Cyprus, Nicosia, Cyprus
| | - Anastasia Constantinidou
- Medical School, University of Cyprus, Nicosia, Cyprus
- Medical Oncology, Bank of Cyprus Oncology Center, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| |
Collapse
|
41
|
Deng H, Deng D, Qi T, Liu Z, Wu L, Yuan J. An IFN-γ-related signature predicts prognosis and immunotherapy response in bladder cancer: Results from real-world cohorts. Front Genet 2023; 13:1100317. [PMID: 36685901 PMCID: PMC9846040 DOI: 10.3389/fgene.2022.1100317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Bladder cancer (BLCA) is featured with high incidence and mortality. Whether the IFN-γ signaling could be used as an immunotherapy determinant for BLCA has not been fully confirmed. In this study, the transcriptome data and clinical information of BLCA samples were collected from The Cancer Genome Atlas (TCGA). Besides, four immunotherapy cohorts including IMvigor210 cohort, Gide cohort, Van Allen cohort, and Lauss cohort were collected. The Xiangya real-world cohort was used for independent validation. An IFN-γ-related signature was developed and validated in BLCA for predicting prognosis, mutation, tumor microenvironment status, and immunotherapy response. This is the first study focusing on the comprehensive evaluation of predictive values on the IFN-γ-related signature in BLCA. The potential clinical application of the IFN-γ-related signature was expected to be further validated with more prospective clinical cohorts.
Collapse
Affiliation(s)
- Hao Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Dingshan Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tiezheng Qi
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhi Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Longxiang Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Junbin Yuan, ; Longxiang Wu,
| | - Junbin Yuan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Junbin Yuan, ; Longxiang Wu,
| |
Collapse
|
42
|
Wei M, Chen A, Zhang J, Ren Y. Novel Oxygen-Dependent Degradable Immunotoxin Regulated by the Ubiquitin-Proteasome System Reduces Nonspecific Cytotoxicity. Mol Pharm 2023; 20:90-100. [PMID: 36305716 DOI: 10.1021/acs.molpharmaceut.2c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The use of bacterial toxins as antitumor agents has received considerable attention. Immunotoxins based on antigen recognition of single-chain antibodies have been widely explored for cancer therapy. Despite their impressive killing effect on tumor cells, immunotoxins still display unspecific toxicity with undesired side effects. High levels of hypoxia-inducible factor 1α (HIF-1α) are well-known indicators of hypoxia in cancer cells. In this study, different linkers were employed to fuse the immunotoxin DAB389-4D5 scFv (DS) with the oxygen-dependent degradation domain (ODDD) of HIF-1α, a domain selectively facilitating the accumulation of HIF-1α under hypoxia, to construct the oxygen-dependent degradable immunotoxin DS-ODDD (DSO). The engineered fusion protein DSO-2 containing a linker (G4S)3 possesses the best killing effect on cancer cells under hypoxia and displayed considerably reduced nonspecific toxicity to normal cells under normoxic conditions. Flow cytometry, immunofluorescence, and immunoblot analyses demonstrated that DSO-2 was degraded via the ubiquitin-proteasome pathway regulated by the oxygen-sensitive mechanism. Western blot analysis indicated that the degradation of DSO-2 significantly decreased the activation of apoptosis-related molecules in normal cells. The engineered immunotoxin with oxygen-sensing properties developed herein is a potential therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Min Wei
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Anxiang Chen
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jian Zhang
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuhong Ren
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
43
|
Feng Y, Tao F, Qiao H, Tang H. A pan-cancer analysis of ABI3BP: a potential biomarker for prognosis and immunoinfiltration. Front Oncol 2023; 13:1159725. [PMID: 37197424 PMCID: PMC10183607 DOI: 10.3389/fonc.2023.1159725] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Objective ABI Family Member 3 Binding Protein (ABI3BP) is an extracellular matrix protein that affects the carcinogenesis of lung and esophageal cancer. However, the relevance of ABI3BP in different forms of cancer is uncertain. Methods ABI3BP expression was interpreted using the Cancer Genome Atlas (TCGA) database, the Genotype Tissue Expression Atlas (GTEx) database, the Human Protein Atlas (HPA) database, the Cancer Cell Line Encyclopedia (CCLE) database, and immunohistochemistry. The R programming language was used to analyze the association between ABI3BP expression and patient prognosis and evaluate the relationship between ABI3BP and the immune characteristics of tumors. Using the GDSC and CTRP databases, a drug sensitivity analysis of ABI3BP was conducted. Results ABI3BP mRNA expression was shown by differential analysis to be down-regulated in 16 tumor types relative to normal tissues, corresponding with its protein expression level as determined by immunohistochemistry. Abnormal expression of ABI3BP accurately predicts the prognosis of patients with renal chromophobe carcinoma (KICH), mesothelioma (MESO), and pancreatic adenocarcinoma (PAAD). Meanwhile, aberrant expression of ABI3BP was associated with immune checkpoints, TMB, MSI, tumor purity, HRD, LOH, and drug sensitivity. A correlation between ABI3BP expression and the amount of infiltration of several immune-related cells in pan-cancer was determined by Immune Score, Stromal Score, and Estimated Score. Conclusion Our results show that ABI3BP might be employed as a molecular biomarker to predict prognosis, treatment susceptibility, and immunological response in patients with pan-cancer.
Collapse
Affiliation(s)
- Yan Feng
- Department of Respiratory Medicine, Qingdao University, Qingdao, China
| | - Fengying Tao
- Department of Oncology Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Han Qiao
- Department of Respiratory Medicine, Qingdao University, Qingdao, China
| | - Huaping Tang
- Department of Respiratory Medicine, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- *Correspondence: Huaping Tang,
| |
Collapse
|
44
|
Sun D, Zhang H, Zhang C, Wang L. An evaluation of KIF20A as a prognostic factor and therapeutic target for lung adenocarcinoma using integrated bioinformatics analysis. Front Bioeng Biotechnol 2022; 10:993820. [PMID: 36619388 PMCID: PMC9816395 DOI: 10.3389/fbioe.2022.993820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
The identification of prognostic and therapeutic biomarkers is essential to reduce morbidity and mortality from lung adenocarcinoma (LUAD). This study aimed to identify a reliable prognostic and therapeutic biomarker for LUAD using integrated bioinformatics. Based on the cancer genome atlas (TCGA) and genome-tissue expression (GTEx) analyses, KIF20A has been identified as the hub gene. Following validation using a series of cohorts, survival analysis, meta-analysis, and univariate Cox analysis was conducted. ESTIMATE and CIBERSORT algorithms were then used to study the association of KIF20A with the tumor microenvironment (TME) and the percentage of tumor-infiltrating immune cells (TICs). In vitro experiments were conducted to determine the function of KIF20A. Finally, there was a negative association between the expression of the KIF20A and overall survival, progression-free survival, and disease-free survival, which was confirmed by meta-analysis and COX analysis. Furthermore, KIF20A also had a potential role of altering the TME and TICs proportions in LUAD. Validations in vitro were performed on A549 and PC-9 cell lines, and we found that the knockdown of KIF20A exhibited inhibitory effects on cell proliferation, resulted in cell cycle arrest during the G2/M phase, and induced cellular apoptosis. Our study demonstrated that KIF20A could be utilized as a reliable prognostic marker and treatment target for LUAD. However, further studies are required to validate these findings.
Collapse
Affiliation(s)
- Dongjie Sun
- College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haiying Zhang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chi Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Lina Wang
- Department of Pediatric Respiration, The First Hospital of Jilin University, Changchun, China,*Correspondence: Lina Wang,
| |
Collapse
|
45
|
Alrhmoun S, Sennikov S. The Role of Tumor-Associated Antigen HER2/neu in Tumor Development and the Different Approaches for Using It in Treatment: Many Choices and Future Directions. Cancers (Basel) 2022; 14:6173. [PMID: 36551661 PMCID: PMC9776683 DOI: 10.3390/cancers14246173] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The treatment of HER2-positive cancers has changed significantly over the past ten years thanks to a significant number of promising new approaches that have been added to our arsenal in the fight against cancer, including monoclonal antibodies, inhibitors of tyrosine kinase, antibody-drug conjugates, vaccination, and particularly, adoptive-T-cell therapy after its great success in hematological malignancies. Equally important is the new methodology for determining patients eligible for targeted HER2 therapy, which has doubled the number of patients who can benefit from these treatments. However, despite the initial enthusiasm, there are still several problems in this field represented by drug resistance and tumor recurrence that require the further development of new more efficient drugs. In this review, we discuss various approaches for targeting the HER2 molecule in cancer treatment, highlighting their benefits and drawbacks, along with the different mechanisms responsible for resistance to HER2-targeted therapies and how to overcome them.
Collapse
Affiliation(s)
- Saleh Alrhmoun
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia
- Department of Immunology, V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
46
|
Wu C, Zhong R, Sun X, Shi J. PSME2 identifies immune-hot tumors in breast cancer and associates with well therapeutic response to immunotherapy. Front Genet 2022; 13:1071270. [PMID: 36583022 PMCID: PMC9793949 DOI: 10.3389/fgene.2022.1071270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BrCa) is a heterogeneous disease, which leads to unsatisfactory prognosis in females worldwide. Previous studies have proved that tumor immune microenvironment (TIME) plays crucial roles in oncogenesis, progression, and therapeutic resistance in Breast cancer. However, biomarkers related to TIME features have not been fully discovered. Proteasome activator complex subunit 2 (PSME2) is a member of proteasome activator subunit gene family, which is critical to protein degradation mediated by the proteasome. In the current research, we comprehensively analyzed the expression and immuno-correlations of Proteasome activator complex subunit 2 in Breast cancer. Proteasome activator complex subunit 2 was significantly upregulated in tumor tissues but associated with well prognosis. In addition, Proteasome activator complex subunit 2 was overexpressed in HER2-positive Breast cancer but not related to other clinicopathological features. Interestingly, Proteasome activator complex subunit 2 was positively related to immune-related processes and identified immuno-hot TIME in Breast cancer. Specifically, Proteasome activator complex subunit 2 was positively correlated with immunomodulators, tumor-infiltrating immune cells (TIICs), immune checkpoints, and tumor mutation burden (TMB) levels. Moreover, the positive correlation between Proteasome activator complex subunit 2 and PD-L1 expression was confirmed in a tissue microarray (TMA) cohort. Furthermore, in an immunotherapy cohort of Breast cancer, patients with pathological complete response (pCR) expressed higher Proteasome activator complex subunit 2 compared with those with non-pathological complete response. In conclusion, Proteasome activator complex subunit 2 is upregulated in tumor tissues and correlated with the immuno-hot tumor immune microenvironment, which can be a novel biomarker for the recognition of tumor immune microenvironment features and immunotherapeutic response in Breast cancer.
Collapse
Affiliation(s)
- Cen Wu
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Ren Zhong
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Xiaofei Sun
- Department of General Surgery, Rudong County People’s Hospital, Nantong, China
| | - Jiajie Shi
- Departments of Breast Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
47
|
Huang J, Xu Z, Yuan Z, Teh BM, Zhou C, Shen Y. Identification of a cuproptosis-related lncRNA signature to predict the prognosis and immune landscape of head and neck squamous cell carcinoma. Front Oncol 2022; 12:983956. [PMID: 36568234 PMCID: PMC9780454 DOI: 10.3389/fonc.2022.983956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Background Cuproptosis is considered a novel copper-induced cell death model regulated by targeting lipoylated TCA cycle proteins. In this study, we established a novel signature based on cuproptosis-related lncRNAs (crlncRNAs) to predict the prognosis and immune landscape of head and neck squamous cell carcinoma. Methods RNA-seq matrix, somatic mutation files, and clinical data were obtained from The Cancer Genome Atlas database. After dividing patients into two sets, a crlncRNA signature was established based on survival related crlncRNAs, which were selected by the univariate Cox analysis and least absolute shrinkage and selection operator Cox regression. To evaluate the model, Kaplan-Meier survival analysis and time-dependent receiver operating characteristic (ROC) were utilized, and a nomogram was established for survival prediction. Immune landscape analysis, drug sensitivity, cluster analysis, tumor mutation burden (TMB) and ceRNA network analysis were conducted subsequently. Results A crlncRNA related prognosis signature was finally constructed with 12 crlncRNAs. The areas under the ROC curves (AUCs) were 0.719, 0.705 and 0.693 respectively for 1, 3, and 5-year's overall survival (OS). Patients in the low-risk group behaved a better prognosis, lower TMB, higher immune function activity and scores. In addition, patients from cluster 2 were more sensitive to chemotherapy and immunotherapy. Conclusion In this study, we constructed a novel crlncRNA risk model to predict the survival of HNSCC patients. This reliable and acceptable prognostic signature may guide and promote the progress of novel treatment strategies for HNSCC patients.
Collapse
Affiliation(s)
- Juntao Huang
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China,School of Medicine, Ningbo University, Ningbo, Zhejiang, China,*Correspondence: Juntao Huang, ; Yi Shen,
| | - Ziqian Xu
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, Ningbo, China
| | - Zhechen Yuan
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China,School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Bing Mei Teh
- Department of Ear Nose and Throat, Head and Neck Surgery, Eastern Health, Box Hill, VA, Australia,Department of Otolaryngology, Head and Neck Surgery, Monash Health, Clayton, VA, Australia,Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VA, Australia
| | - Chongchang Zhou
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China,School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yi Shen
- Department of Otolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China,School of Medicine, Ningbo University, Ningbo, Zhejiang, China,*Correspondence: Juntao Huang, ; Yi Shen,
| |
Collapse
|
48
|
Wang J, Li Z. TREM2 Is a Prognostic Biomarker and Correlated with an Immunosuppressive Microenvironment in Thyroid Cancer. DISEASE MARKERS 2022; 2022:1807386. [PMID: 36438899 PMCID: PMC9683966 DOI: 10.1155/2022/1807386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 03/12/2024]
Abstract
PURPOSES To identify the differentially expressed genes (DEGs) related to the immune microenvironment and elucidate the biological functions of key genes in papillary thyroid cancer (PTC) by analyzing the immune microenvironment. METHODS The relative quantities of immune and matrix components in 507 patients with PTC were calculated from the TCGA database. Analysis of differentially expressed genes in tumor samples throughout the genome, intersection of DEGs obtained from PTC patients, and genome-wide tumor samples and survival analysis were performed. Survival analysis was used for identification of prognostic factor. Immunohistochemical analysis of the TREM2 expression in PTC tissues, flow cytometry, and transwell assays were used to detect the effect of TREM2 on PTC cell proliferation, migration, and invasion. RESULTS There were a total of 1242 upregulated genes with high intersection in the immune score and 124 downregulated genes with low intersection in the stromal score. A total of 1,366 genes in these DEGs may be determinants in the immune microenvironment. GO enrichment and KEGG enrichment analysis revealed that the overall function of DEGs appeared to map onto immune-related activities. Gene intersection and survival analysis showed that there were 435 DEG crosses in PTC patients and genome-wide tumor samples, only CXCL10, CD40LG, KRT14, TRAT1, and TREM2 were associated with patient prognosis, and TCGA showed that only the TREM2 expression was upregulated in PTC. TREM2 knockdown inhibited the cell cycle and cell proliferation, migration, and invasion by PTC cells. TREM2 was associated with the immunosuppressive microenvironment by via NF-κB pathway in PTC. CONCLUSION TREM2 possibly was a potential indicator of altered TME status in PTC, and that TREM2 promoted PTC cell proliferation and cell cycle, migration, and invasion by NF-κB pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, NO.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Zhendong Li
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, NO.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
49
|
Sha YL, Liu Y, Yang JX, Wang YY, Gong BC, Jin Y, Qu TY, Xia FT, Han L, Zhao Q. B3GALT4 remodels the tumor microenvironment through GD2-mediated lipid raft formation and the c-met/AKT/mTOR/IRF-1 axis in neuroblastoma. J Exp Clin Cancer Res 2022; 41:314. [PMID: 36284313 PMCID: PMC9594894 DOI: 10.1186/s13046-022-02523-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Beta-1,3-galactosyltransferase-4 (B3GALT4) plays a critical regulatory role in tumor biology. However, the role of B3GALT4 in modulating the tumor microenvironment (TME) of neuroblastoma (NB) remains unknown. METHODS Public datasets and clinical NB samples were collected to evaluate the expression and clinical significance of GD2 and B3GALT4 in NB patients. CCK-8, colony formation, and transwell assays and experiments in tumor-bearing mouse models were conducted to investigate the function of B3GALT4. Flow cytometry, ELISA, immunohistochemistry, immunofluorescence, western blotting, and chemotaxis assays were conducted to ascertain the immunomodulatory mechanism of B3GALT4. The combined therapeutic effect of the lipid raft inhibitor MβCD and anti-GD2 mAb was validated in a murine model of NB. RESULTS GD2 was overexpressed in NB tissues and high expression of GD2 was associated with poor prognosis in NB patients. B3GALT4 was downregulated in NB tissues, and low expression of B3GALT4 indicated poor prognosis in NB patients. Silencing B3GALT4 significantly enhanced tumor progression both in vitro and in vivo. Meanwhile, the overexpression of B3GALT4 increased the recruitment of CD8+ T lymphocytes via the chemokines CXCL9 and CXCL10. Additionally, B3GALT4 regulated NB-cell GD2 expression and lipid raft formation. Mechanistically, B3GALT4 regulated the expression of CXCL9 and CXCL10 via the c-Met signaling in the lipid rafts and the downstream AKT/mTOR/IRF-1 pathway. The lipid raft inhibitor, MβCD, attenuated B3GALT4 deficiency-induced tumor progression and immune evasion. Last, MβCD combined with anti-GD2 mAb treatment significantly enhanced the antitumor effect and the infiltration of CD8+ T cells. CONCLUSIONS Upregulation of B3GALT4 promotes the secretion of CXCL9 and CXCL10 to recruit CD8+ T lymphocytes via the GD2-mediated lipid rafts and the c-Met/AKT/mTOR/IRF-1 pathway. Moreover, lipid raft inhibitors may enhance the efficacy of anti-GD2 immunotherapy for NB.
Collapse
Affiliation(s)
- Yong-Liang Sha
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jia-Xing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yang-Yang Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bao-Cheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tong-Yuan Qu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Fan-Tong Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
50
|
Fan L, Wang X, Chang Q, Wang Y, Yang W, Liu L. IL2RA is a prognostic indicator and correlated with immune characteristics of pancreatic ductal adenocarcinoma. Medicine (Baltimore) 2022; 101:e30966. [PMID: 36281157 PMCID: PMC9592409 DOI: 10.1097/md.0000000000030966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and incurable cancer with a dismal prognosis. In this study, we aimed to explore potential predictors for the prognosis and immunological characteristics of PDAC. Estimation of stromal and immune cells in malignant tumors, using expression data (ESTIMATE) method was applied to calculate the immune and stromal scores of 206 PDAC samples from GSE71729. R package of "limma" was utilized to identify differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were conducted for functional exploration. Protein-protein interaction (PPI) network and Univariate Cox analysis were conducted to select key prognostic genes of PDAC. Gene set enrichment analysis (GSEA) was applied to investigate the roles of IL2RA in PDAC. Single sample GSEA (ssGSEA) was performed to evaluate the immunological characteristics of PDAC samples. Wilcoxon rank sum test was conducted to compare the difference of immunological characteristics of PDAC samples between low IL2RA and high IL2RA. Spearman correlation analysis was used to explore the correlations of IL2RA expression and immune checkpoint genes. A total of 747 DEGs were identified between low and high immune/stromal groups. Functional exploration revealed upregulated DEGs were associated with immune-related activities, whereas downregulated DEGs were involved in inflammatory-related activities. IL2RA was selected as the critical gene by overlapping the hub genes in PPI network and prognostic genes. Significantly, IL2RA expression was significantly elevated in PDAC and patients with higher IL2RA expression had worse prognoses. The immunological and oncogenic roles of IL2RA in PDAC were evidenced by GSEA. Furthermore, PDAC samples with high IL2RA expression exhibited increased immune infiltration and better immunotherapy responses. IL2RA expression was positively correlated with PDCD1, CD274, CTLA4, IDO1, TDO2, and TIGT. Higher expression of IL2RA predicts worse survival outcomes and increased immune infiltration in PDAC. PDAC patients with high IL2RA expression might potentially benefit from immunotherapy.
Collapse
Affiliation(s)
- Liwen Fan
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xinyu Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing Chang
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yue Wang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Wenjie Yang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Linlin Liu
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
- *Correspondence: Linlin Liu, Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China (e-mail: )
| |
Collapse
|