1
|
Jia J, Wang J, Zhang Y, Bai G, Han L, Niu Y. Deep Learning and Radiomic Signatures Associated with Tumor Immune Heterogeneity Predict Microvascular Invasion in Colon Cancer. Acad Radiol 2025:S1076-6332(25)00432-5. [PMID: 40413149 DOI: 10.1016/j.acra.2025.05.006] [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: 03/24/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/27/2025]
Abstract
RATIONALE AND OBJECTIVES This study aims to develop and validate a deep learning radiomics signature (DLRS) that integrates radiomics and deep learning features for the non-invasive prediction of microvascular invasion (MVI) in patients with colon cancer (CC). Furthermore, the study explores the potential association between DLRS and tumor immune heterogeneity. MATERIALS AND METHODS This study is a multi-center retrospective study that included a total of 1007 patients with colon cancer (CC) from three medical centers and The Cancer Genome Atlas (TCGA-COAD) database. Patients from Medical Centers 1 and 2 were divided into a training cohort (n = 592) and an internal validation cohort (n = 255) in a 7:3 ratio. Medical Center 3 (n = 135) and the TCGA-COAD database (n = 25) were used as external validation cohorts. Radiomics and deep learning features were extracted from contrast-enhanced venous-phase CT images. Feature selection was performed using machine learning algorithms, and three predictive models were developed: a radiomics model, a deep learning (DL) model, and a combined deep learning radiomics (DLR) model. The predictive performance of each model was evaluated using multiple metrics, including the area under the curve (AUC), sensitivity, and specificity. Additionally, differential gene expression analysis was conducted on RNA-seq data from the TCGA-COAD dataset to explore the association between the DLRS and tumor immune heterogeneity within the tumor microenvironment. RESULTS Compared to the standalone radiomics and deep learning models, DLR fusion model demonstrated superior predictive performance. The AUC for the internal validation cohort was 0.883 (95% CI: 0.828-0.937), while the AUC for the external validation cohort reached 0.855 (95% CI: 0.775-0.935). Furthermore, stratifying patients from the TCGA-COAD dataset into high-risk and low-risk groups based on the DLRS revealed significant differences in immune cell infiltration and immune checkpoint expression between the two groups (P < 0.05). CONCLUSION The contrast-enhanced CT-based DLR fusion model developed in this study effectively predicts the MVI status in patients with CC. This model serves as a non-invasive preoperative assessment tool and reveals a potential association between the DLRS and immune heterogeneity within the tumor microenvironment, providing insights to optimize individualized treatment strategies.
Collapse
Affiliation(s)
- Jianye Jia
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.)
| | - Jiahao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.)
| | - Yongxian Zhang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No.1 DongJiaoMinXiang Street, DongCheng District, Beijing 100730, China (Y.Z.)
| | - Genji Bai
- Department of Medical Imaging Center, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian 223300, Jiangsu, PR China (G.B.)
| | - Lei Han
- Department of Medical Imaging, Huaian Hospital Affiliated to Xuzhou Medical University, Huaian 223001, Jiangsu, China (L.H.)
| | - Yantao Niu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China (J.J., J.W., Y.N.).
| |
Collapse
|
2
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
3
|
Zhang Y, Wang H, Dai F, He K, Tuo Z, Wang J, Bi L, Chen X. A pan-cancer analysis of the oncogenic and immunological roles of RGS5 in clear cell renal cell carcinomas based on in vitro experiment validation. Hum Genomics 2025; 19:14. [PMID: 39985100 PMCID: PMC11846387 DOI: 10.1186/s40246-025-00717-w] [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: 09/19/2024] [Accepted: 01/15/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND RGS5, the first gene identified in tumor-resident pericytes, plays a crucial role in angiogenesis. However, its effects on immunology and prognosis in human cancer are still mostly unknown. This study investigates the carcinogenic and immunological roles of RGS5 through a comprehensive pan-cancer analysis. METHODS A standardized pan-cancer dataset for RGS5 was obtained from the public database. R software and relevant packages were utilized to analyze the oncogenic and immunological roles. Clinical samples and cellular experiments were conducted to validate RGS5 expression and its biological function in renal cancer. RESULTS Bioinformatics analysis revealed that RGS5 is dysregulated in a variety of human malignancies and is significantly associated with patient prognosis. Additionally, RGS5 expression is closely linked to tumor heterogeneity and stemness indicators across different cancer types. Co-expression of RGS5 with genes involved in MHC, immune activation, immunosuppressive proteins, chemokines, and chemokine receptors was observed in various tumors. High expression of RGS5 predicts a good prognosis in patients with renal cancer. In the renal cancer cohort, RGS5 expression strongly correlated with the distribution of tumor-associated fibroblasts. Silencing RGS5 expression can affect the proliferation, migration, and invasion of renal carcinoma cells. CONCLUSIONS RGS5 expression in tumors is intricately associated with various clinical features, particularly concerning tumor progression and patient prognosis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Huming Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Fang Dai
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Ke He
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, People's Republic of China
| | - Zhouting Tuo
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Jinyou Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China.
| | - Liangkuan Bi
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China.
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, People's Republic of China.
| | - Xin Chen
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China.
| |
Collapse
|
4
|
Xu K, Zhang H, Dai H, Mao W. Machine learning and multi-omics characterization of SLC2A1 as a prognostic factor in hepatocellular carcinoma: SLC2A1 is a prognostic factor in HCC. Gene 2025; 938:149178. [PMID: 39681148 DOI: 10.1016/j.gene.2024.149178] [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: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by high incidence, significant mortality, and marked heterogeneity, making accurate molecular subtyping essential for effective treatment. Using multi-omics data from HCC patients, we applied diverse clustering algorithms to identify three HCC subtypes (HSs) with distinct prognostic characteristics. Among these, HS1 emerged as an immune-compromised subtype associated with the poorest prognosis. Additionally, we developed a novel, robust, and highly accurate machine learning-guided prognostic signature (MLPS) by integrating multiple machine learning algorithms and their combinations. Our study also identified SLC2A1, the core gene of MLPS, as being highly expressed during advanced stages of tumor progression. Knockdown experiments demonstrated that reducing SLC2A1 expression significantly suppressed the malignant behavior of HCC cells. Furthermore, SLC2A1 expression was linked to responsiveness to dasatinib and vincristine, suggesting potential therapeutic relevance. MLPS and SLC2A1 offer promising tools for individualized prognosis prediction and targeted therapy in HCC, providing new opportunities to improve patient outcomes.
Collapse
Affiliation(s)
- Kangjie Xu
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Binhai County People's Hospital, Jiangsu Province, Yancheng 224000, PR China
| | - Houliang Zhang
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China
| | - Hua Dai
- Yangzhou University Clinical Medical College, Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Jiangsu Province, Yangzhou 225009, PR China.
| | - Weipu Mao
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Shan Y, He D, Yan F, Huang W. Understanding the tumor microenvironment for personalized immunotherapy in early-onset head and neck squamous cell carcinoma. Front Immunol 2025; 15:1522820. [PMID: 39830511 PMCID: PMC11739722 DOI: 10.3389/fimmu.2024.1522820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Early-onset head and neck squamous cell carcinoma (HNSCC) has been increasingly observed in recent years, exhibiting distinct tumor behavior and a unique tumor microenvironment (TME) compared to older age groups. Studies suggest that early-onset HNSCC is associated with specific risk factors and prognostic outcomes, while the underlying mechanisms driving these age-related differences remain unclear. In this review, we systematically examined original studies involving young HNSCC patient samples, focusing on the characteristics of the TME and potential for personalized immunotherapy. While further evidence is needed, our findings indicate that the TME in early-onset HNSCC often exhibits higher aggressiveness and immune suppression. Consequently, tailored immunotherapy may offer a promising therapeutic strategy for this distinct patient population.
Collapse
Affiliation(s)
| | | | | | - Weijia Huang
- Department of Oral & Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Wang J, Ying L, Xiong H, Zhou DR, Wang YX, Che HL, Zhong ZF, Wu GS, Ge YJ. Comprehensive analysis of stearoyl-coenzyme A desaturase in prostate adenocarcinoma: insights into gene expression, immune microenvironment and tumor progression. Front Immunol 2024; 15:1460915. [PMID: 39351232 PMCID: PMC11439642 DOI: 10.3389/fimmu.2024.1460915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate adenocarcinoma (PRAD) is a prevalent global malignancy which depends more on lipid metabolism for tumor progression compared to other cancer types. Although Stearoyl-coenzyme A desaturase (SCD) is documented to regulate lipid metabolism in multiple cancers, landscape analysis of its implications in PRAD are still missing at present. Here, we conducted an analysis of diverse cancer datasets revealing elevated SCD expression in the PRAD cohort at both mRNA and protein levels. Interestingly, the elevated expression was associated with SCD promoter hypermethylation and genetic alterations, notably the L134V mutation. Integration of comprehensive tumor immunological and genomic data revealed a robust positive correlation between SCD expression levels and the abundance of CD8+ T cells and macrophages. Further analyses identified significant associations between SCD expression and various immune markers in tumor microenvironment. Single-cell transcriptomic profiling unveiled differential SCD expression patterns across distinct cell types within the prostate tumor microenvironment. The Gene Ontology and Kyoto Encyclopedia of Genes and Genome analyses showed that SCD enriched pathways were primarily related to lipid biosynthesis, cholesterol biosynthesis, endoplasmic reticulum membrane functions, and various metabolic pathways. Gene Set Enrichment Analysis highlighted the involvement of elevated SCD expression in crucial cellular processes, including the cell cycle and biosynthesis of cofactors pathways. In functional studies, SCD overexpression promoted the proliferation, metastasis and invasion of prostate cancer cells, whereas downregulation inhibits these processes. This study provides comprehensive insights into the multifaceted roles of SCD in PRAD pathogenesis, underscoring its potential as both a therapeutic target and prognostic biomarker.
Collapse
Affiliation(s)
- Jie Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Liang Ying
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - He Xiong
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Duan-Rui Zhou
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yi-Xuan Wang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hui-Lian Che
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhang-Feng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Guo-Sheng Wu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yun-Jun Ge
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases,
Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Chen D, Xu L, Xuan M, Chu Q, Xue C. Unveiling the functional roles of patient-derived tumour organoids in assessing the tumour microenvironment and immunotherapy. Clin Transl Med 2024; 14:e1802. [PMID: 39245957 PMCID: PMC11381553 DOI: 10.1002/ctm2.1802] [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: 03/18/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/10/2024] Open
Abstract
Recent studies have established the pivotal roles of patient-derived tumour organoids (PDTOs), innovative three-dimensional (3D) culture systems, in various biological and medical applications. PDTOs, as promising tools, have been established and extensively used for drug screening, prediction of immune response and assessment of immunotherapeutic effectiveness in various cancer types, including glioma, ovarian cancer and so on. The overarching goal is to facilitate the translation of new therapeutic modalities to guide personalised immunotherapy. Notably, there has been a recent surge of interest in the co-culture of PDTOs with immune cells to investigate the dynamic interactions between tumour cells and immune microenvironment. A comprehensive and in-depth investigation is necessary to enhance our understanding of PDTOs as promising testing platforms for cancer immunotherapy. This review mainly focuses on the latest updates on the applications and challenges of PDTO-based methods in anti-cancer immune responses. We strive to provide a comprehensive understanding of the potential and prospects of PDTO-based technologies as next-generation strategies for advancing immunotherapy approaches.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjuan Xuan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingfei Chu
- Department of State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Li D, Shao F, Yu Q, Wu R, Tuo Z, Wang J, Ye L, Guo Y, Yoo KH, Ke M, Okoli UA, Premkamon C, Yang Y, Wei W, Heavey S, Cho WC, Feng D. The complex interplay of tumor-infiltrating cells in driving therapeutic resistance pathways. Cell Commun Signal 2024; 22:405. [PMID: 39160622 PMCID: PMC11331645 DOI: 10.1186/s12964-024-01776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Drug resistance remains a significant challenge in cancer treatment. Recently, the interactions among various cell types within the tumor microenvironment (TME) have deepened our understanding of the mechanisms behind treatment resistance. Therefore, this review aims to synthesize current research focusing on infiltrating cells and drug resistance suggesting that targeting the TME could be a viable strategy to combat this issue. Numerous factors, including inflammation, metabolism, senescence, hypoxia, and angiogenesis, contribute to drug resistance could be a viable strategy to combat this issue. Overexpression of STAT3 is commonly associated with drug-resistant cancer cells or stromal cells. Current research often generalizes the impact of stromal cells on resistance, lacking specificity and statistical robustness. Thus, future research should take notice of this issue and aim to provide high-quality evidence. Despite the existing limitations, targeting the TME to overcome therapy resistance hold promising and valuable potential.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315211, China
- Department of Pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, Republic of Korea
| | - Mang Ke
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Uzoamaka Adaobi Okoli
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
- Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chaipanichkul Premkamon
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, 404000, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Birmingham, Hong Kong SAR, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
9
|
Li JW, Mao YM, Chen SL, Ye R, Fei YR, Li Y, Tong SY, Yang HW, He YB. The interplay between metal ions and immune cells in glioma: pathways to immune escape. Discov Oncol 2024; 15:348. [PMID: 39134820 PMCID: PMC11319581 DOI: 10.1007/s12672-024-01229-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
This review explores the intricate roles of metal ions-iron, copper, zinc, and selenium-in glioma pathogenesis and immune evasion. Dysregulated metal ion metabolism significantly contributes to glioma progression by inducing oxidative stress, promoting angiogenesis, and modulating immune cell functions. Iron accumulation enhances oxidative DNA damage, copper activates hypoxia-inducible factors to stimulate angiogenesis, zinc influences cell proliferation and apoptosis, and selenium modulates the tumor microenvironment through its antioxidant properties. These metal ions also facilitate immune escape by upregulating immune checkpoints and secreting immunosuppressive cytokines. Targeting metal ion pathways with therapeutic strategies such as chelating agents and metalloproteinase inhibitors, particularly in combination with conventional treatments like chemotherapy and immunotherapy, shows promise in improving treatment efficacy and overcoming resistance. Future research should leverage advanced bioinformatics and integrative methodologies to deepen the understanding of metal ion-immune interactions, ultimately identifying novel biomarkers and therapeutic targets to enhance glioma management and patient outcomes.
Collapse
Affiliation(s)
- Jin-Wei Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi-Ming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Jiangsu Province, China
| | - Shi-Liang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Rui Ye
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi-Ran Fei
- The First Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang Province, China
| | - Yue Li
- The First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Shi-Yuan Tong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hong-Wei Yang
- Department of Clinical Laboratory, Suzhou BOE Hospital, Suzhou, Jiangsu Province, China.
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| |
Collapse
|
10
|
Song Y, Jiang S, Peng Y, Qin C, Du Y, Xu T. Effect of FGFR alteration on prognosis in 1963 urothelial carcinoma patients with immune checkpoint inhibitors: Implying combination of FGFR inhibitor and immunotherapy for FGFR-altered urothelial carcinoma. Pharmacol Res 2024; 205:107230. [PMID: 38788820 DOI: 10.1016/j.phrs.2024.107230] [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: 05/10/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are essential for urothelial carcinoma (UC) treatment. Fibroblast growth factor receptor (FGFR) alterations, as common oncogenic drivers in UC, have been reported to drive T cell depletion of UC immune microenvironment via up-regulating FGFR signaling, which indicated FGFR alterations potentially result in reduced response to ICIs. In addition, the selective pan-FGFR inhibitor showed better clinical benefit in clinical trials, indicating FGFR has emerged as critical therapeutic target via inhibiting FGFR signaling. The present study aims to evaluate prognosis and response to ICIs between FGFR-altered UC patients and FGFR-wildtype UC patients via 1963 UC patients and offers new insights into personalized precision therapy and combination therapy for UC.
Collapse
Affiliation(s)
- Yuxuan Song
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China.
| | - Shan Jiang
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Yun Peng
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing 100044, China; The Institute of Applied Lithotripsy Technology, Peking University, Beijing 100044, China.
| |
Collapse
|
11
|
Bai Z, Peng Y, Xia X, Li Y, Zhong Y, Chen L, Guan Q, Liu W, Zhou Y, Ma L. Inhibiting autophagy enhanced mitotic catastrophe-mediated anticancer immune responses by regulating the cGAS-STING pathway. Cancer Lett 2024; 586:216695. [PMID: 38325769 DOI: 10.1016/j.canlet.2024.216695] [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: 10/11/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
Given the limitations of the response rate and efficacy of immune checkpoint inhibitors (ICIs) in clinical applications, exploring new therapeutic strategies for cancer immunotherapy is necessary. We found that 5-(3,4,5-trimethoxybenzoyl)-4-methyl-2-(p-tolyl)imidazole (BZML), a microtubule-targeting agent, exhibited potent anticancer activity by inducing mitotic catastrophe in A549/Taxol and L929 cells. Nuclear membrane disruption and nuclease reduction provided favorable conditions for cGAS-STING pathway activation in cells with mitotic catastrophe. Similar results were obtained in paclitaxel-, docetaxel- and doxorubicin-induced mitotic catastrophe in various cancer cells. Notably, the surface localization of CALR and MHC-I and the release of HMGB1 were also significantly increased in cells with mitotic catastrophe, but not in apoptotic cells, suggesting that mitotic catastrophe is an immunogenic cell death. Furthermore, activated CD8+T cells enhanced the anticancer effects originating from mitotic catastrophe induced by BZML. Inhibiting the cGAS-STING pathway failed to affect BZML-induced mitotic catastrophe but could inhibit mitotic catastrophe-mediated anticancer immune effects. Interestingly, the expression of p-TBK1 first increased and then declined; however, autophagy inhibition reversed the decrease in p-TBK1 expression and enhanced mitotic catastrophe-mediated anticancer immune effects. Collectively, the inhibition of autophagy can potentiate mitotic catastrophe-mediated anticancer immune effects by regulating the cGAS-STING pathway, which explains why the anticancer immune effects induced by chemotherapeutics have not fully exerted their therapeutic efficacy in some patients and opens a new area of research in cancer immunotherapy.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China.
| | - Yaling Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Xue'er Xia
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yupeng Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yuejiao Zhong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Lingxiang Chen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Yiran Zhou
- Department of General Surgery, Ruijin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China.
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|