1
|
Osborne N, Rupani A, Makarov V, Chan TA, Srivastava RM. Avelumab induces greater Fc-Fc receptor-dependent natural killer cell activation and dendritic cell crosstalk compared to durvalumab. Oncoimmunology 2025; 14:2494995. [PMID: 40311014 PMCID: PMC12051578 DOI: 10.1080/2162402x.2025.2494995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Several FDA-approved anti-PD-L1 (programmed cell death ligand-1) monoclonal antibodies (mAbs) are used to treat cancer. While these mAbs primarily target and intercept PD-L1:PD-1 inhibitory signaling in T-cells, the Fc-domains of these mAbs are distinct, and the unique cellular cascades triggered by differing Fc-domains of PD-L1 mAbs have not been directly investigated. In this study, we compared the innate immune effects of two widely used anti-PD-L1 IgG1 mAbs which bear distinct Fc-domains, avelumab (native-Fc) and durvalumab (mutated-Fc), using two-cell and three-cell co-culture systems containing Natural Killer cells (NK-cells), dendritic cells (DCs) and various tumor cell lines of multiple cancer origins. We show a robust enhancement in NK-cell effector function, DC maturation, reciprocal NK:DC crosstalk and DC editing that is unique to avelumab treatment using multiple functional immune assays. By transcriptomic analysis, we show for the first time pivotal differences in gene sets involved in NK-cell effector function, DC maturation, immunoregulatory interactions, and cytokine production between innate immune cells treated with avelumab versus durvalumab. Furthermore, we report several previously unknown Fc-receptor-associated biological pathways uniquely triggered by avelumab. Our findings elucidate novel mechanisms of Fc-dependent actions of PD-L1 mAbs which may inform their use in future clinical trials.
Collapse
MESH Headings
- Humans
- Dendritic Cells/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Receptors, Fc/metabolism
- Receptors, Fc/immunology
- Cell Line, Tumor
- Coculture Techniques
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Antibodies, Monoclonal/pharmacology
- Cell Communication/drug effects
- Cell Communication/immunology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- Neoplasms/immunology
- Neoplasms/drug therapy
- Antineoplastic Agents, Immunological/pharmacology
Collapse
Affiliation(s)
- Nicole Osborne
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amit Rupani
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A. Chan
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Raghvendra M. Srivastava
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
2
|
Luo R, Liu J, Wang T, Zhao W, Wang Y, Wen J, Wang H, Ding S, Zhou X. The landscape of malignant transition: Unraveling cancer cell-of-origin and heterogeneous tissue microenvironment. Cancer Lett 2025; 621:217591. [PMID: 40054660 PMCID: PMC12040592 DOI: 10.1016/j.canlet.2025.217591] [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: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/12/2025]
Abstract
Understanding disease progression and sophisticated tumor ecosystems is imperative for investigating tumorigenesis mechanisms and developing novel prevention strategies. Here, we dissected heterogeneous microenvironments during malignant transitions by leveraging data from 1396 samples spanning 13 major tissues. Within transitional stem-like subpopulations highly enriched in precancers and cancers, we identified 30 recurring cellular states strongly linked to malignancy, including hypoxia and epithelial senescence, revealing a high degree of plasticity in epithelial stem cells. By characterizing dynamics in stem-cell crosstalk with the microenvironment along the pseudotime axis, we found differential roles of ANXA1 at different stages of tumor development. In precancerous stages, reduced ANXA1 levels promoted monocyte differentiation toward M1 macrophages and inflammatory responses, whereas during malignant progression, upregulated ANXA1 fostered M2 macrophage polarization and cancer-associated fibroblast transformation by increasing TGF-β production. Our spatiotemporal analysis further provided insights into mechanisms responsible for immunosuppression and a potential target to control evolution of precancer and mitigate the risk for cancer development.
Collapse
Affiliation(s)
- Ruihan Luo
- Laboratory of Hepatic AI Translation, Frontier Science Center for Disease-Related Molecular Network and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China.
| | - Jiajia Liu
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tiangang Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Weiling Zhao
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanfei Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jianguo Wen
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hongyu Wang
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; Center for Nursing Research, Cizik School of Nursing, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Shanli Ding
- Graduate School of Biomedical Sciences, The University of MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA; McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Xie J, Liu W, Deng X, Wang H, Ou X, An X, Situ MY, Yang A, Peng C, He R, Xie Y, Tang H, Chen Y, Liang JY, Shao R, Xiao W, Zheng S. Paracrine Orchestration of Tumor Microenvironment Remodeling Induced by GLO1 Potentiates Lymph Node Metastasis in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e00722. [PMID: 40492378 DOI: 10.1002/advs.202500722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/24/2025] [Indexed: 06/12/2025]
Abstract
Breast cancer is the most prevalent form of malignant tumor that frequently metastasizes to axillary lymph nodes (LNs). Nonetheless, the precise mechanisms underlying alterations in the tumor microenvironment (TME) in LN metastasis in breast cancer remain poorly understood. Single-cell RNA sequencing of 28 LN samples from 23 patients is performed, and a comprehensive landscape of the entire ecosystem is generated. Ten major cell types are identified, with the subclusters of each major cell type exhibiting diverse characteristics. Furthermore, multiple signatures are collected to evaluate the key components of the subclusters using multi-omics methodologies. This study finds that myCAFs may hasten LN metastasis, and observed a notable increase in APOE+ macrophages and a higher proportion of exhausted CD8+ T cells, contributing to the immunosuppressive TME. Moreover, cancer cells in metastatic lesions exhibited diverse expression patterns linked to proliferation, metastasis, oxidative phosphorylation, hypoxia, and interferon responses. Using multi-omics approaches and experimental validations, it determines that GLO1 can promote lymphatic angiogenesis, metastasis, and inhibit the proteasomal degradation of GSS, thereby maintaining intracellular glutathione (GSH) and reactive oxygen species (ROS) balance. Collectively, the study offers novel perspectives on the microenvironment remodeling of breast cancer LN metastases, suggesting that GLO1 may be a promising therapeutic target.
Collapse
Affiliation(s)
- Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wenjian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Huan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xueqi Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xin An
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Min-Yi Situ
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Anli Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chuan Peng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rongfang He
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yi Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yuman Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jie-Ying Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ruonan Shao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weikai Xiao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaoquan Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Breast Surgery, Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Zheng Y, He D, Zuo W, Wang W, Wu K, Wu H, Yuan Y, Huang Y, Li H, Lu Y, Zhao L, Wang X, Wang J, Zhang Y, Zou G, Li H, Wang Z, Cao B. Influenza A virus dissemination and infection leads to tissue resident cell injury and dysfunction in viral sepsis. EBioMedicine 2025; 116:105738. [PMID: 40367638 PMCID: PMC12142562 DOI: 10.1016/j.ebiom.2025.105738] [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: 08/20/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Severe respiratory viral infections can lead to viral sepsis (VS), a life-threatening condition characterized by lung and extrapulmonary organ dysfunction. However, the pathology of VS is not clear. Specifically, it is unknown how the cytokine storm and direct virus infection contribute to the damage of extrapulmonary organs. METHODS In this study, we established survival and lethal mouse models of VS by intranasally administering different doses of PR8/H1N1 influenza virus in C57BL/6J male mice, as well as model of bacterial sepsis (BS) caused by Streptococcus pneumoniae as references. Viraemia and extrapulmonary dissemination and infection of the virus were examined. Single-cell sequencing of the lungs and livers was performed at different days post-infection (dpi) in three groups. FINDINGS While bacteria can spread and colonize extensively in extrapulmonary organs, causing multiple organ injuries, IAVs mainly replicate and cause damage in pulmonary cells. Live virus can be isolated in the blood and extrapulmonary organs. Disseminating via the bloodstream, IAVs transiently infect the liver and spleen, causing liver dysfunction and spleen atrophy, without affecting kidney function, despite systematically elevated cytokine levels. Compared to BS, a more significant decrease in the proportion of alveolar macrophages, epithelial cells, endothelial cells, and fibroblasts in the lungs, as well as endothelial cells and Kupffer cells in the liver, was observed in VS. This was accompanied by a longer activated PANoptosis pathway and downregulated genes responsible for barrier function and antigen presentation in the epithelial and endothelial cells. INTERPRETATION Our study suggests that H1N1 influenza virus disseminates through the bloodstream and infects extrapulmonary organs to varying extents, which may lead to differential cell death, organ dysfunction, and trigger VS. FUNDING This research was supported by the National Natural Science Foundation of China (82241056, 82170015, 82030002, 82470007, 824B2001), the National Key R&D Program of China (2023YFC2306300), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2021-I2M-1-048), the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (ZYYCXTD-D-202208), New Cornerstone Science Foundation, National High Level Hospital Clinical Research Funding (2024-NHLHCRF-LX-01-0101, 2024-NHLHCRF-LX-01-0102), Beijing Research Ward Excellence Program (BRWEP2024W114060103), Noncommunicable Chronic Diseases-National Science and Technology Major Project (2023ZD0506200, 2023ZD0506203) and Special Research Fund for Central Universities, Peking Union Medical College (3332024193).
Collapse
Affiliation(s)
- Ying Zheng
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Di He
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wenting Zuo
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weiyang Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Kaiwei Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongping Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yingying Yuan
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100083, China
| | - Yijiao Huang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, 100084, China
| | - Hongyan Li
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yameng Lu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China
| | - Ling Zhao
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiuhong Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiaying Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yulian Zhang
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Guming Zou
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Haibo Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; New Cornerstone Science Laboratory, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Zai Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100083, China; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing, 100054, China; National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China; Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, 100084, China; New Cornerstone Science Laboratory, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
5
|
Kleberg J, Nataraj A, Xiao Y, Podder BR, Jin Z, Tithi TI, Zheng G, Smalley KSM, Moser EK, Safe S, Maharjan CK, Kolb R, Zhang W. Targeting Lineage-Specific Functions of NR4A1 for Cancer Immunotherapy. Int J Mol Sci 2025; 26:5266. [PMID: 40508074 PMCID: PMC12155124 DOI: 10.3390/ijms26115266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/18/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Orphan nuclear receptor 4A1 (NR4A1, Nur77) plays a crucial role in regulating immune cell metabolism and function within the tumor microenvironment (TME), thus influencing cancer progression and serving as a potential therapeutic target for cancer immunotherapy. A comprehensive review discussing the multifaceted roles of NR4A1 in immune cells and the exploitation of that knowledge for therapeutic development is lacking in the field. This review explores diverse functions of NR4A1 in tumor-associated immune cells, including T cells, monocytes, natural killer cells, B cells, dendritic cells, macrophages, and neutrophils. NR4A1 contributes to immune regulation by impacting cytokine production, cell differentiation, and immune cell exhaustion. We highlight how NR4A1 in immune cells within the TME may be either a positive (e.g., macrophages in colon cancer) or negative prognostic factor (e.g., T cells in melanoma), depending on the cancer and immune cell context. Additionally, this review also highlights potential therapeutic strategies targeting NR4A1, leading to its inhibition, activation, or degradation to restore immune cell function and enhance anti-tumor immunity. Such therapies could potentially improve patient outcomes by altering immune cell behaviors, blocking intrinsic tumor growth pathways, or via both mechanisms. However, the development of NR4A1-targeted therapies will be dependent on further research to better understand lineage-specific roles of NR4A1 and the underlying mechanisms across different cancer types and immune cells.
Collapse
Affiliation(s)
- Jeremy Kleberg
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
- Department of Biochemistry and Molecular Biology, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Akhila Nataraj
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
- Department of Health Science, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
| | - Yufeng Xiao
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (Y.X.); (G.Z.)
| | - Bristy R. Podder
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
| | - Tanzia Islam Tithi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; (Y.X.); (G.Z.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Keiran S. M. Smalley
- Department of Tumor Microenvironment and Metastasis, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Emily K. Moser
- Department of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Stephen Safe
- Molecular & Cellular Oncology Laboratory, Physiology and Pharmacology and of Biochemistry and Biophysics, Texas A&M University Veterinary Medicine & Biomedical Sciences, College Station, TX 77843, USA;
| | - Chandra K. Maharjan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (J.K.); (A.N.); (B.R.P.); (Z.J.); (T.I.T.); (R.K.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
6
|
Liu Z, Yang Z, Wu J, Zhang W, Sun Y, Zhang C, Bai G, Yang L, Fan H, Chen Y, Zhang L, Jiang B, Liu X, Ma X, Tang W, Liu C, Qu Y, Yan L, Zhao D, Wu Y, He S, Xu L, Peng L, Chen X, Zhou B, Zhao L, Zhao Z, Tan F, Zhang W, Yi D, Li X, Gao Q, Zhang G, Wang Y, Yang M, Fu H, Guo Y, Hu X, Cai Q, Qi L, Bo Y, Peng H, Tian Z, She Y, Zou C, Zhu L, Cheng S, Zhang Y, Zhong W, Chen C, Gao S, Zhang Z. A single-cell atlas reveals immune heterogeneity in anti-PD-1-treated non-small cell lung cancer. Cell 2025; 188:3081-3096.e19. [PMID: 40147443 DOI: 10.1016/j.cell.2025.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/20/2024] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Anti-PD-(L)1 treatment is standard for non-small cell lung cancer (NSCLC), but patients show variable responses to the same regimen. The tumor immune microenvironment (TIME) is associated with immunotherapy response, yet the heterogeneous underlying therapeutic outcomes remain underexplored. We applied single-cell RNA and TCR sequencing (scRNA/TCR-seq) to analyze surgical tumor samples from 234 NSCLC patients post-neoadjuvant chemo-immunotherapy. Analyses revealed five distinct TIME subtypes with varying major pathological response (MPR) rates. MPR patients had elevated levels of FGFBP2+ NK/NK-like T cells, memory B cells, or effector T cells, while non-MPR patients showed higher CCR8+ Tregs. T cell clonal expansion analyses unveiled heterogeneity in non-MPR patients, marked by varying expansions of Tex-relevant cells and CCR8+ Tregs. Precursor exhausted T cells (Texp cells) correlated with recurrence-free survival, identifying a patient subgroup with reduced recurrence risk despite lack of MPR. Our study dissects TIME heterogeneity in response to chemoimmunotherapy, offering insights for NSCLC management.
Collapse
Affiliation(s)
- Zedao Liu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Wenjie Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuxuan Sun
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Guangyu Bai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hongtao Fan
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yawen Chen
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Benyuan Jiang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaoyan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaoshi Ma
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University), Shenzhen 518020, China
| | - Wei Tang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chang Liu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Yang Qu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Yilong Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Shun He
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lishan Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiaowei Chen
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhangyi Zhao
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wanting Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dingcheng Yi
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | | | - Qianqian Gao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yongjie Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo 315010, China
| | - Honghao Fu
- Department of General Thoracic Surgery, Jining First People's Hospital, Affiliated Hospital of Shandong First Medical University, Jining 272000, China
| | - Yongjun Guo
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xueda Hu
- Analytical Biosciences Limited, Beijing, China
| | - Qingyuan Cai
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Lu Qi
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China; Changping Laboratory, Beijing 102206, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Zhigang Tian
- National Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Chang Zou
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China; Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College of Jinan University), Shenzhen 518020, China.
| | - Linnan Zhu
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Sijin Cheng
- Changping Laboratory, Beijing 102206, China; Chongqing Medical University, Chongqing, China.
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Zhongyuan Cell Therapy and Immunotherapy Laboratory, Zhengzhou 450000, China.
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing 100871, China; Chongqing Medical University, Chongqing, China.
| |
Collapse
|
7
|
Shang Y, Pang Y, Liu T, Wang W. Application of mass cytometry in the immune microenvironment of breast cancer. Med Oncol 2025; 42:215. [PMID: 40388018 DOI: 10.1007/s12032-025-02770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
The rapid development of immunotherapy has shown preliminary clinical efficacy and significant anti-tumor effects in some cancer patients. Although immunotherapy has been approved for breast cancer, some breast cancer patients still do not benefit from it due to issues such as immunotherapy insensitivity and resistance. Mass cytometry, as a mature single-cell proteomic analysis method, with its high-throughput capabilities, has been widely used in the analysis of tumor immune microenvironments and immune cell subpopulations. Using mass cytometry to analyze the immune microenvironment of breast cancer and explore new immunotherapy targets can help improve the current status of breast cancer immunotherapy and develop personalized treatment plans for more patients. This review surveys the recent advancements in analyzing the single-cell components of breast cancer using mass cytometry technology and reviews the immune microenvironment of breast cancer as well as potential targets for immunotherapy. These results provide new insights for the subsequent research of the immune microenvironment of breast cancer and targeted immunotherapy.
Collapse
Affiliation(s)
- Yuefeng Shang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Yuheng Pang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tong Liu
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen Wai, Fengtai District, Beijing, 100069, People's Republic of China.
| |
Collapse
|
8
|
Razmara AM, Lammers M, Judge SJ, Murphy WJ, Gaskill CE, Culp WT, Gingrich AA, Morris ZS, Rebhun RB, Brown CT, Vail DM, Kent MS, Canter RJ. Single cell atlas of canine natural killer cells identifies distinct circulating and tissue resident gene profiles. Front Immunol 2025; 16:1571085. [PMID: 40443661 PMCID: PMC12119461 DOI: 10.3389/fimmu.2025.1571085] [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: 02/04/2025] [Accepted: 04/22/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Natural killer (NK) cells in mice and humans are key effectors of the innate immune system with complex immunoregulatory functions, and diverse subsets have been identified with distinct characteristics and roles. Companion dogs with spontaneous cancer have been validated as models of human disease, including cancer immunology and immunotherapy, and greater understanding of NK cell heterogeneity in dogs can inform NK biology across species and optimize NK immunotherapy for both dogs and people. Methods Here, we assessed canine NK cell populations by single-cell RNA sequencing (scRNAseq) across blood, lung, liver, spleen, and placenta with comparison to human NK cells from blood and the same tissues to better characterize the differential gene expression of canine and human NK cells regarding ontogeny, heterogeneity, patterns of activation, inhibition, and tissue residence. Results Overall, we observed tissue-specific NK cell signatures consistent with immature NK cells in the placenta, mature and activated NK cells in the lung, and NK cells with a mixed activated and inhibited signature in the liver with significant cross-species homology. Discussion Together, our results point to heterogeneous canine NK populations highly comparable to human NK cells, and we provide a comprehensive atlas of canine NK cells across organs which will inform future cross-species NK studies and further substantiate the spontaneous canine model to optimize NK immunotherapy across species.
Collapse
Affiliation(s)
- Aryana M. Razmara
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Marshall Lammers
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sean J. Judge
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Cameron E. Gaskill
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - William T.N. Culp
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - Alicia A. Gingrich
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Robert B. Rebhun
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - C. Titus Brown
- Department Population Health and Reproduction, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - David M. Vail
- Department of Medical Sciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States
| | - Michael S. Kent
- Department of Surgical and Radiological Sciences, University of California Davis School of Veterinary Medicine, Davis, CA, United States
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
9
|
Shorer O, Pinhasi A, Yizhak K. Single-cell meta-analysis of T cells reveals clonal dynamics of response to checkpoint immunotherapy. CELL GENOMICS 2025; 5:100842. [PMID: 40187353 DOI: 10.1016/j.xgen.2025.100842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/03/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
Despite the crucial role of T cell clones in anti-tumor activity, their characterization and association with clinical outcomes following immune checkpoint inhibitors are lacking. Here, we analyzed paired single-cell RNA sequencing/T cell receptor sequencing of 767,606 T cells across 460 samples spanning 6 cancer types. We found a robust signature of response based on expanded CD8+ clones that differentiates responders from non-responders. Analysis of persistent clones showed transcriptional changes that are differentially induced by therapy in the different response groups, suggesting an improved reinvigoration capacity in responding patients. Moreover, a gene trajectory analysis revealed changes in the pseudo-temporal state of de novo clones that are associated with clinical outcomes. Lastly, we found that clones shared between tumor and blood are more abundant in non-responders and execute distinct transcriptional programs. Overall, our results highlight differences in clonal transcriptional states that are linked to patient response, offering valuable insights into the mechanisms driving effective anti-tumor immunity.
Collapse
Affiliation(s)
- Ofir Shorer
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Asaf Pinhasi
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Keren Yizhak
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel; The Taub Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
10
|
Tyler M, Gavish A, Barbolin C, Tschernichovsky R, Hoefflin R, Mints M, Puram SV, Tirosh I. The Curated Cancer Cell Atlas provides a comprehensive characterization of tumors at single-cell resolution. NATURE CANCER 2025:10.1038/s43018-025-00957-8. [PMID: 40341230 DOI: 10.1038/s43018-025-00957-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/24/2025] [Indexed: 05/10/2025]
Abstract
Recent years have seen a rapid proliferation of single-cell cancer studies, yet most of these studies profiled few tumors, limiting their statistical power. Combining data and results across studies holds great promise but also involves various challenges. We recently began to address these challenges by curating a large collection of cancer single-cell RNA-sequencing datasets, leveraging it for systematic analyses of tumor heterogeneity. Here we greatly extend this repository to 124 datasets for over 40 cancer types, together comprising 2,836 samples, with improved data annotations, visualizations and exploration. Using this vast cohort, we generate an updated map of recurrent expression programs in malignant cells and systematically quantify context-dependent gene expression and cell-cycle patterns across cell types and cancer types. These data, annotations and analysis results are all freely available for exploration and download through the Curated Cancer Cell Atlas, a central community resource that opens new avenues in cancer research.
Collapse
Affiliation(s)
- Michael Tyler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.
| | - Avishai Gavish
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Chaya Barbolin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Roi Tschernichovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Mints
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Sidharth V Puram
- Department of Otolaryngology-Head and Neck Surgery and Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- The Robert Ebert and Greg Stubblefield Head and Neck Tumor Center at Siteman Cancer Center, St. Louis, MO, USA
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
11
|
Guo H, Hu J, Wang Z, Xu F, Liu Y, Cui L, Zhang H, Xie C, Yao R, Jin H, Guo Z, Wang T, Li L, Lin Y, Wang X, Li H, Xia X. RAC2 inhibition enhances tumor sensitivity to NK cell-mediated cytotoxicity. J Immunother Cancer 2025; 13:e010931. [PMID: 40316303 PMCID: PMC12049936 DOI: 10.1136/jitc-2024-010931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/14/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Natural killer (NK) cells are recognized for their ability to kill tumor cells for tumor control, but tumor cells often develop resistance to evade NK cell-mediated cytotoxicity. Identification of molecular mechanisms by which tumor cells evade from NK cell-mediated killing may offer novel therapeutic strategies for potentiating NK-based cancer immunotherapy. METHODS An in vitro tumor-NK cell co-culture system was employed to identify the most significantly altered genes in tumor cells following NK cell interaction. The cell death rate of tumor cells by NK cell exposure was quantified using flow cytometry. EL4 and HCT116 tumor models in C57BL/6, BALB/c-nu, and NOD/SCID mice were used for evaluating tumor growth differences induced by Rac2 knockdown or knockout. The cellular and molecular impact of Rac2 knockdown or knockout on the sensitivity of tumor cells to NK cell-mediated cytotoxicity was assessed using quantitative PCR, immunofluorescence, and mutation analysis. RESULTS By screening expression levels of the Ras homology (Rho) GTPase family genes in tumor cells after co-culture with NK cells, we identified RAC2 as a key regulator of tumor cell resistance to NK cell-mediated cytotoxicity among the Rho GTPase family members. Furthermore, knockout of RAC2 in human colorectal cancer cells leads to increased tumor susceptibility to NK cell-mediated cytotoxicity in a xenograft tumor model. Mechanistically, the absence of RAC2 enhances tumor cell sensitivity to NK cell-mediated killing by facilitating cell-cell contact. CONCLUSIONS These findings indicate that the inhibition of RAC2 in tumor cells substantially enhances their susceptibility to NK cell-mediated cytotoxicity, thereby providing a potential therapeutic target for optimizing NK cell therapy.
Collapse
Affiliation(s)
- Hui Guo
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jie Hu
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zining Wang
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Feifei Xu
- Department of Oncology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yongxiang Liu
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Lei Cui
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Huanling Zhang
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Chunyuan Xie
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ruhui Yao
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Huan Jin
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zixuan Guo
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tiantian Wang
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lin Li
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yanxun Lin
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiaojuan Wang
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Heping Li
- Department of Medical Oncology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Xiaojun Xia
- Department of Experiment Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
12
|
Jiang Q, Yu W, Ma J, Zhao M, Zou J, Mir S, Zhang J, Germain RN, Hassan R. Robust differentiation of NK cells from MSLN.CAR-IL-15-engineered human iPSCs with enhanced antitumor efficacy against solid tumors. SCIENCE ADVANCES 2025; 11:eadt9932. [PMID: 40315330 PMCID: PMC12047432 DOI: 10.1126/sciadv.adt9932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Human induced pluripotent stem cells (iPSCs) offer a promising source for chimeric antigen receptor (CAR)-engineered natural killer (NK) products. However, complex iPSC-NK (iNK) manufacturing challenges clinical use. Here, we identified LiPSC-GR1.1 as a superior iPSC line for iNK production. By engineering LiPSC-GR1.1 with a mesothelin (MSLN)-targeting CAR and interleukin-15 (IL-15), we achieved robust differentiation of iPSCs into mature activated iNK cells with enhanced tumor killing efficacy, superior tumor homing, and vigorous proliferation. Single-cell transcriptomic analysis revealed that transforming growth factor-β (TGF-β)-producing tumor cells up-regulated major histocompatibility complex molecules and down-regulated MSLN post-CAR-IL-15 iNK treatment. Tumor-infiltrating CAR-IL-15 iNK cells exhibited high levels of CAR, IL-15, and NK-activating receptors, negligible checkpoint exhaustion markers, and extremely low levels of NK suppressive factors CISH, TGFBR2, and BATF, enabling them to sustain activation, metabolic fitness, and effective tumor killing within TGF-β-rich hypoxic tumor microenvironment. Overall, we developed MSLN.CAR-IL-15-engineered GR1.1-iNK therapy with enhanced antitumor efficacy for solid tumor treatment.
Collapse
Affiliation(s)
- Qun Jiang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Weiming Yu
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging, NIAID and NCI, NIH, Bethesda, MD, USA
| | - James Ma
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mingming Zhao
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Sameer Mir
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jingli Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging, NIAID and NCI, NIH, Bethesda, MD, USA
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
13
|
Qu WF, Zhu GQ, Yang R, Chu TH, Guan ZQ, Huang R, Tian MX, Jiang XF, Tao CY, Fang Y, Gao J, Wu XL, Chen JF, Zhao QF, Wang Y, Bu YC, Zhou J, Fan J, Liu WR, Tang Z, Shi YH. Targeting HMGB2 acts as dual immunomodulator by bolstering CD8 + T cell function and inhibiting tumor growth in hepatocellular carcinoma. SCIENCE ADVANCES 2025; 11:eads8597. [PMID: 40315321 PMCID: PMC12047442 DOI: 10.1126/sciadv.ads8597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/27/2025] [Indexed: 05/04/2025]
Abstract
T cell exhaustion is a critical obstacle for durable treatment response in hepatocellular carcinoma (HCC). Developing drugs that control tumor growth and simultaneously bolster immune function is of great significance. Although high-mobility group box 2 (HMGB2) has been reported to be crucial to HCC prognosis, its role in the tumor microenvironment remains unclear. Here, we found HMGB2+ CD8+ T cells as being associated with immune exhaustion and resistance to anti-PD-1 treatment through single-cell RNA sequencing. Mechanistically, HMGB2 impaired the oxidative phosphorylation in CD8+ T cells and inactivated the interferon-γ response in tumor cells, reducing the antitumor effector function. Tannic acid, a specific inhibitor of HMGB2, synergized with PD-1 antibody to attenuate tumor growth and reverse T cell exhaustion. Our findings highlight the unique role of HMGB2 as an immune exhaustion associated molecule. Targeting HMGB2 on both CD8+ T cells and tumor cells contributed to promising treatment strategies for HCC.
Collapse
Affiliation(s)
- Wei-Feng Qu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
- Department of Thyroid and Breast Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gui-Qi Zhu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Yang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Tian-Hao Chu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Qi Guan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Run Huang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
- Department of Thyroid and Breast Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Xin Tian
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi-Fei Jiang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen-Yang Tao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Fang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Gao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Ling Wu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia-Feng Chen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian-Fu Zhao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Wang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Chao Bu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia Fan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei-Ren Liu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Zheng Tang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying-Hong Shi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Song Z, Wang Y, Zhu M, Zhang P, Li Z, Geng X, Cao X, Zheng J, Tang J, Chen L. Exploring ribosome biogenesis in lung adenocarcinoma to advance prognostic methods and immunotherapy strategies. J Transl Med 2025; 23:503. [PMID: 40316986 PMCID: PMC12048935 DOI: 10.1186/s12967-025-06489-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/13/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) presents a considerable danger to human health and has evolved into a major public health concern. Ribosome biogenesis (RiboSis) is a critical process for synthesizing ribosomes, closely associated with cancer initiation, progression, and treatment resistance, potentially serving as a target for future cancer therapies. METHODS Utilizing single-cell RNA sequencing (scRNA-seq) technology, a single-cell atlas of LUAD was delineated, focusing on the analysis of T cell subpopulations. Cells were scored based on the expression patterns of 331 genes associated with RiboSis across different cell types, and monocle2 was employed to analyze the developmental trajectory of CD4+ T cells. Employing various machine learning algorithms, a ribosome biogenesis-related signature (RBS) was constructed and compared to 140 published LUAD prognostic models. The relationship between RBS risk scores and various factors in LUAD patients, including prognosis, the tumor immune microenvironment (TIME), responsiveness to immunotherapy, and sensitivity to pharmacological treatments was specifically analyzed. Immunohistochemistry was utilized to validate the expression levels of immune markers in the high- and low- RBS groups, and in vitro experiments were performed to validate the functional role of the pivotal gene KIF23 in the progression of LUAD. RESULTS Using single-cell analysis, two distinct T cell subtypes were identified: CD8+ interferon (IFN) response T cells and CD4+ stress response T cells. It was observed that CD4+ naive-like T cells exhibit high expression of RiboSis-related genes, with a gradual decrease in RiboSis activity as CD4+ T cells develop. Compared to other prognostic models, RBS demonstrated superior performance in prognosis prediction. The low-RBS group exhibited a tumor microenvironment (TME) more favorable for efficient immune monitoring and reaction, higher responsiveness to immunotherapy, and a better prognosis. Immunohistochemistry confirmed higher expression levels of immune markers in the low-RBS group, while in vitro experiments validated the promoting role of KIF23 in LUAD cell proliferation, migration and invasion. CONCLUSION This study delves into the relationship between RiboSis and LUAD cell subpopulations, identifying a potent prognostic biomarker for LUAD. This biomarker aids in assessing immunotherapy efficacy in LUAD patients, ultimately enhancing their prognosis and guiding clinical decision-making.
Collapse
Affiliation(s)
- Zipei Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuheng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Miaolin Zhu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhihua Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Geng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xincen Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianan Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jianwei Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Liu S, Sun H, Song T, Liang C, Deng L, Zhu H, Zhao F, Li S. Comprehensive characterization of T cell subtypes in lung adenocarcinoma: Prognostic, predictive, and therapeutic implications. Transl Oncol 2025; 55:102332. [PMID: 40184717 PMCID: PMC12002896 DOI: 10.1016/j.tranon.2025.102332] [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: 11/25/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND T cells are crucial for immunosurveillance and tumor eradication, with their dysregulation or absence in the tumor microenvironment linked to immunotherapy resistance. In lung adenocarcinoma (LUAD), this resistance is a significant barrier to effective treatment, highlighting the need for robust biomarkers and therapeutic targets to improve clinical outcomes. METHODS T cell-related markers were identified through single-cell RNA sequencing analysis. The TCGA dataset was used for consensus clustering to define molecular subtypes associated with distinct survival outcomes and immune profiles. A T cell-related prognostic signature was developed by integrating LUAD datasets from TCGA, GSE31210, GSE50081, and GSE68465 using 10 machine learning algorithms. Further analysis linked risk scores to immune infiltration and drug sensitivity. The role of a hub gene in CD4+ T cell function and its involvement in tumor immunity was explored through in vitro experiments and molecular biology techniques. RESULTS Cluster analysis identified three LUAD subtypes, with cluster1 showing the best prognosis and immune characteristics. A Lasso + PLSRcox-based signature was a significant risk factor for predicting LUAD patient outcomes, outperforming traditional clinicopathological factors. The risk score correlated with immune microenvironment features, immune cell infiltration, and sensitivity to immunotherapy and chemotherapy. CPA3 expression was elevated in activated CD4+ T cells, particularly in Th1 cells, promoting differentiation and IFN-γ secretion. Overexpression of CPA3 enhanced tumor cell apoptosis and increased Granzyme B and IFN-γ levels, highlighting its role in immune responses. CONCLUSION We developed a powerful prognostic signature in LUAD that accurately predicts clinical outcomes and can guide immunotherapy and chemotherapy responses.
Collapse
Affiliation(s)
- Shiquan Liu
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Thoracic Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Hao Sun
- Xinqiao Hospital, Army Military Medical University, Chongqing, China; Faculty of Science, Autonomous University of Madrid, Spainish National Research Council (UAM-CSIC), Madrid, Spain
| | - Tianye Song
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ce Liang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lele Deng
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haiyong Zhu
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
16
|
Chen Y, Huang J, Fan Y, Huang L, Cai X. Understanding the cellular and molecular heterogeneity in colorectal cancer through the use of single-cell RNA sequencing. Transl Oncol 2025; 55:102374. [PMID: 40163910 PMCID: PMC11993189 DOI: 10.1016/j.tranon.2025.102374] [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: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
The very prevalent nature, genetic variability, and intricate tumor microenvironment (TUME) of colorectal cancer (COREC) are its defining features. In order to better understand the molecular and cellular make-up of COREC, this work used single-cell RNA sequencing (SRNAS) to isolate and characterize important cell types as well as their interactions within the TUME. Our analysis of 51,204 cells yielded six distinct types: epithelial, fibroblast, endothelial, T&NK, B, and myeloid. C3 B cells were shown to be the most active in immunological regulation, according to chemokine signaling study, which was one of seven clusters of B cells that were thoroughly subtyped. The examination of copy number variation (CONUV) revealed a great deal of genetic variability, especially in epithelial cells. We traced the activity of three key transcription factor clusters (M1, M2, and M3) across all B cell subtypes using transcription factor analysis. We created a predictive model that correctly sorts patients according to survival results by using marker genes from C3 B cells. In addition, the relationship between genetic changes and the immune system was better understood by tumor mutational burden (TUMUB) and immune infiltration studies. Our research sheds light on the genetic complexity and cellular variety of COREC, which in turn opens up new possibilities for targeted treatments and individualized approaches to patient care.
Collapse
Affiliation(s)
| | - Jian Huang
- Wenzhou Central Hospital, Wenzhou, China
| | - Yufang Fan
- Wenzhou Central Hospital, Wenzhou, China
| | | | | |
Collapse
|
17
|
Wang Z, Dai R, Wang M, Lei L, Zhang Z, Han K, Wang Z, Guo Q. KanCell: dissecting cellular heterogeneity in biological tissues through integrated single-cell and spatial transcriptomics. J Genet Genomics 2025; 52:689-705. [PMID: 39577768 DOI: 10.1016/j.jgg.2024.11.009] [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: 07/21/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
KanCell is a deep learning model based on Kolmogorov-Arnold networks (KAN) designed to enhance cellular heterogeneity analysis by integrating single-cell RNA sequencing and spatial transcriptomics (ST) data. ST technologies provide insights into gene expression within tissue context, revealing cellular interactions and microenvironments. To fully leverage this potential, effective computational models are crucial. We evaluate KanCell on both simulated and real datasets from technologies such as STARmap, Slide-seq, Visium, and Spatial Transcriptomics. Our results demonstrate that KanCell outperforms existing methods across metrics like PCC, SSIM, COSSIM, RMSE, JSD, ARS, and ROC, with robust performance under varying cell numbers and background noise. Real-world applications on human lymph nodes, hearts, melanoma, breast cancer, dorsolateral prefrontal cortex, and mouse embryo brains confirmed its reliability. Compared with traditional approaches, KanCell effectively captures non-linear relationships and optimizes computational efficiency through KAN, providing an accurate and efficient tool for ST. By improving data accuracy and resolving cell type composition, KanCell reveals cellular heterogeneity, clarifies disease microenvironments, and identifies therapeutic targets, addressing complex biological challenges.
Collapse
Affiliation(s)
- Zhenghui Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Ruoyan Dai
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Mengqiu Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Lixin Lei
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zhiwei Zhang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Kaitai Han
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zijun Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Qianjin Guo
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China.
| |
Collapse
|
18
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
19
|
He A, Huang Z, Chen X, Qi K, Zhang S, Li F, Lu H, Wang J, Peng J, Song C. Decoding the role of lipid metabolism in NSCLC: From macrophage subtype identification to prognostic model development. FASEB J 2025; 39:e70467. [PMID: 40277347 DOI: 10.1096/fj.202500124] [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: 01/21/2025] [Accepted: 03/07/2025] [Indexed: 04/26/2025]
Abstract
Lipid metabolism plays a pivotal role in shaping the tumor microenvironment, particularly by influencing macrophage function. This study aimed to identify lipid-associated macrophage (LAM) marker genes involved in the onset and progression of non-small cell lung cancer (NSCLC) through integrated single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) analyses. Mutation and RNA-seq data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were analyzed to explore the relationship between lipid metabolism pathways and NSCLC progression. scRNA-seq analysis revealed macrophage subtypes closely associated with lipid metabolism, with three key marker genes-S100A10, HLA-DMB, and CTSL-identified as predictive factors for patient prognosis. A prognostic risk scoring model was constructed and validated using survival analysis and ROC curves, demonstrating high accuracy in stratifying NSCLC patients by risk. Further in vivo experiments using subcutaneous tumor xenografts and lung metastasis models showed that S100A10 and CTSL promoted tumor growth and metastasis, while HLA-DMB inhibited these processes. Immune infiltration analysis highlighted the immunological relevance of the identified marker genes, providing insights into their functional roles. This study underscores the critical influence of LAMs in NSCLC progression and highlights a robust prognostic model that offers potential therapeutic targets for improving patient outcomes.
Collapse
Affiliation(s)
- Aoxiao He
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianglai Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kai Qi
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shan Zhang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Li
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jinhua Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chao Song
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Wei C, Liao K, Chen HJ, Xiao ZX, Meng Q, Liu ZK, Lu YX, Sheng H, Mo HY, Wu QN, Han Y, Zeng ZL, Guan XY, Luo HY, Ju HQ, Xu RH. Nuclear mitochondrial acetyl-CoA acetyltransferase 1 orchestrates natural killer cell-dependent antitumor immunity in colorectal cancer. Signal Transduct Target Ther 2025; 10:138. [PMID: 40289129 PMCID: PMC12034769 DOI: 10.1038/s41392-025-02221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Tumor metabolism often interferes with the immune microenvironment. Although natural killer (NK) cells play pivotal roles in antitumor immunity, the connection between NK cells and tumor metabolism remains unclear. Our systematic analysis of multiomics data and survival data from colorectal cancer (CRC) patients uncovered a novel association between mitochondrial acetyl-CoA acetyltransferase 1 (ACAT1) and NK cell infiltration that influences disease progression. ACAT1, a metabolic enzyme involved in reversible conversion of acetoacetyl-CoA to two molecules of acetyl-CoA, exhibits nuclear protein acetylation activity through its translocation. Under immune stimulation, mitochondrial ACAT1 can be phosphorylated at serine 60 (S60) and enters the nucleus; however, this process is hindered in nutrient-poor tumor microenvironments. Nuclear ACAT1 directly acetylates lysine 146 of p50 (NFKB1), attenuating its DNA binding and transcriptional repression activity and thereby increasing the expression of immune-related factors, which in turn promotes NK cell recruitment and activation to suppress colorectal cancer growth. Furthermore, significant associations are found among low nuclear ACAT1 levels, decreased S60 phosphorylation, and reduced NK cell infiltration, as well as poor prognosis in CRC. Our findings reveal an unexpected function of ACAT1 as a nuclear acetyltransferase and elucidate its role in NK cell-dependent antitumor immunity through p50 acetylation.
Collapse
Affiliation(s)
- Chen Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Kun Liao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Hao-Jie Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Zi-Xuan Xiao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Qi Meng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Ze-Kun Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Yun-Xin Lu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Hui Sheng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Hai-Yu Mo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Qi-Nian Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Yi Han
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
| | - Zhao-Lei Zeng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Hui-Yan Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China
| | - Huai-Qiang Ju
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China.
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China.
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| |
Collapse
|
21
|
Whalen KA, Henry CC, Mehta NK, Rakhra K, Yalcin S, Meetze K, Gibson NW, Baeuerle PA, Michaelson JS. CLN-619, a MICA/B monoclonal antibody that promotes innate immune cell-mediated antitumor activity. J Immunother Cancer 2025; 13:e008987. [PMID: 40274283 PMCID: PMC12020772 DOI: 10.1136/jitc-2024-008987] [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: 02/01/2024] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Major histocompatibility complex class I-related protein A and B (MICA/B) are ligands for the natural killer group 2 member D (NKG2D) receptor and are broadly expressed on tumor cells but minimally on normal tissues. When cytotoxic NKG2D-expressing immune cells engage MICA/B, the ligand-expressing cells are targeted for lysis. Cancer cells can evade NKG2D-mediated destruction by shedding MICA/B from their cell surface via proteases present in the tumor microenvironment. CLN-619 is a humanized IgG1 monoclonal antibody (mAb) which binds MICA/B and inhibits shedding resulting in accumulation of MICA/B on the tumor cell surface. CLN-619 may thereby have therapeutic effects in a broad range of malignancies by re-establishing the MICA/B-NKG2D axis to enable NKG2D-mediated, as well as Fc-gamma receptor-mediated, tumor cell lysis. METHODS CLN-619 was characterized for binding epitope and affinity, effects on surface and soluble levels of MICA/B, and in vitro tumor cell killing. In mouse models, the mAb was tested for tumor growth inhibition. The contribution of the Fc-gamma (Fcγ) 1 domain to CLN-619 activity was also assessed. RESULTS CLN-619 bound with high affinity to the alpha-3 domain of MICA/B without encumbering the interaction with NKG2D on natural killer cells. CLN-619 increased the level of cell surface expression of MICA/B and concomitantly decreased the levels of soluble MICA/B in cell culture assays. Treatment of cancer cell lines with CLN-619 induced antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. CLN-619 resulted in potent inhibition of tumor growth in multiple xenograft models and increased survival of mice in a disseminated cancer model. CONCLUSIONS CLN-619 inhibited the shedding of MICA/B to effectively restore cytotoxic signaling pathways in immune cells. Potent antitumor activity of CLN-619 as a monotherapy was observed in several preclinical models. Activity of CLN-619 required a functional Fcγ1 domain, suggesting the requirement of simultaneous engagement of NKG2D and cluster of differentiation 16A (CD16A) on immune cells for optimal cytotoxicity. The preclinical data reported here support the assessment of CLN-619 in patients with cancer.
Collapse
Affiliation(s)
- Kerry A Whalen
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | - Naveen K Mehta
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | - Kavya Rakhra
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | - Kristan Meetze
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
22
|
Li X, Deng J, Liu X, Zhou Y, Bi T, Chen J, Wang J. Tissue-resident immune cells in cervical cancer: emerging roles and therapeutic implications. Front Immunol 2025; 16:1541950. [PMID: 40330461 PMCID: PMC12053169 DOI: 10.3389/fimmu.2025.1541950] [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: 12/09/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
The favorable prognosis of "hot" tumors is widely acknowledged in oncology. Recently, the concept of tertiary lymphoid structures (TLS) has renewed appreciation for local immune cells within tumor tissues. Tissue-resident immune cells, a newly identified subset of tumor-infiltrating lymphocytes, are emerging as potential key players in tumor infiltration and TLS formation, due to their ability to reside indefinitely within tissues and mount effective responses to local antigens. Cervical cancer (CC), the fourth most common cause of cancer-related mortality among women globally, has experienced comparatively limited progress in delineating its tumor immune microenvironment compared to other malignancies. Notably, the role of tissue-resident immune cells within the CC milieu remains inadequately characterized. This comprehensive review aims to synthesize current knowledge and critically evaluate the putative roles of these cells in CC pathogenesis, providing new insights on the intricate dynamics of the local tumor microenvironment.
Collapse
Affiliation(s)
- Xidie Li
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Juan Deng
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Xiaoping Liu
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Yan Zhou
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Tingting Bi
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Jingjing Chen
- Department of Breast Surgery, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| | - Jinjin Wang
- Department of Obstetrics and Gynecology, Zhuzhou Central Hospital, Zhuzhou, Hunan, China
| |
Collapse
|
23
|
Huang X, You R, Liu F, Jian Z, Zhou G, Yin H, Wu M, Sun T, Duan Z, Xu W, Zhang S, Yang X, Jiao H, Yang S, Wang Q, Yin J, Tang H, Lin M, Tan L. Identification and validation of poor prognosis immunoevasive subtype of esophageal cancer with tumor-infiltrating SAMD3 + NK cell abundance. Cancer Immunol Immunother 2025; 74:177. [PMID: 40252130 PMCID: PMC12009252 DOI: 10.1007/s00262-025-04028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/19/2025] [Indexed: 04/21/2025]
Abstract
INTRODUCTION Esophageal cancer (EC) remains highly lethal due to tumor microenvironment (TME)-mediated immune evasion. While natural killer (NK) cells are central to antitumor immunity, their functional states in EC are poorly characterized. METHODS We integrated bulk RNA-seq (TCGA/GEO) and single-cell data to construct an NK cell-derived prognostic signature (NK score) via LASSO-Cox regression. Immunofluorescence was applied to assess the clinical relevance of SAMD3 + NK cells in EC. Using both xenograft mouse models and in vitro co-culture procedures, the impact of SAMD3 on NK cell function was confirmed. RESULTS In EC patients, the prognostic NK score-which is generated from important NK cell markers including SAMD3-was substantially correlated with a worse chance of survival. NK cells within the TME had significant levels of SAMD3 expression, as seen by immunofluorescence labeling. Moreover, NK cells with SAMD3 knockdown exhibited enhanced antitumor activity, leading to decreased tumor development in the xenograft model. DISCUSSION Our results demonstrate the predictive significance of NK cell markers in EC and pinpoint SAMD3 as a critical modulator of NK cell activity. We pioneer SAMD3 + NK cells as architects of TME immunosuppression in EC. Our findings nominate SAMD3 inhibition as a combinatorial strategy to overcome immune checkpoint blockade resistance.
Collapse
Affiliation(s)
- Xu Huang
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Runze You
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Fangyi Liu
- The School of Basic Medical Sciences, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Zitao Jian
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Guanyou Zhou
- The School of Basic Medical Sciences, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Hao Yin
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Mengyuan Wu
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Tiantao Sun
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Zhiyun Duan
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Wenyi Xu
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Shaoyuan Zhang
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Xinyu Yang
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Heng Jiao
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Shuyi Yang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Qingle Wang
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jun Yin
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Han Tang
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Miao Lin
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- The School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| | - Lijie Tan
- Departments of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
- The School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Departments of Thoracic, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
| |
Collapse
|
24
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Feng Y, Zhang X, Wang G, Yang F, Li R, Yin L, Chen D, Wang W, Wang M, Hu Z, Sh Y, Xing N. Comprehensive Integrated Analysis Reveals the Spatiotemporal Microevolution of Cancer Cells in Patients with Bone-Metastatic Prostate Cancer. Biomedicines 2025; 13:909. [PMID: 40299503 PMCID: PMC12024866 DOI: 10.3390/biomedicines13040909] [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: 03/06/2025] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Bone metastasis is a frequent and life-threatening event in advanced cancers, affecting up to 70-85% of prostate cancer patients. Understanding the cellular and molecular mechanisms underlying bone metastasis is essential for developing targeted therapies. This study aimed to systematically characterize the heterogeneity and microenvironmental adaptation of prostate cancer bone metastases using single-cell transcriptomics. Methods: We integrated the largest single-cell transcriptome dataset to date, encompassing 124 samples from primary prostate tumors, various bone metastatic sites, and non-malignant tissues (e.g., benign prostatic hyperplasia, normal bone marrow). After quality control, 602,497 high-quality single-cell transcriptomes were analyzed. We employed unsupervised clustering, gene expression profiling, mutation analysis, and metabolic pathway reconstruction to characterize cancer cell subtypes and tumor microenvironmental remodeling. Results: Cancer epithelial cells dominated the tumor microenvironment but exhibited pronounced heterogeneity, posing challenges for conventional clustering methods. By integrating genetic and metabolic features, we revealed key evolutionary trajectories of epithelial cancer cells during metastasis. Notably, we identified a novel epithelial subpopulation, NEndoCs, characterized by unique differentiation patterns and distinct spatial distribution across metastatic niches. We also observed significant metabolic reprogramming and recurrent mutations linked to prostate-to-bone microenvironmental transitions. Conclusions: This study comprehensively elucidates the mutation patterns, metabolic reprogramming, and microenvironment adaptation mechanisms of bone metastasis in prostate cancer, providing key molecular targets and clinical strategies for the precise treatment of bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Yinghua Feng
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China;
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiuli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing 100034, China;
| | - Guangpeng Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Feiya Yang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ruifang Li
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Lu Yin
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong Chen
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wenkuan Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mingshuai Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhiyuan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuan Sh
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nianzeng Xing
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China;
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
26
|
Cheng H, Yan Y, Zhang B, Ma Z, Fu S, Ji Z, Zou Z, Wang Q. Single-cell transcriptomics reveals immunosuppressive microenvironment and highlights tumor-promoting macrophage cells in Glioblastoma. PLoS One 2025; 20:e0312764. [PMID: 40193323 PMCID: PMC11975071 DOI: 10.1371/journal.pone.0312764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/13/2024] [Indexed: 04/09/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive primary brain malignancy in adults. Nevertheless, the cellular heterogeneity and complexity within the GBM microenvironment (TME) are still not fully understood, posing a significant obstacle in the advancement of more efficient immunotherapies for GBM. In this study, we conducted an integrated analysis of 48 tumor fragments from 24 GBM patients at the single-cell level, uncovering substantial molecular diversity within immune infiltrates. We characterized molecular signatures for five distinct tumor-associated macrophages (TAMs) subtypes. Notably, the TAM_MRC1 subtype displayed a pronounced M2 polarization signature. Additionally, we identified a subtype of natural killer (NK) cells, designated CD56dim_DNAJB1. This subtype is characterized by an exhausted phenotype, evidenced by an elevated stress signature and enrichment in the PD-L1/PD-1 checkpoint pathway. Our findings also highlight significant cell-cell interactions among malignant glioma cells, TAM, and NK cells within the TME. Overall, this research sheds light on the functional heterogeneity of glioma and immune cells in the TME, providing potential targets for therapeutic intervention in this immunologically cold cancer.
Collapse
Affiliation(s)
- Han Cheng
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Yan Yan
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Biao Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhuolin Ma
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Siwen Fu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhi Ji
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Ziyi Zou
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Qin Wang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
27
|
Yang Y, Liu Z, Wang Z, Fu X, Li Z, Li J, Xu Z, Cen B. Large-scale bulk and single-cell RNA sequencing combined with machine learning reveals glioblastoma-associated neutrophil heterogeneity and establishes a VEGFA + neutrophil prognostic model. Biol Direct 2025; 20:45. [PMID: 40188324 PMCID: PMC11972500 DOI: 10.1186/s13062-025-00640-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/22/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Neutrophils play a key role in the tumor microenvironment (TME); however, their functions in glioblastoma (GBM) are overlooked and insufficiently studied. A detailed analysis of GBM-associated neutrophil (GBMAN) subpopulations may offer new insights and opportunities for GBM immunotherapy. METHODS We analyzed single-cell RNA sequencing (scRNA-seq) data from 127 isocitrate dehydrogenase (IDH) wild-type GBM samples to characterize the GBMAN subgroups, emphasizing developmental trajectories, cellular communication, and transcriptional networks. We implemented 117 machine learning combinations to develop a novel risk model and compared its performance to existing glioma models. Furthermore, we assessed the biological and molecular features of the GBMAN subgroups in patients. RESULTS From integrated large-scale scRNA-seq data (498,747 cells), we identified 5,032 neutrophils and classified them into four distinct subtypes. VEGFA+GBMAN exhibited reduced inflammatory response characteristics and a tendency to interact with stromal cells. Furthermore, these subpopulations exhibited significant differences in transcriptional regulation. We also developed a risk model termed the "VEGFA+neutrophil-related signature" (VNRS) using machine learning methods. The VNRS model showed higher accuracy than previously published risk models and was an independent prognostic factor. Additionally, we observed significant differences in immunotherapy responses, TME interactions, and chemotherapy efficacy between high-risk and low-risk VNRS score groups. CONCLUSION Our study highlights the critical role of neutrophils in the TME of GBM, allowing for a better understanding of the composition and characteristics of GBMAN. The developed VNRS model serves as an effective tool for evaluating the risk and guiding clinical treatment strategies for GBM. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yufan Yang
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ziyuan Liu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhongliang Wang
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiang Fu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianlong Li
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Departments of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Bohong Cen
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong- Macao, Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
28
|
Lai W, Li Y, Luo OJ. MIST: An interpretable and flexible deep learning framework for single-T cell transcriptome and receptor analysis. SCIENCE ADVANCES 2025; 11:eadr7134. [PMID: 40184452 PMCID: PMC11970455 DOI: 10.1126/sciadv.adr7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Joint analysis of transcriptomic and T cell receptor (TCR) features at single-cell resolution provides a powerful approach for in-depth T cell immune function research. Here, we introduce a deep learning framework for single-T cell transcriptome and receptor analysis, MIST (Multi-insight for T cell). MIST features three latent spaces: gene expression, TCR, and a joint latent space. Through analyses of antigen-specific T cells, and T cell datasets related to lung cancer immunotherapy and COVID19, we demonstrate MIST's interpretability and flexibility. MIST easily and accurately resolves cell function and antigen specificity by vectorizing and integrating transcriptome and TCR data of T cells. In addition, using MIST, we identified the heterogeneity of CXCL13+ subsets in lung cancer infiltrating CD8+ T cells and their association with immunotherapy, providing additional insights into the functional transition of CXCL13+ T cells related to anti-PD-1 therapy that were not reported in the original study.
Collapse
Affiliation(s)
- Wenpu Lai
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
29
|
Chen YC, Bazewicz CG, Dinavahi SS, Huntington ND, Schell TD, Robertson GP. Emerging Role of the p53 Pathway in Modulating NK Cell-Mediated Immunity. Mol Cancer Ther 2025; 24:523-535. [PMID: 39470047 DOI: 10.1158/1535-7163.mct-24-0325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
The p53 pathway plays an important role in role in cancer immunity. Mutation or downregulation of the proteins in the p53 pathway are prevalent in many cancers, contributing to tumor progression and immune dysregulation. Recent findings suggest that the activity of p53 within tumor cells, immune cells, and the tumor microenvironment can play an important role in modulating NK cell-mediated immunity. Consequently, efforts to restore p53 pathway activity are being actively pursued to modulate this form of immunity. This review focuses on p53 activity regulating the infiltration and activation of NK cells in the tumor immune microenvironment. Furthermore, the impact of p53 and its regulation of NK cells on immunogenic cell death within solid tumors and the abscopal effect are reviewed. Finally, future avenues for therapeutically restoring p53 activity to improve NK cell-mediated antitumor immunity and optimize the effectiveness of cancer therapies are discussed.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Christopher G Bazewicz
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- oNKo-Innate Pty Ltd. Moonee Ponds, Victoria, Australia
| | - Todd D Schell
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
30
|
Lonberg N. The Problem with Syngeneic Mouse Tumor Models. Cancer Immunol Res 2025; 13:456-462. [PMID: 39996612 DOI: 10.1158/2326-6066.cir-24-1046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/16/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025]
Abstract
The advent of syngeneic mouse tumor models provided the scientific foundation for cancer immunotherapies now in widespread use. However, in many respects, these models do not faithfully recapitulate the interactions between cancer cells and the immune systems of human patients who have solid tumors because they represent a very early stage in the immune response to the newly transplanted cancer cells compared with the relatively mature stage found in human patients at the time of treatment. The lack of translatability of syngeneic models is probably responsible for many failed clinical trials conducted at considerable expense, involving far too many patients with cancer who received no benefit. Better mouse models would substantially accelerate the pace of discovery of new immunotherapies. Until these models emerge, a better understanding of the differences between the existing syngeneic models and human cancers may provide a more efficient path for moving experimental drugs into clinical development. To accomplish this, we must consider mice transplanted with syngeneic tumor cells to be in vivo assays, potentially useful for understanding the mechanism of action of immunotherapies rather than disease models.
Collapse
|
31
|
Gobbini E, Hubert M, Doffin AC, Eberhardt A, Hermet L, Li D, Duplouye P, Ghamry-Barrin S, Berthet J, Benboubker V, Grimont M, Sakref C, Perrot J, Tondeur G, Harou O, Lopez J, Dubois B, Dalle S, Caux C, Caramel J, Valladeau-Guilemond J. The Spatial Organization of cDC1 with CD8+ T Cells is Critical for the Response to Immune Checkpoint Inhibitors in Patients with Melanoma. Cancer Immunol Res 2025; 13:517-526. [PMID: 39774795 DOI: 10.1158/2326-6066.cir-24-0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/05/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Dendritic cells (DC) are promising targets for cancer immunotherapies because of their central role in the initiation and control of immune responses. The type 1 conventional DC (cDC1) population is of particular interest because of its ability to cross-present antigens to CD8+ T cells. cDC1s also secrete cytokines that allow Th1 cell polarization and NK cell activation and recruitment. However, the spatial organization and specific functions of cDC1s in response to immunotherapy remain to be clearly characterized in human tumors. In this study, we used a multiplexed immunofluorescence analysis pipeline coupled with computational image analysis to determine the spatial organization of cDC1s in skin lesions from a cohort of patients with advanced melanoma treated with immune checkpoint inhibitors (ICI). For this, we performed a whole-slide image analysis of cDC1 infiltration, distribution, and spatial interaction with key immune partners such as CD8+ T cells and plasmacytoid DCs. We also analyzed LAMP3+ DCs, which correspond to a mature subset of tumor-infiltrating DCs. Distance and cell network analyses demonstrated that cDC1s exhibited a scattered distribution compared with tumor-infiltrating plasmacytoid DCs and LAMP3+ DCs, which were preferentially organized in dense areas with high homotypic connections. The proximity and interactions between CD8+ T cells and cDC1s were positively associated with the response to ICIs. In conclusion, our study unravels the complex spatial organization of cDC1s and their interactions with CD8+ T cells in lesions of patients with melanoma, shedding light on the pivotal role of these cells in shaping the response to ICIs.
Collapse
Affiliation(s)
- Elisa Gobbini
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Institut Curie, Oncology Department, Paris, France
| | - Margaux Hubert
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Anne-Claire Doffin
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Anais Eberhardt
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Léo Hermet
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Danlin Li
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Pierre Duplouye
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Sarah Ghamry-Barrin
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Justine Berthet
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Valentin Benboubker
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Maxime Grimont
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Candice Sakref
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- LabEx DEVweCAN, Lyon, France
| | - Jimmy Perrot
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Garance Tondeur
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Olivier Harou
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Jonathan Lopez
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Bertrand Dubois
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
| | - Stephane Dalle
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Christophe Caux
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Centre Léon Bérard, Lyon, France
- Laboratoire d'Immunothérapie des Cancers de Lyon (LICL), Lyon, France
- LabEx DEVweCAN, Lyon, France
| | - Julie Caramel
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jenny Valladeau-Guilemond
- INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Univ Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Centre Léon Bérard, Lyon, France
- LabEx DEVweCAN, Lyon, France
| |
Collapse
|
32
|
Sun P, Xu H, Guo C, Yang L, Zhang X, Lu B, Chen L, Huang J. TMEM115 as an Oncogenic and Immunological Biomarker in Hepatocellular Carcinoma. Liver Int 2025; 45:e70048. [PMID: 40052693 DOI: 10.1111/liv.70048] [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: 08/15/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Transmembrane (TMEM) proteins are involved in fundamental biological processes such as material transport and signal transduction. TMEM115 is a member of the TMEM protein family, but its significance in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigate the clinical predictive significance and potential functions of TMEM115 in HCC. METHODS Bioinformatics was used to investigate TMEM115 mRNA expression and immune infiltration score. Through multiplex immunohistochemistry analysis, we assessed its protein expression and association with HCC patient clinical features, prognosis and immune cell infiltration in HCC. Through in vitro and in vivo experiments, we evaluated the biological functions of TMEM115 in HCC cells and its impact on the immune microenvironment. RESULTS TMEM115 mRNA and protein levels were significantly higher in HCC tissues compared to paracancerous liver tissues. Its protein expression correlated with clinical characteristics and overall survival in HCC patients. In HCC tissues, higher TMEM115 protein expression corresponded to lower proportions of CD66b+ neutrophils and CD8+ T cells and a higher proportion of CD4+ T cells. Furthermore, patients with low TMEM115 expression displayed higher programmed cell death ligand-1 and lower lymphocyte activation gene 3 protein expression. Functionally, TMEM115 knockdown inhibited the proliferation, migration and invasion of HCC cells. In orthotopic models, TMEM115 knockdown inhibited the growth of HCC and affected the infiltration of immune cells. CONCLUSIONS Our findings show TMEM115 as a promising prognostic indicator for HCC and hold promise in predicting responses to immune therapy, emphasising its potential clinical relevance and intricate involvement in the immune microenvironment of HCC.
Collapse
Affiliation(s)
- Pingping Sun
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Haiyan Xu
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Chengfeng Guo
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Lei Yang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Xiaojing Zhang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Bing Lu
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Lei Chen
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jianfei Huang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
- Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
33
|
Tredicine M, Mucci M, Recchiuti A, Mattoscio D. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett 2025; 599:927-951. [PMID: 39973474 PMCID: PMC11995684 DOI: 10.1002/1873-3468.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
The arachidonic acid (AA) pathway promotes tumor progression by modulating the complex interactions between cancer and immune cells within the microenvironment. In this Review, we summarize the knowledge acquired thus far concerning the intricate mechanisms through which eicosanoids either promote or suppress the antitumor immune response. In addition, we will discuss the impact of eicosanoids on immune cells and how they affect responsiveness to immunotherapy, as well as potential strategies for manipulating the AA pathway to improve anticancer immunotherapy. Understanding the molecular pathways and mechanisms underlying the role played by AA and its metabolites in tumor progression may contribute to the development of more effective anticancer immunotherapies.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Matteo Mucci
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Antonio Recchiuti
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| |
Collapse
|
34
|
Qian L, Wu L, Miao X, Xu J, Zhou Y. The role of TIGIT-CD226-PVR axis in mediating T cell exhaustion and apoptosis in NSCLC. Apoptosis 2025; 30:784-804. [PMID: 39725799 DOI: 10.1007/s10495-024-02052-2] [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] [Accepted: 11/24/2024] [Indexed: 12/28/2024]
Abstract
The treatment of non-small cell lung cancer (NSCLC) remains a critical challenge in oncology, primarily due to the dysfunction and exhaustion of T cells within the tumor microenvironment, which greatly limits the effectiveness of immunotherapy. This study investigates the regulatory role of the T cell immunoglobulin and ITIM domain (TIGIT)-CD226-PVR signaling axis in the exhaustion and apoptosis of cluster of differentiation (CD)27+/CD127+T cells in NSCLC. Utilizing single-cell sequencing technology, we conducted a comprehensive gene expression analysis of T cells in a mouse model of NSCLC. Bioinformatics analysis revealed that the TIGIT-CD226-PVR signaling axis is highly active in the CD27+/CD127+T cell subset and is closely associated with their functional decline and exhaustion. In vitro experiments further demonstrated that inhibiting the TIGIT-PVR pathway while activating the CD226-PVR pathway significantly restored T cell proliferation and effector function. Importantly, in vivo studies showed that targeting this axis can significantly alleviate T cell exhaustion, enhance their cytotoxicity against NSCLC cells, and promote apoptosis, thereby improving the efficacy of immunotherapy.
Collapse
MESH Headings
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Apoptosis/genetics
- Animals
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- T Lineage-Specific Activation Antigen 1
- Humans
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/genetics
- Mice
- Receptors, Virus/metabolism
- Receptors, Virus/genetics
- Signal Transduction
- Cell Line, Tumor
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/immunology
- Cell Proliferation
- T-Cell Exhaustion
Collapse
Affiliation(s)
- Liang Qian
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Ling Wu
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Xiaohui Miao
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Jiao Xu
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China
| | - Yao Zhou
- Department of Respiratory and Critical Care Medicine, WuJin Hospital Afliated With Jiangsu University, WuJin Clinical College of Xuzhou Medical University, No.2, Yongning North Road, Changzhou, 213017, Jiangsu, China.
| |
Collapse
|
35
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
36
|
Li CX, Huang C, Chen DS. scPANDA: PAN-Blood Data Annotator with a 10-Million Single-Cell Atlas. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2025; 40:68-87. [PMID: 40164519 DOI: 10.24920/004472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
OBJECTIVES Recent advancements in single-cell RNA sequencing (scRNA-seq) have revolutionized the study of cellular heterogeneity, particularly within the hematological system. However, accurately annotating cell types remains challenging due to the complexity of immune cells. To address this challenge, we develop a PAN-blood single-cell Data Annotator (scPANDA), which leverages a comprehensive 10-million-cell atlas to provide precise cell type annotation. METHODS The atlas, constructed from data collected in 16 studies, incorporated rigorous quality control, preprocessing, and integration steps to ensure a high-quality reference for annotation. scPANDA utilizes a three-layer inference approach, progressively refining cell types from broad compartments to specific clusters. Iterative clustering and harmonization processes were employed to maintain cell type purity throughout the analysis. Furthermore, the performance of scPANDA was evaluated in three external datasets. RESULTS The atlas was structured hierarchically, consisting of 16 compartments, 54 classes, 4,460 low-level clusters (pd_cc_cl_tfs), and 611 high-level clusters (pmid_cts). Robust performance of the tool was demonstrated in annotating diverse immune scRNA-seq datasets, analyzing immune-tumor coexisting clusters in renal cell carcinoma, and identifying conserved cell clusters across species. CONCLUSIONS scPANDA exemplifies effective reference mapping with a large-scale atlas, enhancing the accuracy and reliability of blood cell type identification.
Collapse
Affiliation(s)
- Chang-Xiao Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Can Huang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Dong-Sheng Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China.
| |
Collapse
|
37
|
Dong J, Fan L, Wu Q, Zheng Z. Retinoid X receptor γ predicts the prognosis and is associated with immune infiltration in kidney renal clear cell carcinoma: a qRT-PCR, TCGA and in silico research. BMC Urol 2025; 25:62. [PMID: 40155870 PMCID: PMC11951502 DOI: 10.1186/s12894-025-01744-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 03/14/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Kidney clear cell carcinoma (KIRC) stands as one of the most prevalent primary malignant tumors, showcasing significant heterogeneity within the urological system. However, the precise molecular mechanisms underpinning tumorigenesis in KIRC remain elusive. While Retinoid X receptor γ (RXRG) has been implicated in various diseases and human cancers, its specific role in KIRC remains undetermined. This research aimed to investigate the involvement of RXRG in KIRC pathogenesis. METHODS Quantitative real-time polymerase chain reaction was performed to evaluate the expression levels of RXRG in KIRC. Utilizing RNA-seq data and corresponding clinicopathological information from The Cancer Genome Atlas (TCGA) database, we embarked on an analysis to ascertain the prognostic significance of RXRG in KIRC. Furthermore, bioinformatics analyses were employed to delineate the preliminary molecular mechanisms through which RXRG operates in KIRC tumorigenesis. RESULTS Our findings revealed a significant downregulation of RXRG in KIRC tumor tissues compared to normal kidney tissues, as evidenced in local and TCGA cohorts. Diminished RXRG expression correlated with adverse clinicopathological characteristics, including larger tumor size, higher clinical stage, and advanced histologic grade. Cox regression analyses unveiled that reduced RXRG expression was associated with poorer overall survival (OS) and disease-free survival (DFS) rates in KIRC patients. Bioinformatics analyses indicated that the RXRG-related differentially expressed genes (DEGs) were involved in tumorigenesis and metabolism by regulating a series of signaling pathways. Using ssGSEA, we found that RXRG expression was significantly associated with NK cells and macrophages. CONCLUSION Our study provides new insights and evidence that RXRG is involved in the tumorigenesis of KIRC and may be a suitable target for immunotherapy in KIRC.
Collapse
Affiliation(s)
- Jianda Dong
- Department of Neck Surgery, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lailai Fan
- Department of Urinary Surgery, The Second Affiliated Hospital and Yuying, Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiaolin Wu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouci Zheng
- Department of Neck Surgery, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
38
|
Qiu J, Shen Z, Jiang G, Ni Q. Study on Heparanase Promoting Breast Cancer Cell Proliferation and Inhibiting NK Cell Cytolytic Activity Via the PD-1/PD-L1 Pathway. Clin Breast Cancer 2025:S1526-8209(25)00080-1. [PMID: 40253275 DOI: 10.1016/j.clbc.2025.03.012] [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: 12/26/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVE To explore the mechanism of action of heparanase (HPSE) in breast cancer (BC), particularly its impact on BC cell proliferation and natural killer (NK) cell cytolytic activity through the programmed cell death protein 1 (PD-1)/PD ligand 1 (PD-L1) pathway. METHODS HPSE expression levels were analyzed in BC cells. MDA-MB-231 cells, a widely used triple-negative BC model, were treated and assessed using RT-qPCR and Western blotting for gene and protein expression. CCK-8, Transwell assays, and flow cytometry were used to evaluate cell proliferation, migration, invasion, and apoptosis. NK cells isolated from healthy donors were treated with HPSE and co-cultured with BC cells to assess cytolytic activity through interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) release measurements. A nude mouse xenograft model was established to examine the in vivo effects of HPSE on tumor growth and NK cell function. RESULTS HPSE overexpression enhanced BC cell proliferation, migration, and invasion, while reducing apoptosis. It also upregulated PD-1 and PD-L1 expression, leading to impaired NK cell cytolytic activity and decreased secretion of TNF-α and IFN-γ. Inhibition of the PD-1/PD-L1 pathway partially reversed the pro-tumor effects of HPSE and restored NK cell cytolytic function. In vivo, HPSE overexpression promoted tumor growth and reduced NK cell activity within tumor tissues. CONCLUSION These findings reveal HPSE as a key regulator of BC immune evasion, likely via modulation of the PD-1/PD-L1 axis. Targeting HPSE, particularly in combination with PD-1/PD-L1 inhibitors, may offer a novel therapeutic strategy for BC treatment.
Collapse
Affiliation(s)
- Jian Qiu
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Thyroid and Breast Surgery, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
| | - Zhongyi Shen
- Health Management Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province, China
| | - Guoqin Jiang
- Thyroid and Breast Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Qichao Ni
- Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
39
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [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: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
40
|
Liu C, Liu X, Cao P, Xin H, Li X, Zhu S. Circadian rhythm related genes identified through tumorigenesis and immune infiltration-guided strategies as predictors of prognosis, immunotherapy response, and candidate drugs in skin cutaneous malignant melanoma. Front Immunol 2025; 16:1513750. [PMID: 40191195 PMCID: PMC11968383 DOI: 10.3389/fimmu.2025.1513750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Background Skin cutaneous malignant melanoma (SKCM) is among the most aggressive forms of skin cancer, notorious for its rapid progression and poor prognosis under late diagnosis. This study investigates the role of circadian rhythm-related genes (CRGs) in SKCM addressing a gap in understanding how CRGs affect tumor progression and patient outcomes. Methods An analysis of CRGs expression was conducted on SKCM samples derived from The Cancer Genome Atlas datasets(TCGA). Moreover, a correlation between various subtypes and their clinical features was identified. The study employed various bioinformatics methods, including differential expression analysis, consensus clustering, and survival analysis, to investigate the role of CRGs. The functional consequences of various CRG expression patterns were further investigated using immune infiltration analysis and gene set variation analysis (GSVA). A scoring system based on CRGs was developed to predict overall survival (OS) and treatment responses in SKCM patients. The predictive accuracy of this score system was then tested, and a nomogram was used to improve its clinical usefulness. Results Key findings from this study include significant genetic alterations in circadian rhythm-related genes (CRGs) in skin cutaneous melanoma (SKCM), such as mutations and CNVs. Two molecular subtypes with distinct clinical outcomes and immune profiles were identified. A prognostic model based on six CRGs (CMTM, TNPO1, CTBS, UTRN, HK2, and LIF) was developed and validated with TCGA and GEO datasets, showing high predictive accuracy for overall survival (OS). A high CRGs score correlated with poor OS, immune checkpoint expression, and reduced sensitivity to several chemotherapeutics, including AKT inhibitor VIII and Camptothecin. Conclusions This work provides valuable insights into the circadian regulation of SKCM and underscores the potential of CRGs as biomarkers for prognosis and targets for therapeutic interventions. The novel molecular subtypes and CRGs prognostic scoring model introduced in this study offer significant contributions to the understanding and management of SKCM.
Collapse
Affiliation(s)
- Chengling Liu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xingchen Liu
- Department of Pathology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Pengjuan Cao
- Department of Endocrinology and Traditional Chinese Medicine, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Haiming Xin
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xin Li
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Sailing Zhu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| |
Collapse
|
41
|
Lin Y, Lin Q, Guan Q, Chen D, Zhou Y, Li S. Immune cell infiltration as a prognostic factor in endometrial cancer: a meta-analysis. Am J Cancer Res 2025; 15:1335-1345. [PMID: 40226477 PMCID: PMC11982738 DOI: 10.62347/bxzm8857] [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: 09/27/2024] [Accepted: 12/13/2024] [Indexed: 04/15/2025] Open
Abstract
The immune system's role in cancer development and progression is receiving increasing attention. Endometrial cancer, common gynecological malignancy, has exhibited promising responses to immunotherapies. This study aims to assess the prognostic significance of various immune cell subsets in endometrial cancer, focusing on potential novel biomarkers and therapeutic targets. A systematic literature review and meta-analysis were conducted. Eleven eligible studies, comprising 2,319 patients with endometrial cancer, were included. The primary outcome was the association between levels of immune cell types, particularly CD8+ T cells, and overall prognosis. The meta-analysis found that high levels of tumor-infiltrating lymphocytes (TILs), particularly CD8+ T cells, were significantly associated with better overall prognosis in endometrial cancer patients. These findings suggest that the tumor immune microenvironment plays a crucial role in endometrial cancer prognosis. This meta-analysis indicates that higher levels of CD8+ T cells in the tumor microenvironment are linked to improved prognosis in endometrial cancer, underscoring the immune system's potential in prognostication and therapy.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| | - Qiaoming Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| | - Qi Guan
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| | - Danru Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| | - Yan Zhou
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| | - Sang Li
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital Fuzhou 350014, Fujian, PR China
| |
Collapse
|
42
|
Meng Z, Li J, Wang H, Cao Z, Lu W, Niu X, Yang Y, Li Z, Wang Y, Lu S. NLRP4 unlocks an NK/macrophages-centered ecosystem to suppress non-small cell lung cancer. Biomark Res 2025; 13:44. [PMID: 40087771 PMCID: PMC11909883 DOI: 10.1186/s40364-025-00756-4] [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: 10/04/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Tumor immune evasion extends beyond T cells, affecting innate immune elements like natural killer cells (NK) and macrophages within the tumor-immune microenvironment (TIME). Nevertheless, translational strategies to trigger collaboration of NK cells and macrophages to initiate sufficient anti-tumor cytoxicity remain scarce and are urgently needed. METHODS In this study, TCGA datasets was used to confirm the prognosis value of the expression level of NLR family pyrin domain containing 4 (NLRP4) in NSCLC and the tumor tissues microarray was used to further check its clinical-relevance at protein-level. Subsequently, a tumor cell line with stable NLRP4 overexpression was established and subcutaneous tumor models in C57BL/6J mice were used to validate the anti-tumor characteristics of NLRP4. After analyzing the tumor microenvironment using flow cytometry and multiplex immunofluorescence, we further validated our findings through co-culture transwell assays and TCGA analysis. Utilizing bulk-RNA sequencing, proteomics, and mass spectrometry of mouse tumor tissues, we innovatively identified the downstream pathways of NLRP4 and verified them through co-immunoprecipitation (co-IP) and Western blot (WB) experiments. RESULTS NLRP4 could trigger a distinct anti-tumor ecosystem organized by TIGIT+TNFA+ NK and iNOS+ M1 in lung cancer, discovered in TCGA analysis and verified in murine model. NLRP4-eco exerted tumor-suppression capacity through chemokine reprogramming including CCL5 and CXCL2. Meanwhile, the cytoxicity of NK could be facilitated by iNOS+M1. Mechanistically, NLRP4 stimulated PI3K/Akt-NF-kB axis through suppression of the activity of PP2A. Besides, knockdown of CCL5 and blockade of CXCL2-CXCR2 axis abolished chemotaxis of TIGIT+TNFA+ NK and iNOS+ M1 respectively, as well as for LB-100, a PP2A inhibitor. CONCLUSION Altogether, we delineated NLRP4's unexplored facets and discovered an NLRP4-driven anti-tumor ecosystem composed of TIGIT+TNFA+ NK and iNOS+ M1. Finally, targeting PP2A by its inhibitor successfully mimicked the anti-tumor capacity of the overexpression of NLRP4.
Collapse
Affiliation(s)
- Zhouwenli Meng
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jian Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Hui Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhengqi Cao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Xiaomin Niu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yi Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| |
Collapse
|
43
|
Xue L, Gao L, Zhou S, Yan C, Zhang X, Lin W, Li H, Shen Y, Wang X. Single-cell RNA sequencing revealed changes in the tumor microenvironment induced by radiotherapy for cervical cancer and the molecular mechanism of mast cells in immunosuppression. Funct Integr Genomics 2025; 25:63. [PMID: 40082276 DOI: 10.1007/s10142-025-01564-7] [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: 11/30/2024] [Revised: 01/25/2025] [Accepted: 02/23/2025] [Indexed: 03/16/2025]
Abstract
Radiotherapy (RT) is an important treatment for cervical cancer (CC), effectively controlling tumor growth and improving survival rates. However, radiotherapy-induced cell heterogeneity and its underlying mechanisms remain unclear, which may potentially impact treatment efficacy. This study aims to investigate tumor microenvironment changes following radiotherapy for CC, hoping to provide evidence to improve the therapeutic effects of radiotherapy. For the first time, we applied single-cell RNA sequencing (scRNA-seq) to analyze tissue samples from three CC patients pre- and post-radiotherapy. We obtained gene expression data from 52,506 cells to identify the cellular changes and molecular mechanisms induced by radiotherapy. Radiotherapy significantly alters cellular composition and gene expression within the tumor microenvironment (TME), notably upregulating mast cell expression. Mast cells are involved in multiple cell axes in the CC ecosystem after radiotherapy, and play a pivotal role in tumor immunosuppression and matrix remodeling. scRNA-seq revealed gene expression variations among cell types after radiotherapy, underscoring the importance of specific cell types in modulating the TME post-treatment. This study revealed the molecular mechanism of radiotherapy for CC and the role of mast cells, providing a foundation for optimizing the personalized treatment of CC.
Collapse
Affiliation(s)
- Lujiadai Xue
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China
| | - Linzhi Gao
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China
| | - Shimin Zhou
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China
| | - Chaofan Yan
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China
| | - Xian Zhang
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China
| | - Wei Lin
- Department of Gynecology, The First Peoples Hospital of Changde City, No 388 People's East Road, Wuling District, Changde City, 415000, China
| | - Hu Li
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China.
| | - Yuan Shen
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China.
| | - Xiaoyu Wang
- Department of Gynecology, Tianhe District, The First Affiliated Hospital of Jinan University, No.613 West Huangpu Avenue, Guangzhou City, 510000, China.
| |
Collapse
|
44
|
Maddineni S, Sharma K, Mohammad IA, Ruggiero-Sherman AD, Stepanek I, Shin JH, Bando JK, Sunwoo JB. An intraepithelial ILC1-like natural killer cell subset produces IL-13. Front Immunol 2025; 16:1521086. [PMID: 40114916 PMCID: PMC11922857 DOI: 10.3389/fimmu.2025.1521086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Natural killer (NK) cells are innate immune effectors with considerable heterogeneity and potent antitumor capabilities. Intraepithelial ILC1 (ieILC1)-like NK cells, a population of cytotoxic tissue-resident innate lymphoid cells, have recently been documented in the microenvironment of head and neck squamous cell carcinomas (HNSCC) and other solid tumors. These cells have antitumor cytolytic potential and are potent producers of type 1 cytokines, including IFNγ. Here, we identify a subpopulation of ex vivo differentiated ieILC1-like NK cells that produce IL-13 upon stimulation. Functional characterization revealed that these cells co-expressed IFNγ and IL-13 while maintaining an ILC1 transcriptional signature. IL-13 was induced either upon co-culture with tumor cell lines, or in response to TGF-β and IL-15. IL-13-expressing ieILC1-like NK cells were identified among tumor infiltrating lymphocytes expanded from patient HNSCC tumors, in support of their in vivoexistence in primary tumors. These data demonstrate additional heterogeneity within the ieILC1-like NK cell population than previously appreciated and highlight a unique form of ILC plasticity in which cells with clear ILC1 transcriptional profiles express a type 2 cytokine. With the known roles of IL-13 in cancer cell growth dynamics and immunoregulation, the identification of this subset within tumor microenvironments presents a potential target for therapeutic manipulation.
Collapse
Affiliation(s)
- Sainiteesh Maddineni
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Krishna Sharma
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Imran A. Mohammad
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Alistaire D. Ruggiero-Sherman
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Ivan Stepanek
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - June Ho Shin
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Jennifer K. Bando
- Department of Microbiology & Immunology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - John B. Sunwoo
- Department of Otolaryngology — Head & Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
45
|
Zhang X, Chen Y, Liu X, Li G, Zhang S, Zhang Q, Cui Z, Qin M, Simon HU, Terzić J, Kocic G, Polić B, Yin C, Li X, Zheng T, Liu B, Zhu Y. STING in cancer immunoediting: Modeling tumor-immune dynamics throughout cancer development. Cancer Lett 2025; 612:217410. [PMID: 39826670 DOI: 10.1016/j.canlet.2024.217410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Cancer immunoediting is a dynamic process of tumor-immune system interaction that plays a critical role in cancer development and progression. Recent studies have highlighted the importance of innate signaling pathways possessed by both cancer cells and immune cells in this process. The STING molecule, a pivotal innate immune signaling molecule, mediates DNA-triggered immune responses in both cancer cells and immune cells, modulating the anti-tumor immune response and shaping the efficacy of immunotherapy. Emerging evidence has shown that the activation of STING signaling has dual opposing effects in cancer progression, simultaneously provoking and restricting anti-tumor immunity, and participating in every phase of cancer immunoediting, including immune elimination, equilibrium, and escape. In this review, we elucidate the roles of STING in the process of cancer immunoediting and discuss the dichotomous effects of STING agonists in the cancer immunotherapy response or resistance. A profound understanding of the sophisticated roles of STING signaling pathway in cancer immunoediting would potentially inspire the development of novel cancer therapeutic approaches and overcome the undesirable protumor effects of STING activation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yan Chen
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xi Liu
- Department of Cardiology, ordos central hospital, Ordos, People's Republic of China
| | - Guoli Li
- Department of Colorectal and Anal Surgery, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, People's Republic of China
| | - Shuo Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Minglu Qin
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, 16816, Germany
| | - Janoš Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Split, Croatia
| | - Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Croatia
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, People's Republic of China.
| | - Bing Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; School of Stomatology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Yuanyuan Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
46
|
Li C, Liu C, Ma H, Zhang Z, Zhang J. Lymphocytes-Associated Extracellular Vesicles Activate Natural Killer Cells in HNSCC. Cancer Sci 2025; 116:633-642. [PMID: 39749376 PMCID: PMC11875761 DOI: 10.1111/cas.16440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Small extracellular vesicles (sEVs) facilitate intercellular communication and play a pivotal role in tumor progression. Accumulated evidence has indicated the diversity of sEVs but with limited results revealing the landscape of heterogeneity of sEVs. The heterogeneity of cargo RNA in sEVs presents the different cell origins and indicates different functions. Here, we analyzed the heterogeneity of sEVs at droplet levels from single-cell RNA sequencing results of head and neck squamous cell carcinoma (HNSCC) with the previously reported algorithm SEVtras. With the sEVs secretion activity calculated by SEVtras, we also found that the T cells held the major role of sEVs secretion. In addition, we found these sEVs secreted by T cells increased the cytotoxic ability of natural killer cells (NK cells), which illustrated an indirect manner for the anti-tumor function of T cells. These results revealed the heterogeneity of cargo RNA of sEVs in HNSCC and underlined a sEVs-dependent manner in which T cells act on NK cells and anti-tumor immunity.
Collapse
Affiliation(s)
- Chuwen Li
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Chun Liu
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Hailong Ma
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghaiPeople's Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| |
Collapse
|
47
|
Xiao L, Shen Z, Pan Z, Qiu Y, Huang D, Liu Y, Liu C, Zhang X. High-dimensional deconstruction of HNSC reveals clinically distinct cellular states and ecosystems that are associated with prognosis and therapy response. J Transl Med 2025; 23:254. [PMID: 40025504 PMCID: PMC11872339 DOI: 10.1186/s12967-025-06299-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/23/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Characterizing the variety of cell types in the tumor microenvironment (TME) and their organization into cellular communities is vital for elucidating the biological diversity of cancer and informing therapeutic strategies. METHODS Here, we employed a machine learning-based algorithm framework, EcoTyper, to analyze single-cell transcriptomes from 139 patients with head and neck squamous cell carcinoma (HNSC)and gene expression profiles from 983 additional HNSC patients, aiming to delineate the fundamental cell states and ecosystems integral to HNSC. RESULTS A diverse landscape of 66 cell states and 9 ecosystems within the HNSC microenvironment was identified, revealing classical cell types while also expanding upon previous immune classifications. Survival analysis revealed that specific cell states and ecotypes (ecosystems) are associated with patient prognosis, underscoring their potential as indicators of clinical outcomes. Moreover, distinct cell states and ecotypes exhibited varying responses to immunotherapy and chemotherapy, with several showing promise as predictive biomarkers for treatment efficacy. CONCLUSION Our large-scale integrative transcriptome analysis provides high-resolution insights into the cellular states and ecosystems of HNSC, facilitating the discovery of novel biomarkers and supporting the development of precision therapies.
Collapse
Affiliation(s)
- Lei Xiao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Zhe Shen
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Chao Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
48
|
Sibai M, Cervilla S, Grases D, Musulen E, Lazcano R, Mo CK, Davalos V, Fortian A, Bernat A, Romeo M, Tokheim C, Barretina J, Lazar AJ, Ding L, DUTRENEO Study Investigators, Grande E, Real FX, Esteller M, Bailey MH, Porta-Pardo E. The spatial landscape of cancer hallmarks reveals patterns of tumor ecological dynamics and drug sensitivity. Cell Rep 2025; 44:115229. [PMID: 39864059 DOI: 10.1016/j.celrep.2024.115229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 08/15/2024] [Accepted: 12/31/2024] [Indexed: 01/28/2025] Open
Abstract
Tumors are complex ecosystems of interacting cell types. The concept of cancer hallmarks distills this complexity into underlying principles that govern tumor growth. Here, we explore the spatial distribution of cancer hallmarks across 63 primary untreated tumors from 10 cancer types using spatial transcriptomics. We show that hallmark activity is spatially organized, with the cancer compartment contributing to the activity of seven out of 13 hallmarks, while the tumor microenvironment (TME) contributes to the activity of the rest. Additionally, we discover that genomic distance between tumor subclones correlates with differences in hallmark activity, even leading to clone-hallmark specialization. Finally, we demonstrate interdependent relationships between hallmarks at the junctions of TME and cancer compartments and how they relate to sensitivity to different neoadjuvant treatments in 33 bladder cancer patients from the DUTRENEO trial. In conclusion, our findings may improve our understanding of tumor ecology and help identify new drug biomarkers.
Collapse
Affiliation(s)
- Mustafa Sibai
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain
| | - Sergi Cervilla
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Daniela Grases
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Eva Musulen
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain; Department of Pathology, Hospital Universitari General de Catalunya Grupo-QuirónSalud, Sant Cugat del Vallès, Spain
| | - Rossana Lazcano
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chia-Kuei Mo
- Department of Medicine and Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Veronica Davalos
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Arola Fortian
- Institut de Recerca Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Adrià Bernat
- Institut de Recerca Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Margarita Romeo
- Institut de Recerca Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Collin Tokheim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jordi Barretina
- Institut de Recerca Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Alexander J Lazar
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Li Ding
- Department of Medicine and Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Enrique Grande
- Medical Oncology Department. MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Francisco X Real
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain; Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Matthew H Bailey
- Department of Biology and Simmons Center for Cancer Research, Brigham Young University, Provo, UT, USA
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| |
Collapse
Collaborators
Enrique Grande, Teresa Alonso-Gordoa, Mario Álvarez-Maestro, Elena Andrada, Ainara Azueta, Raquel Benítez Javier Burgos, Daniel Castellano, M Angel Climent, Mario Domínguez, Ignacio Durán Albert Font, Isabel Galante, Patricia Galván, Juan F García, Xavier García Del Muro, Félix Guerrero-Ramos, Núria Malats, Miriam Marqués, Pablo Maroto, Jaime Martínez de Villarreal, Ane Moreno-Oya, Jesús M Paramio, Alvaro Pinto, Aleix Prat, Javier Puente, Oscar Reig, Francisco X Real,
Collapse
|
49
|
Zhao Y, He H, Huang L, Yu L. Comprehensive analysis of lipid metabolic signatures identified CEBPD promotes breast cancer cell proliferation. Sci Rep 2025; 15:6570. [PMID: 39994306 PMCID: PMC11850814 DOI: 10.1038/s41598-025-90869-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Breast cancer (BRCA) remains the leading cause of cancer-related mortality worldwide, with lipid metabolism emerging as a critical factor in tumor progression that influences cell proliferation, migration, and immune response. Insights into lipid metabolism signatures and associated genes may offer new prognostic and therapeutic avenues. In this study, we leveraged scRNA-seq and bulk transcriptome data to assess the expression patterns and prognostic significance of lipid metabolism-related genes in BRCA. Through single-cell transcriptomic analysis of primary BRCA samples, we identified a specific set of lipid metabolism signature genes and constructed a prognostic risk model based on these signatures. This model enables patient stratification by risk scores, supporting an integrated analysis of lipid metabolism, immune landscape, and clinical outcomes. Importantly, we identified CEBPD, ABCA1, and CYP27A1 as independent prognostic genes linked to lipid metabolism, with functional assays revealing an inhibitory role for CEBPD in BRCA cell proliferation. Our findings underscore the influence of adipocytes in BRCA progression and propose CEBPD as a potential target for therapeutic intervention. This study provides a foundation for further exploration of metabolism-based strategies to enhance BRCA outcomes.
Collapse
Affiliation(s)
- Yu Zhao
- People's Hospital of Qianxinan Prefecture, Xingyi, Guizhou, China
| | - Huan He
- People's Hospital of Qianxinan Prefecture, Xingyi, Guizhou, China
| | - Linyan Huang
- People's Hospital of Qianxinan Prefecture, Xingyi, Guizhou, China
| | - Linna Yu
- People's Hospital of Qianxinan Prefecture, Xingyi, Guizhou, China.
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
50
|
Liu Y, Lyu D, Yao Y, Cui J, Liu J, Bai Z, Zhao Z, Li Y, Lu B, Dong K, Pan X. The comprehensive potential of AQP1 as a tumor biomarker: evidence from kidney neoplasm cohorts, cell experiments and pan-cancer analysis. Hum Genomics 2025; 19:15. [PMID: 39988693 PMCID: PMC11849320 DOI: 10.1186/s40246-025-00726-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
Aquaporin1 (AQP1) facilitates water transport. Its ability to be a biomarker at the pan-cancer level remains uninvestigated. We performed immunohistochemical staining on tissues from 370 individuals with kidney neoplasms to measure AQP1 expression. We utilized Kaplan-Meier survival analysis, Chi-square tests, and multivariate Cox regression analyses to assess the prognostic relevance of AQP1 expression. In the pan-cancer context, we explored AQP1's competing endogenous RNAs network, protein-protein interactions, genomic changes, gene set enrichment analysis (GSEA), the correlation of AQP1 expression with survival outcomes, drug sensitivity, drug molecular docking, tumor purity and immunity. AQP1 shRNA expressing 786-O cells were established. Cell proliferation was assessed by Cell Counting Kit-8 and colony formation. Transwell migration, invasion, and cell scratch assays were conducted. In our study, AQP1 expression was an independent protective factor for OS and PFS in renal cancer patients. AQP1 expression significantly correlated with survival outcomes in renal cancers, LGG, SARC, HNSC and UVM. PI-103 sensitivity was related to AQP1 expression and had potential binding cite with AQP1 protein. Knockdown of AQP1 reduced cell proliferation, migration and invasion. Our study uncovered AQP1 as a biomarker for favorable survival outcomes in renal cancers. Furthermore, the bioinformatic analysis promoted its implication in pan-cancer scope.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- BGI research, BGI-Hangzhou, Hangzhou, 310012, China
| | - Donghao Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- BGI research, BGI-Hangzhou, Hangzhou, 310012, China
| | - Jinming Cui
- Ulink College of Shanghai, Shanghai, 201615, China
| | - Jiangui Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zikuan Bai
- Shanghai YK Pao School, Shanghai, 201620, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuanan Li
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Department of urology, Chinese PLA general hospital of central theater command, Wuhan, 430061, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|