1
|
Zhu B, Wan H, Ling Z, Jiang H, Pei J. Machine learning and single-cell analysis uncover distinctive characteristics of CD300LG within the TNBC immune microenvironment: experimental validation. Clin Exp Med 2025; 25:167. [PMID: 40382513 PMCID: PMC12085369 DOI: 10.1007/s10238-025-01690-3] [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: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Investigating the essential function of CD300LG within the tumor microenvironment in triple-negative breast cancer (TNBC). Transcriptomic and single-cell data from TNBC were systematically collected and integrated. Four machine learning algorithms were employed to identify distinct target genes in TNBC patients. Specifically, CIBERSORT and ssGSEA algorithms were utilized to elucidate immune infiltration patterns, whereas TIDE and TCGA algorithms predicted immune-related outcomes. Moreover, single-cell sequencing data were analyzed to investigate the function of CD300LG-positive cells within the tumor microenvironment. Finally, immunofluorescence staining confirmed the significance of CD300LG in tumor phenotyping. After machine learning screening and independent dataset validation, CD300LG was identified as a unique prognostic biomarker for triple-negative breast cancer. Enrichment analysis revealed that CD300LG expression is strongly linked to immune infiltration and inflammation-related pathways, especially those associated with the cell cycle. The presence of CD8+ T cells and M1-type macrophages was elevated in the CD300LG higher group, whereas the abundance of M2-type macrophage infiltration showed a significant decrease. Immunotherapy prediction models indicated that individuals with low CD300LG expression exhibited better responses to PD-1 therapy. Additionally, single-cell RNA sequencing and immunofluorescence analyses uncovered a robust association between CD300LG and genes involved in tumor invasion. CD300LG plays a pivotal role in the tumor microenvironment of TNBC and represents a promising therapeutic target.
Collapse
Affiliation(s)
- Baoxi Zhu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Thyroid and Breast Surgery, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, China
| | - Hong Wan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zichen Ling
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Han Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Pei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
2
|
Wang X, Xu Z, Lin Z, Wu D, Tang Y, Pei Z, Gao Y, He J. Molecular subtypes based on ferroptosis-related genes and tumor microenvironment infiltration characterization in small cell lung cancer. Front Immunol 2025; 16:1574434. [PMID: 40433367 PMCID: PMC12106331 DOI: 10.3389/fimmu.2025.1574434] [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: 02/10/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background Ferroptosis is an iron-dependent form of regulated cell death associated with cancer. However, the characteristics of ferroptosis in small cell lung cancer (SCLC) are still uncertain. This study aimed to explore the application value of ferroptosis-related genes (FRGs) classification in prognosis and characteristics prediction to provide clues for targeted SCLC therapy. Method We systematically characterized mRNA expression and genetic alterations of FRGs in SCLC, evaluating their expression pattern in 181 samples from 3 datasets. Unsupervised clustering analysis was performed to identify the molecular subtypes based on FRGs. We then conducted association analyses between FRG subtypes and various tumor microenvironment (TME) characteristics, traditional key transcript factor subtypes, clinical features, transcriptional and post-transcriptional regulation, drug response, and the efficacy of immunotherapy. Furthermore, the novel classification was validated in an independent cohort of 34 samples from Beijing. Result In this study, we identified three distinct ferroptosis subtypes in SCLC: S1, S2, and S3. We found that patients in S2 had the poorest prognosis. The FRG classification was correlated with the NOTCH pathway, MYC pathway, Neuroendocrine (NE), and epithelial-to-mesenchymal transition (EMT) process. Additionally, the FRG classification was strongly associated with TME 4 subtypes. To validate the classification, we employed an independent cohort. The FRG classification could also help to guide the prediction of chemical drugs. Finally, the heatmap showed the landscape of FRG subtypes, TME subtypes, NE subtypes, key transcription subtypes, age, gender, and stage. Conclusion Our identification of new SCLC subtypes provides novel insights into tumor biology and has potential clinical implications for the management of SCLC.
Collapse
Affiliation(s)
- Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Clinical Trials Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, Taiyuan, China
| | - Zhenyi Xu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhen Lin
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Dawei Wu
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Tang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihua Pei
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Yibo 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, China
| | - Jie He
- 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, China
| |
Collapse
|
3
|
Li R, Lv W, Wang DL, Chen N. A Systematic Review of Immune Cell Roles in Breast Cancer Immunotherapy. Cancer Rep (Hoboken) 2025; 8:e70217. [PMID: 40356222 PMCID: PMC12069222 DOI: 10.1002/cnr2.70217] [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: 11/29/2024] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent malignancy among women and is associated with high mortality and significant clinical challenges. Although conventional treatments such as surgery, chemotherapy, and radiotherapy have significantly improved patient survival, their efficacy remains limited by severe side effects and treatment resistance. In recent years, advances in immunotherapy have underscored the pivotal role of immune cells in treating BC. RECENT FINDINGS This systematic review summarizes the current knowledge on the roles of immune cells within the BC tumor microenvironment (TME), including their phenotypes, functions, and implications for immunotherapy. Following PRISMA guidelines, 71 studies published between 2010 and 2024 were analyzed. The results indicate that immune cell populations-such as tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), natural killer (NK) cells, dendritic cells (DCs), and myeloid-derived suppressor cells (MDSCs)-are integral to tumor progression and therapeutic response. However, their functional heterogeneity and plasticity remain key obstacles to the development of effective and personalized immunotherapeutic strategies. CONCLUSION Further research is needed to clarify the mechanisms governing immune cell behavior within the BC TME and to advance precision immunotherapy. Such insights will lay the foundation for individualized treatment approaches, ultimately improving patient outcomes and quality of life (QoL).
Collapse
Affiliation(s)
- Rui Li
- Shandong Provincial Hospital, Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Wei Lv
- Department of General SurgeryShandong Provincial HospitalJinanShandongChina
| | | | - Na Chen
- Department of Internal MedicineShandong Provincial HospitalJinanShandongChina
| |
Collapse
|
4
|
Cai J, Zhang P, Cai Y, Zhu G, Chen S, Song L, Du J, Wang B, Dai W, Zhou J, Fan J, Yu Y, Dai Z. Lactylation-Driven NUPR1 Promotes Immunosuppression of Tumor-Infiltrating Macrophages in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413095. [PMID: 40305758 PMCID: PMC12120759 DOI: 10.1002/advs.202413095] [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] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 04/13/2025] [Indexed: 05/02/2025]
Abstract
While checkpoint immunotherapy effectively mobilizes T-cell responses against tumors, its success in hepatocellular carcinoma (HCC) is frequently undermined by immunosuppressive myeloid cells within the tumor microenvironment. This study investigates the role of nuclear protein 1 (NUPR1), a gene prominently expressed in tumor-associated macrophages (TAMs), in mediating this suppression and influencing immunotherapy outcomes. Through comprehensive analysis of single-cell RNA sequencing (scRNA-seq) datasets and functional assays in vitro and in vivo, NUPR1 is identified as a critical regulator within TAMs. The upregulation of NUPR1 is associated with enhanced M2 macrophage polarization and increased expression of immune checkpoints PD-L1 and SIRPA, resulting in CD8+ T cell exhaustion and a diminished response to immunotherapy. Mechanistically, NUPR1 inhibits the ERK and JNK signaling pathways, thereby creating an immunosuppressive milieu conducive to tumor progression. Additionally, tumor-derived lactate is shown to upregulate NUPR1 expression in macrophages via histone lactylation, perpetuating a feedback loop that intensifies immune suppression. Pharmacological targeting of NUPR1 reverses M2 polarization, curtails tumor growth, and augments the efficacy of PD-1 blockade in preclinical models, positioning NUPR1 as both a potential biomarker for immunotherapy responsiveness and a therapeutic target to boost immunotherapy efficacy in HCC.
Collapse
Affiliation(s)
- Jialiang Cai
- Liver Cancer InstituteZhongshan HospitalFudan UniversityShanghai200032China
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityMinistry of EducationShanghai200032China
| | - Peiling Zhang
- Liver Cancer InstituteZhongshan HospitalFudan UniversityShanghai200032China
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityMinistry of EducationShanghai200032China
| | - Yufan Cai
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Guiqi Zhu
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Department of Liver Surgery and TransplantationZhongshan HospitalFudan UniversityShanghai200032China
- Research Unit of Liver Cancer Recurrence and MetastasisChinese Academy of Medical SciencesBeijing100000China
| | - Shiping Chen
- Liver Cancer InstituteZhongshan HospitalFudan UniversityShanghai200032China
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityMinistry of EducationShanghai200032China
| | - Lina Song
- Liver Cancer InstituteZhongshan HospitalFudan UniversityShanghai200032China
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityMinistry of EducationShanghai200032China
| | - Junxian Du
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Biao Wang
- Department of Radiation OncologyZhongshan HospitalFudan UniversityShanghai200032China
| | - Weixing Dai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jian Zhou
- Department of Liver Surgery and TransplantationZhongshan HospitalFudan UniversityShanghai200032China
- Research Unit of Liver Cancer Recurrence and MetastasisChinese Academy of Medical SciencesBeijing100000China
| | - Jia Fan
- Department of Liver Surgery and TransplantationZhongshan HospitalFudan UniversityShanghai200032China
- Research Unit of Liver Cancer Recurrence and MetastasisChinese Academy of Medical SciencesBeijing100000China
| | - Yiyi Yu
- Department of Medical OncologyZhongshan HospitalFudan University180 Fenglin RoadShanghai200032China
- Cancer CenterZhongshan Hospital Fudan UniversityShanghai200032China
| | - Zhi Dai
- Liver Cancer InstituteZhongshan HospitalFudan UniversityShanghai200032China
- State Key Laboratory of Genetic EngineeringFudan UniversityShanghai200032China
- Key Laboratory of Carcinogenesis and Cancer InvasionFudan UniversityMinistry of EducationShanghai200032China
| |
Collapse
|
5
|
Gao Y, Li B, Jin Y, Cheng J, Tian W, Ying L, Hong L, Xin S, Lin B, Liu C, Sun X, Zhang J, Zhang H, Xie J, Deng X, Dai X, Liu L, Zheng Y, Zhao P, Yu G, Fang W, Bao X. Spatial multi-omics profiling of breast cancer oligo-recurrent lung metastasis. Oncogene 2025:10.1038/s41388-025-03388-y. [PMID: 40234722 DOI: 10.1038/s41388-025-03388-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025]
Abstract
Primary breast cancer (BC) and metastatic tumors exhibit distinct tumor microenvironment (TME) ecosystems, and the heterogeneity of the TME of BC poses challenges to effective therapies. Evaluating the TME at the single-cell and spatial profiles offers potential for more precise treatments. However, due to the challenge of obtaining surgical specimens of both primary BC and oligo-recurrent lung metastasis simultaneously for high-resolution spatial analysis, the TME of lung-specific metastases using paired samples remains largely unexplored. In this study, we developed a comprehensive strategy using imaging mass cytometry (IMC), spatial proteomics, single-nucleus RNA-seq (snRNA-seq) and multiplex immunofluorescence to explore the spatial topology of lung-specific metastasis and the underlying biological mechanisms based on formalin-fixed paraffin-embedded (FFPE) samples from BC and paired lung metastasis. A total of 250,600 high-quality cells with spatial information revealed by IMC depicted the spatial differences in the TME between BC and lung metastasis. A significant increase in HLA-DR+ epithelial cells, endothelial cells and exhausted T cells was detected in lung metastases compared to primary sites, with this difference accentuated in the triple-negative subtype. Moreover, a distinct cellular hub comprising endothelial cells and HLA-DR+ epithelial cells implies the potential promising effect of anti-angiogenic therapy and immunotherapy in BC with lung metastasis, which was further validated by multiplex immunofluorescence analysis. Spatial proteomics further explored the underlying mechanism of TME components identified by IMC analysis. snRNA-seq validated the enrichment of endothelial cells in lung metastasis than that in BC at a whole FFPE slide level. In conclusion, this study determines the spatial multi-omics profiling of TME components at a single-cell resolution using paired samples of primary BC and lung oligo-metastasis. The comprehensive analysis may contribute to the development of therapeutic options.
Collapse
Affiliation(s)
- Yang Gao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Bin Li
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yuzhi Jin
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Jinlin Cheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Weihong Tian
- Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213001, China
| | - Lixiong Ying
- Department of Medical Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Libing Hong
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Shan Xin
- Department of Genetics, Yale School of Medicine, New Haven, USA
| | - Bo Lin
- Innovation Centre for Information, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, 310053, China
| | - Chuan Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Xuqi Sun
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Jun Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - 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
| | - 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
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Lulu Liu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Guangchuan Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
6
|
Ge Q, Meng J, Wang Z, Anwaier A, Lu J, Tian X, Wang Y, Yang J, Zhang H, Ye D, Xu W. Spatially segregated APOE + macrophages restrict immunotherapy efficacy in clear cell renal cell carcinoma. Theranostics 2025; 15:5312-5336. [PMID: 40303328 PMCID: PMC12036886 DOI: 10.7150/thno.109097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Immunotherapy has revolutionized cancer treatment and holds great potential for them, including metastatic clear cell renal cell carcinoma (ccRCC). However, immune resistance remains a major obstacle, limiting its efficacy and durability. Understanding the mechanisms of immune tolerance in the tumor microenvironment (TME) is pivotal for overcoming these challenges and enhancing therapeutic outcomes. Methods: Over 2000 samples, including a real-world cohort of 230 advanced ccRCC patients treated with immune checkpoint blockade (ICB) were analyzed. Single-cell RNA sequencing data from 13 tumor regions were categorized into ICB-exposed, ICB-resistant, and ICB-responsive groups. Multiple robust algorithms and multiplex immunofluorescence were used to explore TME composition and macrophage heterogeneity. Spatial communication dynamics were further investigated. In vitro experiments were performed to evaluate the impact of SPP1 on 786-O and 769-P cells. Co-culture experiments with THP-1-derived macrophages, followed by Western blot, flow cytometry, and functional assays, were performed to investigate SPP1-mediated macrophage polarization and its impact on tumor progression. Results: The results revealed an elevated presence of Apolipoprotein E (APOE)+ macrophages in ICB-resistant ccRCC. Notably, higher APOE+ macrophage proportion indicated shorter prognosis and worse response to ICB (P < 0.001). Elevated expression of CCAAT Enhancer Binding Protein Delta (CEBPD) was markedly linked to several immunosuppressive pathways, hindering T cell recruitment, promoting exhaustion, ultimately diminishing poorer prognosis and worse ICB efficacy. Meanwhile, upregulated Secreted Phosphoprotein 1 (SPP1) significantly enhances the proliferation, clonal formation, and migration of ccRCC cells. Tumor-derived SPP1. Additionally, SPP1 signaling from malignant cells appeared to recruit APOE+ macrophages to tumor margins, and promotes macrophage polarization into APOE+ M2-like macrophages. In the vicinity of the tumor, these APOE+ macrophages shape immunosuppressive TME by releasing abundant TGF-β signals, limiting anti-tumor effector T cells activity in ICB-resistant tumors, and contributing to tumor progression. Conclusion: This study reveals the critical role of APOE+ macrophages in promoting immune suppression and resistance to ICB therapy in ccRCC. By promoting T cell exhaustion and immunosuppressive signaling, particularly via localized TGF-β, these spatially segregated macrophages undermine treatment efficacy. Targeting APOE+ macrophages, especially in conjunction with ICB, presents a promising strategy to overcome immune resistance and enhance outcomes for ccRCC patients.
Collapse
Affiliation(s)
- Qintao Ge
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, 230022, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, 230022, P.R. China
| | - Zhongyuan Wang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jiahe Lu
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jianfeng Yang
- Department of Urology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, P.R. China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| |
Collapse
|
7
|
Liu Z, Mao Y, Wang S, Zheng H, Yang K, Yang L, Huang P. A bibliometric and visual analysis of the impact of senescence on tumor immunotherapy. Front Immunol 2025; 16:1566227. [PMID: 40292294 PMCID: PMC12021824 DOI: 10.3389/fimmu.2025.1566227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Recently, many studies have focused on the relationship between senescence and immunotherapy in cancer treatment. However, relatively few studies have examined the intrinsic links between the three. Whether these studies can act synergistically in the fight against cancer and the specific links between them are still unclear. Methods We extracted, quantified, and visualized data from the literature (n = 2396) for the period 2004-2023 after rigorous quality control using citespace, GraphPad Prism, the R software package, and VOSviewer. Results Linear fit analyses were generated to predict the number of annual publications and citations as a function of the top-performing authors, journals, countries, and affiliations academically over the past two decades such as Weiwei, Aging-us, China, and the UT MD Anderson Cancer Center. Vosviewer-based hierarchical clustering further categorized study characteristics into six clusters, including two major clusters of immunotherapy research, immunosenescence-related research factors, and timeline distributions suggesting that cellular senescence and tumor progression is a relatively new research cluster that warrants further exploration and development. Study characterization bursts and linear regression analyses further confirmed these findings and revealed other important results, such as aging (a = 1.964, R² = 0.6803) and immunotherapy (a = 16.38, R² = 0.8812). Furthermore, gene frequency analysis in this study revealed the most abundant gene, APOE, and SIRT1-7 proteins. Conclusion The combination of aging therapies with tumor immunotherapies is currently in its preliminary stages. Although senescence has the greatest impact on ICB therapies, mechanistic investigations, and drug development for APOE and sirt1-7 (Sirtuins family) targets may be the key to combining senescence therapies with immunotherapies in the treatment of tumors.
Collapse
Affiliation(s)
- Zixu Liu
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yuchen Mao
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Shukai Wang
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Haoyu Zheng
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kangping Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Liang Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Huang
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Zhang J, Zhang X, Wu R, Dong CS. Unveiling purine metabolism dysregulation orchestrated immunosuppression in advanced pancreatic cancer and concentrating on the central role of NT5E. Front Immunol 2025; 16:1569088. [PMID: 40236698 PMCID: PMC11996659 DOI: 10.3389/fimmu.2025.1569088] [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: 01/31/2025] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Background The dismal efficacy of immunotherapy for Pancreatic cancer (PC) can be predominantly ascribed to its distinctive cold-tumor properties. The by-products of purine metabolic reprogramming are extensively engaged in tumor immune modulation, influencing the functions and recruitment of immune cells and molding an immune microenvironment that is propitious for tumor growth. Methods We harnessed single-cell transcriptomics and spatial transcriptomics to concurrently analyze the purine metabolism (PM) features of the PC microenvironment. We quantitatively appraised the PM traits of diverse cell subsets via scoring algorithms such as AUCell and Ucell. Moreover, cell development and cell-cell interaction analysis elucidated the alterations in TME induced by PM dysregulation. Additionally, we defined the PM disorder characteristics of PC patients and utilized this to assess the immune phenotypes and prognoses of the patient population. Also, we identified the crucial intermediate genes that impact PM reprogramming and the establishment of an immunosuppressive environment within the TME of PC, and validated them through spatial sectioning and cell co-culture experiments. Results Multi - dimensional transcriptome data elucidated the unique heterogeneity of PM in the PC microenvironment, which manifested that tumor cells and fibroblasts demonstrating higher PM scores in the TME. Cellchat analysis revealed that malignant cells with elevated PM expression were concomitantly associated with frequent interactions with CAFs as well as high expression of ligand-receptor pairs and transcription factors. Spatial data further corroborated this finding. Furthermore, the newly constructed PM disorder criteria indicated that patients with high PM levels were associated with a lack of response to immunotherapy and an immunosuppressive microenvironment. Finally, this study identified the singular role of NT5E in the immunosuppression resulting from PM reprogramming in PC. CCK8 and invasion experiments following the co-culture model demonstrated that intervention targeting NT5E could reverse the augmented malignancy of PC induced by co-cultured CAFs. NT5E is potentially a key target for reversing the "stiff-cancer" characteristics of PC. Conclusion This study demonstrates that PM metabolic disorders could impinge upon tumor immunotherapy and exacerbate the immunosuppression engendered by the progression of PC fibrosis. Therapeutic strategies targeting PM or NT5E may offer a ray of hope for patients with advanced PDAC.
Collapse
Affiliation(s)
- Junqian Zhang
- Henan Key Laboratory of Cancer Epigenetics; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaobo Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ruixin Wu
- Preclinical Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-Sheng Dong
- Cancer Institute of Traditional Chinese Medicine/Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
10
|
Xie J, Xie Y, Tan W, Ye Y, Ou X, Zou X, He Z, Wu J, Deng X, Tang H, He L, Li K, Luo P, Bai K, Huang G, Li J. Deciphering the role of ELAVL1: Insights from pan-cancer multiomics analyses with emphasis on nasopharyngeal carcinoma. J Transl Int Med 2025; 13:138-155. [PMID: 40443402 PMCID: PMC12116272 DOI: 10.1515/jtim-2025-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025] Open
Abstract
Background and Objectives Cancer continues to be a predominant cause of mortality worldwide, underscoring the critical need to identify and develop novel biomarkers to improve prognostic accuracy and therapeutic approaches. The dysregulation of ELAVL1 is linked to various diseases, including cancer. Nevertheless, its role across different cancer types remains insufficiently investigated. Methods We conducted a systematic investigation into the expression patterns, prognostic significance, genomic alterations, modifications, and functional implications of ELAVL1 in pan-cancer types. Besides, we performed in vitro and in vivo experiments to confirm the role of ELAVL1 in nasopharyngeal carcinoma (NPC). Results By utilizing multi-omics datasets, we found obvious overexpression of ELAVL1 in various cancer types at both the mRNA and protein levels, with predominant expression in malignant cells. Survival analysis revealed that increased ELAVL1 expression was linked to unfavorable outcomes in certain cancers; however, its effect difers among various cancer types. Additionally, we found that the genomic alterations and modifications of ELAVL1 were related to tumor progression. We discovered that ELAVL1 was elevated in NPC tissues. In addition, survival analysis indicated that NPC patients with higher ELAVL1 expression had worse prognoses. Functional assays demonstrated that ELAVL1 suppression led to decreased proliferation and migration in NPC cell lines. Moreover, ELAVL1 knockdown effectively inhibited NPC progression in the lymph node and lung metastasis models. Conclusions In summary, ELAVL1 exhibits diverse and complex involvement in tumor progression. Targeting it might inhibit tumor progression, making it a promising biomarker and therapeutic target for enhancing cancer treatment outcomes.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Yi Xie
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Wencheng Tan
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Yimeng Ye
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Xueqi Ou
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiong Zou
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Zhiqing He
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Jiarong Wu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Xinpei Deng
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Hailin Tang
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Longjun He
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Kailai Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Kunhao Bai
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Guoxian Huang
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| | - Jianjun Li
- Department of Oncology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
- Department of Endoscopy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, China
| |
Collapse
|
11
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
12
|
He D, Yang Z, Zhang T, Luo Y, Peng L, Yan J, Qiu T, Zhang J, Qin L, Liu Z, Sun M. Multi-omics and machine learning-driven CD8 + T cell heterogeneity score for head and neck squamous cell carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102413. [PMID: 40027882 PMCID: PMC11869859 DOI: 10.1016/j.omtn.2024.102413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/03/2024] [Indexed: 03/05/2025]
Abstract
The heterogeneity of head and neck squamous cell carcinoma (HNSCC) poses a significant challenge to treatment, underscoring the urgent need for more precise and personalized therapeutic approaches. CD8+ T cells, integral components of the tumor immune microenvironment, have emerged as key targets for immunotherapy. Our research has established a correlation between a decrease in CD8+ T cell score and a poor clinical prognosis, highlighting the prognostic value of this biomarker. By analyzing the gene expression related to CD8+ T cells, we have differentiated HNSCC into cold and hot tumor subtypes, uncovering disparities in clinical prognosis and responses to immunotherapy. Utilizing eight machine learning methods, we identified the key gene OLR1. Single-cell analysis of HNSCC tissues and peripheral blood, along with spatial transcriptome analysis, revealed that OLR1 predominantly functions in macrophages, modulating the immune microenvironment of HNSCC. The expression level of OLR1 may serve as a predictive marker for immunotherapy responses. Moreover, drug sensitivity analysis and molecular docking studies have indicated that simvastatin and pazopanib are potential inhibitors of OLR1. These findings suggest that simvastatin and pazopanib could open up innovative potential therapeutic avenues for individuals with HNSCC.
Collapse
Affiliation(s)
- Di He
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tian Zhang
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yaxian Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Lianjie Peng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jiatao Yan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tao Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jingyu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Luying Qin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Zhichao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
13
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
14
|
Xin Y, Ma Q, Deng Q, Wang T, Wang D, Wang G. Analysis of single-cell and spatial transcriptomics in TNBC cell-cell interactions. Front Immunol 2025; 16:1521388. [PMID: 40079015 PMCID: PMC11897037 DOI: 10.3389/fimmu.2025.1521388] [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: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly malignant tumor in women, characterized by high morbidity, mortality, and recurrence rates. Although surgical treatment, radiotherapy, and chemotherapy are the mainstays of current treatment methods, the high heterogeneity of TNBC results in unsatisfactory outcomes with low 5-year survival rates. Rapid advancements in omics technology have propelled the understanding of TNBC molecular biology. The emergence of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) has significantly enhanced knowledge of tumor heterogeneity and the distribution, functionality, and intercellular interactions of various cell types within the tumor microenvironment, including tumor cells, T cells, B cells, macrophages, and fibroblasts. The present study provides an overview of the technical characteristics of scRNA-seq and ST, highlighting their applications in exploring TNBC heterogeneity, cell spatial distribution patterns, and intercellular interactions. This review aims to enhance the comprehension of TNBC at the cellular level for the development of effective therapeutic targets.
Collapse
Affiliation(s)
- Yan Xin
- Department of Anesthesiology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Qiji Ma
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Qiang Deng
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Tielin Wang
- College of Acupuncture, Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Gang Wang
- Department of Breast and Thyroid Surgery, The Affliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
15
|
Zhang J, Li L, Tang A, Wang C, Wang Y, Hu Y, He G, Liao W, Zhou R. Pan-cancer analysis of the transcriptional expression of histone acetylation enzymes in solid tumors defines a new classification scheme for gliomas. Front Immunol 2025; 15:1523034. [PMID: 39906742 PMCID: PMC11790639 DOI: 10.3389/fimmu.2024.1523034] [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: 11/05/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction The altered expression of genes encoding histone acetyltransferases (HATs) and histone deacetylases (HDACs) has been implicated in the tumorigenesis and progression of various solid tumors. However, systematic characterization of the transcriptomic landscape and clinical relevance of HATs and HDACs in pan-cancer contexts remains lacking. Methods Transcriptome and clinical data of 9,483 patients across 31 tumor types from The Cancer Genome Atlas were collected for systematic pan-cancer analysis. Additional glioma-specific datasets (Chinese Glioma Genome Atlas, GlioVis, GSE43378, and GSE182109) were also collected to validate the transcriptional characteristics of HATs and HDACs in gliomas. Consensus clustering analysis was applied to identify distinct expression patterns of HATs and HDACs. Results Based on the transcriptomic data of 25 genes encoding 9 HATs and 16 HDACs, we identified five major subtypes across 31 cancer types (AC-I to AC-V). Notably, the AC-V subtype comprised over 95% of glioma patients, suggesting glioma patients exhibited distinct expression patterns of histone acetylation-modifying enzymes compared to patients with other solid tumors. Therefore, we re-conducted the consensus clustering analysis specifically within the context of gliomas and identified five subtypes, denoted "AC-GI" to "AC-GV", which were characterized by differences in HATs/HDACs expression patterns, biological and immune status, genetic alterations, and clinical outcomes. The AC-GII patients exhibited the best prognosis and were sensitive to temozolomide, while AC-GV patients had the poorest prognosis and the lowest sensitivity to temozolomide among all subtypes. Moreover, based on the Connectivity Map database analysis and experimental verification, we identified several pan-HDAC inhibitors that could serve as sensitizers for temozolomide therapy in AC-GV patients, such as panobinostat and scriptaid. Considering the distinctive clinical characteristics of patients with AC-GII and AC-GV, we constructed the "ACG score" model capable of effectively recognizing patients with these subtypes and predicting patient prognosis. Conclusion Herein, we established novel biologically and clinically relevant molecular classifications for pan-solid tumors and gliomas based on transcriptional expression profiles of HATs and HDACs. Moreover, the ACG score model, calculated by the transcriptional expression of 29 genes, was not only an independent prognostic factor for glioma patients, but can also provide valuable references for promoting more effective therapeutic strategies.
Collapse
Affiliation(s)
- Junhao Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lingbo Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Aiwei Tang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chucheng Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yupeng Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongqi Hu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangting He
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Cancer Center, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
- Foshan Key Laboratory of Translational Medicine in Cancer, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Wang L, Sun M, Li J, Wan L, Tan Y, Tian S, Hou Y, Wu L, Peng Z, Hu X, Zhang Q, Huang Z, Han M, Peng S, Pan Y, Ren Y, Zhang M, Chen D, Liu Q, Li X, Qin ZY, Xiang J, Li M, Zhu J, Chen Q, Luo H, Wang S, Wang T, Li F, Bian XW, Wang B. Intestinal Subtype as a Biomarker of Response to Neoadjuvant Immunochemotherapy in Locally Advanced Gastric Adenocarcinoma: Insights from a Prospective Phase II Trial. Clin Cancer Res 2025; 31:74-86. [PMID: 39495175 DOI: 10.1158/1078-0432.ccr-24-2436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/22/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE Neoadjuvant immunochemotherapy (NAIC) markedly induces pathologic regression in locally advanced gastric adenocarcinoma. However, specific biomarkers are still lacking to effectively identify the beneficiary patients for NAIC. PATIENTS AND METHODS A prospective, single-arm, phase II study was conducted to treat locally advanced gastric adenocarcinoma with NAIC (NCT05515796). Correlation between clinicopathologic characteristics and neoadjuvant efficacy was investigated. Bulk RNA sequencing data from 104 samples (from 75 patients in two independent cohorts) and single-cell RNA sequencing data from 105 treatment-naïve gastric adenocarcinomas were comprehensively analyzed to decipher the association of epithelial and microenvironmental characteristics and clinical responses. RESULTS The prespecified primary endpoints were achieved: pathologic complete regression rate was 30%, major pathologic regression rate was 43%, and the regimen was well tolerated. Analysis of baseline clinical-pathologic parameters revealed the intestinal subtype of Lauren's classification as a key feature stratifying patients with increased sensitivity to NAIC. Mechanistically, an increased pool of DNA damage repair-active cancer cells and enrichment of CLEC9A+ dendritic cells in the tumor microenvironment were associated with enhanced responsiveness of the intestinal subtype gastric adenocarcinoma to NAIC. More importantly, an intestinal subtype-specific signature model was constructed by the machine learning algorithm NaiveBayes via integrating the transcriptomic features of both DNA damage repair-active cancer cells and CLEC9A+ dendritic cells, which accurately predicted the efficacy of NAIC in multiple independent gastric adenocarcinoma cohorts. CONCLUSIONS Intestinal subtype is a histologic biomarker of enhanced sensitivity of gastric adenocarcinoma to NAIC. The intestinal subtype-specific signature model is applicable to guide NAIC for patients with locally advanced gastric adenocarcinoma.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Mengting Sun
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Jinyang Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Linghong Wan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Yuting Tan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- School of Medicine, Chongqing University, Chongqing, P.R. China
| | - Shuoran Tian
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Yongying Hou
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Linyu Wu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Ziyi Peng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Xiao Hu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- School of Medicine, Chongqing University, Chongqing, P.R. China
| | - Qihua Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Zening Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, P.R. China
| | - Mengyi Han
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Shiyin Peng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- School of Medicine, Chongqing University, Chongqing, P.R. China
| | - Yuwei Pan
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- School of Medicine, Chongqing University, Chongqing, P.R. China
| | - Yuanfeng Ren
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Mengsi Zhang
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Dongfeng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Qin Liu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Xianfeng Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Zhong-Yi Qin
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Junyv Xiang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Mengxia Li
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Jianwu Zhu
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Qiyue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, P.R. China
| | - Huiyan Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Tao Wang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Fan Li
- Division of Gastric and Colorectal Surgery, Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
| | - Bin Wang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- Institute of Pathology and Southwest Cancer Center, and Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, P.R. China
- Jinfeng Laboratory, Chongqing, P.R. China
| |
Collapse
|
17
|
Peng S, Wu M, Yan Q, Xu G, Xie Y, Tang G, Lin J, Yuan Z, Liang X, Yuan Z, Weng J, Bai L, Wang X, Yu H, Huang M, Luo Y, Liu X. Disrupting EDEM3-induced M2-like macrophage trafficking by glucose restriction overcomes resistance to PD-1/PD-L1 blockade. Clin Transl Med 2025; 15:e70161. [PMID: 39754316 DOI: 10.1002/ctm2.70161] [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: 09/08/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Immunotherapy is beneficial for some colorectal cancer (CRC) patients, but immunosuppressive networks limit its effectiveness. Cancer-associatedfibroblasts (CAFs) are significant in immune escape and resistance toimmunotherapy, emphasizing the urgent need for new treatment strategies. METHODS Flow cytometric, Western blotting, proteomics analysis, analysis of public database data, genetically modified cell line models, T cell coculture, crystal violetstaining, ELISA, metabonomic and clinical tumour samples were conducted to assess the role of EDEM3 in immune escape and itsmolecular mechanisms. We evaluated theeffects of FMD plus 2-DG on antitumour immunity using multipleximmunofluorescence, flow cytometry, cytokine profiling, TUNEL assays, xenografttumours, and in vivo studies. RESULTS We show thatCAFs upregulate PD-L1 glycosylation and contribute to immune evasion byglycosyltransferase EDEM3. Additionally, EDEM3 plays a role in tumour immunityduring tumour progression. However, the EDEM3-mediated upregulation of PD-L1 expression underpins PD-1/PD-L1 blockade resistance in vivo. This finding contradictsthe previous trend that positive PD-L1 expression indicates a strong responseto PD-1/PD-L1 blockade. Mechanistically, high-EDEM3 expression facilitates M2-like This finding contradictsthe previous trend that positive PD-L1 expression indicates a strong responseto PD-1/PD-L1 blockade.Mechanistically, polarizationand chemotactic migration of macrophages, which are enriched in theperipheral region of tumours compared to thecore region, precluding access of CD8+ T cells to tumourfoci. Furthermore, we EDEM3 predominantly activates the recruited M2-like macrophagesvia a glucose metabolism-dependent mechanism. Manipulationof glucose utilization by a fasting-mimicking diet(FMD) plus 2-DG treatmentsynergistically with PD-1 antibody elicits potent antitumour activity byeffectively decreasing tumour glycosylated PD-L1 expression, augmenting the CD8+effector T cell infiltration and activation while concurrently reducing the infiltration.TheCAFs-EDEM3-M2-like macrophage axis plays a critical role in promotingimmunotherapy resistance. infiltration.TheCAFs-EDEM3-M2-like macrophage axis plays a critical role in promotingimmunotherapy resistance. CONCLUSIONS Our study suggests that blocking EDEM3-induced M2-like macro phage trafficking by FMD plus 2-DG is a promising and effective strategy to overcomeresistance to checkpoint blockade therapy offeringhope for improved treatment outcomes. KEY POINTS Cancer-associated fibroblasts (CAFs) can enhance PD-L1 glycosylation through the glycosyltransferase EDEM3, contributing to immune evasion during tumour progression. EDEM3 predominantly activates the recruit M2-like macrophages via a glucose metabolism-dependent mechanism. Blocking glucose utilization antagonizes recruiting and polarizing M2-like macrophages synergistically with PD-1 antibody to improve anticancer immunity.
Collapse
Affiliation(s)
- Shaoyong Peng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
| | - Minshan Wu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Yan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gaopo Xu
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yumo Xie
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guannan Tang
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinxin Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zixu Yuan
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoxia Liang
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ze Yuan
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingrong Weng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liangliang Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaolin Wang
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huichuan Yu
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meijin Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoxia Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- School of Life Sciences, Innovation Center of the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Xiao Z, Liu X, Wang Y, Jiang S, Feng Y. Comprehensive analysis of single-cell and bulk RNA sequencing reveals postoperative progression markers for non-muscle invasive bladder cancer and predicts responses to immunotherapy. Discov Oncol 2024; 15:649. [PMID: 39532830 PMCID: PMC11557814 DOI: 10.1007/s12672-024-01548-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Non-muscle-invasive bladder cancer (NMIBC) is renowned for its high recurrence, invasiveness, and poor prognosis. Consequently, developing new biomarkers for risk assessment and investigating innovative therapeutic targets postoperative in NMIBC patients are crucial to aid in treatment planning. APPROACHES Differential gene expression analysis was performed using multiple Gene Expression Omnibus (GEO) datasets to identify differentially expressed genes (DEGs) between NMIBC and normal tissue, as well as between NMIBC and muscle-invasive bladder cancer (MIBC). Functional enrichment analysis was conducted based on the DEGs identified. Subsequently, prognosis-related genes were selected using Kaplan-Meier (KM) analysis and Cox regression analysis. The Boruta algorithm was utilized to further screen for core DEGs related to postoperative progression in NMIBC based on the aforementioned prognosis-related genes. Single-cell and clinical correlation studies were performed to verify their expression across various stages of bladder cancer. To investigate the link between core genes and the immune microenvironment, single-sample gene set enrichment analysis (ssGSEA) was utilized, and Receiver Operating Characteristic (ROC) and KM analyses were performed to confirm predictive power for immune therapy outcomes. Machine learning (ML) models were constructed using the DepMap dataset to predict the efficacy of core gene inhibitors in treating bladder cancers. The prognostic performance of the core genes was evaluated using ROC curve analysis. An online prediction tool was developed based on the core genes to provide prognostic predictions. Finally, RT-qPCR, CCK-8, and Transwell assays were used to verify the pro-tumor effects of the GINS2 in bladder cancer. RESULTS A total of 70 DEGs were identified, among which 11 prognostic genes were obtained through KM analysis, and an additional 8 prognostic genes were obtained through COX analysis. The Boruta algorithm selected AURKB, GINS2, and UHRF1 as the three core DEGs. Single-cell and clinical variable correlation analyses indicated that the core genes promoted the progression of bladder cancer. The analysis of immune infiltration revealed a strong positive association between the core genes and both activated CD4 T cells and Type 2 helper T cells. Two random forest (RF) models were constructed to effectively predict the treatment effect of bladder cancer after targeted inhibition of AURKB and GINS2. In addition, an online nomogram tool was developed to effectively predict the risk of postoperative progression in NMIBC patients undergoing TURBT. Finally, RT-qPCR, CCK8, and Transwell assays showed that GINS2 promoted the growth and progression of bladder cancer. CONCLUSION AURKB, GINS2, and UHRF1 have the potential to enhance postoperative management of NMIBC patients undergoing transurethral resection of bladder tumor (TURBT) and can predict immunotherapy response, establishing them as promising therapeutic targets.
Collapse
Affiliation(s)
- Zhiliang Xiao
- Department of Urology, The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Xin Liu
- Department of Urology, The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Yuan Wang
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Sicong Jiang
- Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China.
| | - Yan Feng
- Department of Urology, The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, China.
| |
Collapse
|
19
|
Pan L, Huang H, Zhang P, Li H, Lu L, Wei M, Zheng P, Wang Q, Guo J, Qin Y. Immunofluorescence-Verified Sphingolipid Signatures Indicate Improved Prognosis in Liver Cancer Patients. J Cancer 2024; 15:6239-6255. [PMID: 39513103 PMCID: PMC11540515 DOI: 10.7150/jca.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy, with its pathogenesis involving a complex interplay of molecular mechanisms, including cell cycle dysregulation, evasion of apoptosis, enhanced angiogenesis, and aberrant immune responses. Precision medicine approaches that target specific molecular subtypes through multi-omics integration hold promise for improving patient survival. Among the various molecular players, sphingolipids have emerged as pivotal regulators of tumor growth and apoptosis, positioning them as key targets in the search for novel anticancer therapies. Methods: To identify critical genes involved in sphingolipid metabolism (SM), we employed the AUCell algorithm and correlation analysis in conjunction with scRNA-seq data. A robust prognostic risk model was developed using Cox proportional hazards and Lasso regression, and its predictive performance was validated using an independent cohort from the International Cancer Genome Consortium (ICGC). The model's evaluation also incorporated analyses of the tumor microenvironment (TME), immunotherapy responses, mutational landscape, and pathway enrichment across different risk strata. Finally, we conducted multiplex immunofluorescence assays to investigate the functional role of ZC3HAV1 in HCC. Results: Our analysis yielded a 9-gene signature risk model with strong prognostic capabilities, effectively stratifying HCC patients into high- and low-risk groups, with significant differences in survival outcomes. Notably, the model revealed distinct variations in the immune microenvironment and responsiveness to immunotherapy between the risk groups. Further experimental validation identified ZC3HAV1 as a key gene, with multiplex immunofluorescence suggesting its involvement in promoting malignant progression in HCC through modulation of the epithelial-mesenchymal transition (EMT). Conclusion: This sphingolipid metabolism-based prognostic model is not only predictive of survival in HCC but also indicative of immunotherapy efficacy in certain patient subsets. Our findings underscore the crucial role of sphingolipid metabolism in shaping the immune microenvironment, offering new avenues for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Lujuan Pan
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
| | - Huijuan Huang
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Hua Li
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Libai Lu
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Mingwei Wei
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Pin Zheng
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Junyu Guo
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Yueqiu Qin
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, Guangxi, China
| |
Collapse
|
20
|
Zhang B, Liu J, Mo Y, Zhang K, Huang B, Shang D. CD8 + T cell exhaustion and its regulatory mechanisms in the tumor microenvironment: key to the success of immunotherapy. Front Immunol 2024; 15:1476904. [PMID: 39372416 PMCID: PMC11452849 DOI: 10.3389/fimmu.2024.1476904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
A steady dysfunctional state caused by chronic antigen stimulation in the tumor microenvironment (TME) is known as CD8+ T cell exhaustion. Exhausted-like CD8+ T cells (CD8+ Tex) displayed decreased effector and proliferative capabilities, elevated co-inhibitory receptor generation, decreased cytotoxicity, and changes in metabolism and transcription. TME induces T cell exhaustion through long-term antigen stimulation, upregulation of immune checkpoints, recruitment of immunosuppressive cells, and secretion of immunosuppressive cytokines. CD8+ Tex may be both the reflection of cancer progression and the reason for poor cancer control. The successful outcome of the current cancer immunotherapies, which include immune checkpoint blockade and adoptive cell treatment, depends on CD8+ Tex. In this review, we are interested in the intercellular signaling network of immune cells interacting with CD8+ Tex. These findings provide a unique and detailed perspective, which is helpful in changing this completely unpopular state of hypofunction and intensifying the effect of immunotherapy.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinming Liu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuying Mo
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kexin Zhang
- Central Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Westlake University, Hangzhou, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Hawsawi YM, Khoja B, Aljaylani AO, Jaha R, AlDerbi RM, Alnuman H, Khan MI. Recent progress and applications of single-cell sequencing technology in breast cancer. Front Genet 2024; 15:1417415. [PMID: 39359479 PMCID: PMC11445024 DOI: 10.3389/fgene.2024.1417415] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technology enables the precise analysis of individual cell transcripts with high sensitivity and throughput. When integrated with multiomics technologies, scRNA-seq significantly enhances the understanding of cellular diversity, particularly within the tumor microenvironment. Similarly, single-cell DNA sequencing has emerged as a powerful tool in cancer research, offering unparalleled insights into the genetic heterogeneity and evolution of tumors. In the context of breast cancer, this technology holds substantial promise for decoding the intricate genomic landscape that drives disease progression, treatment resistance, and metastasis. By unraveling the complexities of tumor biology at a granular level, single-cell DNA sequencing provides a pathway to advancing our comprehension of breast cancer and improving patient outcomes through personalized therapeutic interventions. As single-cell sequencing technology continues to evolve and integrate into clinical practice, its application is poised to revolutionize the diagnosis, prognosis, and treatment strategies for breast cancer. This review explores the potential of single-cell sequencing technology to deepen our understanding of breast cancer, highlighting key approaches, recent advancements, and the role of the tumor microenvironment in disease plasticity. Additionally, the review discusses the impact of single-cell sequencing in paving the way for the development of personalized therapies.
Collapse
Affiliation(s)
- Yousef M Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Basmah Khoja
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | | | - Raniah Jaha
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Rasha Mohammed AlDerbi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Huda Alnuman
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Mohammed I Khan
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Shen G, Wang Q, Li Z, Xie J, Han X, Wei Z, Zhang P, Zhao S, Wang X, Huang X, Xu M. Bridging Chronic Inflammation and Digestive Cancer: The Critical Role of Innate Lymphoid Cells in Tumor Microenvironments. Int J Biol Sci 2024; 20:4799-4818. [PMID: 39309440 PMCID: PMC11414386 DOI: 10.7150/ijbs.96338] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/09/2024] [Indexed: 09/25/2024] Open
Abstract
The incidence and mortality of digestive system-related cancers have always been high and attributed to the heterogeneity and complexity of the immune microenvironment of the digestive system. Furthermore, several studies have shown that chronic inflammation in the digestive system is responsible for cancer incidence; therefore, controlling inflammation is a potential strategy to stop the development of cancer. Innate Lymphoid Cells (ILC) represent a heterogeneous group of lymphocytes that exist in contrast to T cells. They function by interacting with cytokines and immune cells in an antigen-independent manner. In the digestive system cancer, from the inflammatory phase to the development, migration, and metastasis of tumors, ILC have been found to interact with the immune microenvironment and either control or promote these processes. The conventional treatments for digestive tumors have limited efficacy, therefore, ILC-associated immunotherapies are promising strategies. This study reviews the characterization of different ILC subpopulations, how they interact with and influence the immune microenvironment as well as chronic inflammation, and their promotional or inhibitory role in four common digestive system tumors, including pancreatic, colorectal, gastric, and hepatocellular cancers. In particular, the review emphasizes the role of ILC in associating chronic inflammation with cancer and the potential for enhanced immunotherapy with cytokine therapy and adoptive immune cell therapy.
Collapse
Affiliation(s)
- Guanliang Shen
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xinda Han
- Xinglin College, Nantong University, Nantong, Jiangsu, China
| | - Zehao Wei
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Songyun Zhao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xiumei Wang
- Affiliated Cancer Hospital of Inner Mongolia Medical University, 010020, Inner Mongolia, China
| | | | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
23
|
Zhang JN, Yang BB, Li LW, Xu H, Wang B, Yi ZL, Zhou XR, Liu H. Multi-omics pan-cancer analysis reveals the prognostic values and immunological functions of PPA2, with a spotlight on breast cancer. Front Immunol 2024; 15:1435502. [PMID: 39176095 PMCID: PMC11338811 DOI: 10.3389/fimmu.2024.1435502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Background Recently, the role of inorganic pyrophosphatase 2 (PPA2) has been remaining merely superficial in many tumors. Hence, the aim was to analyze the potential functions of PPA2 in pan-cancer, focusing on its role in breast cancer. Methods A systematic pan-cancer analysis conducted primarily utilizing various open databases such as TCGA and GTEx. We explored the clinical value of PPA2 as well as various biological functions, including expression levels and subcellular localization, multi-dimensional immune-correlation analysis, co-expression networks, and gene heterogeneity. In addition, we not only verified the function of PPA2 through cell experiments but also analyzed PPA2 at the single-cell level and its drug sensitivity. Results PPA2 is abnormally expressed in various tumors, and it is mainly distributed in mitochondria. Furthermore, the indicators (OS, DSS, DFI, and PFI) of analysis hint that PPA2 exhibits significant prognostic value. At the same time, the genomic heterogeneity (including TMB, MSI, MATH, and NEO) of PPA2 in pan-cancer was analyzed. Across multiple tumors, the results showed a close correlation between PPA2 expression levels and different immune signatures (such as immune cell infiltration). All of these indicate that PPA2 could potentially be applied in the guidance of immunotherapy. We also have demonstrated that PPA2 promoted the process of breast cancer. Finally, some potential therapeutic agents (such as Fulvestrant) targeting the abnormal expression of PPA2 are revealed. Conclusion In conclusion, the results demonstrated the great value of PPA2 in pan-cancer research, as well as its potential as a therapeutic target for breast tumors.
Collapse
Affiliation(s)
- Jia-Ning Zhang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Bei-Bei Yang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Lin-Wei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Hao Xu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Bin Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Zi-Lu Yi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xi-Rui Zhou
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- The Second Surgical Department of Breast Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
24
|
Li MP, Long SP, Liu WC, Long K, Gao XH. EMT-related gene classifications predict the prognosis, immune infiltration, and therapeutic response of osteosarcoma. Front Pharmacol 2024; 15:1419040. [PMID: 39170698 PMCID: PMC11335561 DOI: 10.3389/fphar.2024.1419040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS), a bone tumor with high ability of invasion and metastasis, has seriously affected the health of children and adolescents. Many studies have suggested a connection between OS and the epithelial-mesenchymal transition (EMT). We aimed to integrate EMT-Related genes (EMT-RGs) to predict the prognosis, immune infiltration, and therapeutic response of patients with OS. METHODS We used consensus clustering to identify potential EMT-Related OS molecular subtypes. Somatic mutation, tumor immune microenvironment, and functional enrichment analyses were performed for each subtype. We next constructed an EMT-Related risk signature and evaluated it by Kaplan-Meier (K-M) analysis survival and receiver operating characteristic (ROC) curves. Moreover, we constructed a nomogram to more accurately predict OS patients' clinical outcomes. Response effects of immunotherapy in OS patients was analyzed by Tumor Immune Dysfunction and Exclusion (TIDE) analysis, while sensitivity for chemotherapeutic agents was analyzed using oncoPredict. Finally, the expression patterns of hub genes were investigated by single-cell RNA sequencing (scRNA-seq) data analysis. RESULTS A total of 53 EMT-RDGs related to prognosis were identified, separating OS samples into two separate subgroups. The EMT-high subgroup showed favourable overall survival and more active immune response. Significant correlations were found between EMT-Related DEGs and functions as well as pathways linked to the development of OS. Additionally, a risk signature was established and OS patients were divided into two categories based on the risk scores. The signature presented a good predictive performance and could be recognized as an independent predictive factor for OS. Furthermore, patients with higher risk scores exhibited better sensitivity for five drugs, while no significant difference existed in immunotherapy response between the two risk subgroups. scRNA-seq data analysis displayed different expression patterns of the hub genes. CONCLUSION We developed a novel EMT-Related risk signature that can be considered as an independent predictor for OS, which may help improve clinical outcome prediction and guide personalized treatments for patients with OS.
Collapse
Affiliation(s)
- Meng-Pan Li
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Si-Ping Long
- The Fourth Clinical Medical College of Nanchang University, Nanchang, China
| | - Wen-Cai Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Long
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Xing-Hua Gao
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
25
|
Wang Z, Tang R, Wang H, Li X, Liu Z, Li W, Peng G, Zhou H. Bioinformatics analysis of the role of lysosome-related genes in breast cancer. Comput Methods Biomech Biomed Engin 2024:1-20. [PMID: 39054687 DOI: 10.1080/10255842.2024.2379936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
This study aimed to investigate the roles of lysosome-related genes in BC prognosis and immunity. Transcriptome data from TCGA and MSigDB, along with lysosome-related gene sets, underwent NMF cluster analysis, resulting in two subtypes. Using lasso regression and univariate/multivariate Cox regression analysis, an 11-gene signature was successfully identified and verified. High- and low-risk populations were dominated by HR+ sample types. There were differences in pathway enrichment, immune cell infiltration, and immune scores. Sensitive drugs targeting model genes were screened using GDSC and CCLE. This study constructed a reliable prognostic model with lysosome-related genes, providing valuable insights for BC clinical immunotherapy.
Collapse
Affiliation(s)
- Zhongming Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Ruiyao Tang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huazhong Wang
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Xizhang Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Zhenbang Liu
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Wenjie Li
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Gui Peng
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| | - Huaiying Zhou
- Department of Breast Oncology, The Third People's Hospital of Yongzhou, Yongzhou City, Hunan Province, China
| |
Collapse
|
26
|
Li Y, Yang X, Geng C, Liu Y, Tang T, Zhang L, Liu F, Zhang M, Hao J, Ma L. Identification of molecular subtypes based on chromatin regulator-related genes and experimental verification of the role of ASCL1 in conferring chemotherapy resistance to breast cancer. Front Immunol 2024; 15:1390261. [PMID: 38726001 PMCID: PMC11079216 DOI: 10.3389/fimmu.2024.1390261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
Objective The aim of this study was to identify the molecular subtypes of breast cancer based on chromatin regulator-related genes. Methods The RNA sequencing data of The Cancer Genome Atlas-Breast Cancer cohort were obtained from the official website, while the single-cell data were downloaded from the Gene Expression Omnibus database (GSE176078). Validation was performed using the Molecular Taxonomy of Breast Cancer International Consortium dataset. Furthermore, the immune characteristics, tumor stemness, heterogeneity, and clinical characteristics of these molecular subtypes were analyzed. The correlation between chromatin regulators and chemotherapy resistance was examined in vitro using the quantitative real-time polymerase chain reaction (qRT-PCR) and Cell Counting Kit-8 (CCK8) assays. Results This study identified three stable molecular subtypes with different prognostic and pathological features. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction analyses revealed that the differentially expressed genes were associated with disease processes, such as mitotic nuclear division, chromosome segregation, condensed chromosome, and specific chromosome region. The T stage and subtypes were correlated with the clinical features. Tumor heterogeneity (mutant-allele tumor heterogeneity, tumor mutational burden, purity, and homologous recombination deficiency) and tumor stemness (RNA expression-based stemness score, epigenetically regulated RNA expression-based stemness score, DNA methylation-based stemness score, and epigenetically regulated DNA methylation-based stemness score) significantly varied between the three subtypes. Furthermore, Western blotting, qRT-PCR, and CCK8 assays demonstrated that the expression of ASCL1 was positively correlated with chemotherapy resistance in breast cancer. Conclusion This study identified the subtypes of breast cancer based on chromatin regulators and analyzed their clinical features, gene mutation status, immunophenotype, and drug sensitivity. The results of this study provide effective strategies for assessing clinical prognosis and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Yilun Li
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xiaolu Yang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Cuizhi Geng
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yunjiang Liu
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tiantian Tang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lina Zhang
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fei Liu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meng Zhang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Li Ma
- Department of Breast Disease Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|