1
|
Chen P, Li J, Tian R. Construction of a novel prognostic model based on lncRNAs-related to DNA damage repair for predicting the prognosis of clear cell renal cell carcinoma. Ann Med 2025; 57:2480755. [PMID: 40172678 PMCID: PMC11966993 DOI: 10.1080/07853890.2025.2480755] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND CcRCC has the characteristics of high aggression, high metastasis, high mortality, wide tumour heterogeneity and variable clinical course. The purpose of this study was to explore the potential value of lncRNAs-related to DNA damage repair (DDR) in predicting the prognosis of ccRCC by construction and verification a novel prognostic model. METHODS RNA-seq data and clinical data of ccRCC were downloaded from public databases. Subsequently, Pearson correlation analysis and differential expression analysis were performed to identify DElncRNAs-related to DDR. Then, through univariate Cox analysis and LASSO analysis, the DElncRNAs-related to DDR associated with prognosis were screened for the construction of novel risk score prognostic model. In addition, functional annotation, tumour mutation burden, immune correlation and drug sensitivity analyses were performed based on risk score to assess the characteristics of patients in different risk score groups. RESULTS Based on univariate Cox analysis and LASSO analysis, four best DElncRNAs-related to DDR were selected. Subsequently, a novel risk score prognostic model based on these four DElncRNAs was constructed through LASSO. Multivariate Cox analysis showed that risk score and age were independent prognostic factors for ccRCC (p < 0.05). Functional enrichment analysis showed that DDR-related biological processes were mainly enriched in the high risk group. The highly mutated genes in the high and low risk groups were the same (VHL, PBRM1 and TTN), but they also had their own unique mutated genes. Pearson correlation analysis showed that the risk score was significantly (p < 0.05) positively correlated with the infiltration degree of CD8 T cells evaluated by six algorithms. In addition, it was found that the high and low risk groups had different sensitivities to the drugs Etoposide, Imatinib, Sorafenib, Bosutinib and Sunitinib. CONCLUSION A novel prognostic model was constructed based on four DElncRNAs-related to DDR. The model has satisfactory accuracy in predicting survival of ccRCC patients.
Collapse
Affiliation(s)
- Peng Chen
- Department of Urology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jian Li
- Department of Urology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Renli Tian
- Department of Urology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Li Z, Zhong Y, Ye D, Yang J, Chen L. Revealing NAPSA's role in ccRCC: Insights from single-cell RNA sequencing. Gene 2025; 959:149478. [PMID: 40194687 DOI: 10.1016/j.gene.2025.149478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is aggressive and heterogeneous, resulting in poor prognosis due to frequent metastasis. Napsin A, an aspartic proteinase encoded by the NAPSA gene, is involved in protein processing and is expressed in the kidney and lung, but its function is not well understood. Studying ccRCC's molecular characteristics, including Napsin A, is vital for enhancing diagnostics and treatment. METHODS Single-cell RNA sequencing data from the GEO database (GSE210042) were analyzed, including seven tumor and two normal samples. The Seurat package was used for data preprocessing, clustering, and visualization. Differential expression and enrichment analyses were conducted between tumor and normal cells, and cell-to-cell communication was assessed between NAPSA + and NAPSA- cells. The correlation between NAPSA expression and EMT score was analyzed using TCGA-KIRC data. In vitro experiments involved transfecting OS-RC-2 and Caki-1 ccRCC cell lines with siRNA targeting NAPSA. Effect on the cellular EMT process induced by TGF-β1 was assessed by immunofluorescence staining. RESULTS NAPSA was primarily expressed in podocytes and ccRCC epithelial cells, with significantly reduced levels in tumor tissues associated with poor prognosis. NAPSA downregulation may influence various biological pathways and enhance communication with tumor-associated macrophages and mast cells. Silencing NAPSA increased TGF-β1-induced epithelial-mesenchymal transition (EMT). CONCLUSION The study highlights NAPSA's expression characteristics and potential role in ccRCC, suggesting it may serve as a biomarker. Further research is needed to elucidate NAPSA's mechanisms and explore its applications in precision medicine.
Collapse
Affiliation(s)
- Zhichao Li
- Doctoral Candidate in School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750001, Ningxia, China; Shanghai Jiahui International Hospital, Shanghai, China
| | - Yuanjie Zhong
- Graduate Student in Ningxia Medical University, Yinchuan 750001, Ningxia, China
| | - Dan Ye
- Ningxia Medical University, Yinchuan 750001 Ningxia, China
| | - Jincheng Yang
- Department of Urology, Yinchuan First People's Hospital, Deputy Chief Physician, Yinchuan 750001, Ningxia, China
| | - Linbao Chen
- Yinchuan Women and Children Healthcare Hospital, Yinchuan 750001, Ningxia, China; Yinchuan First People's Hospital, Yinchuan 750001, Ningxia, China.
| |
Collapse
|
3
|
Qiu H, Liu F, Qiu M, Yang J, Peng X. Monotropein attenuates renal cell carcinoma cell progression and M2 macrophage polarization by weakening NF-κB. Int Urol Nephrol 2025; 57:1785-1795. [PMID: 39776402 DOI: 10.1007/s11255-024-04358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE The study aimed to investigate the effect and mechanism of monotropein on renal cell carcinoma (RCC). METHODS After monotropein and NF-κB receptor activator (RANKL) treatment, cell proliferation, invasion, and apoptosis were evaluated using CCK-8, Transwell, and flow cytometry. Primary macrophages co-cultured with monotropein-treated RCC cells were analyzed to evaluate macrophage polarization using qRT-PCR, western blot, and ELISA assays by detecting the expression of M2 markers (CD206, CD168) and cytokines (IL-10, TGF-β). Additionally, the therapeutic efficacy of monotropein was examined using an RCC mouse xenograft model. RESULTS Monotropein could inhibit the proliferation, invasion, and M2 macrophage polarization and accelerate the apoptosis of RCC cells. Mechanistically, monotropein suppressed NF-κB pathway activation in RCC cells and reduced the expression of NF-κB downstream targets, including Bcl-2, c-Myc, and MMP9. RANKL could eliminate the effect of monotropein on RCC progression. In primary macrophages co-cultured with monotropein-treated RCC cells, monotropein downregulated M2 polarization markers and cytokines, further supporting its role in modulating the tumor microenvironment. In mouse models, monotropein reduced RCC tumor growth, induced apoptosis, and blocked NF-κB pathway. CONCLUSIONS Monotropein prevents RCC malignant progression and reduces M2 macrophage polarization by suppressing the NF-κB pathway, suggesting that monotropein may serve as a potential therapeutic agent for RCC by targeting both tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Heping Qiu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Fei Liu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China
| | - Mei Qiu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, 341099, China
| | - Juliang Yang
- Department of Urology, The Affiliated Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou, 341099, China
| | - Xiang Peng
- Department of Urology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, East Lake District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
4
|
Yang A, Zhou M, Gao Y, Zhang Y. Mechanisms of CD8 + T cell exhaustion and its clinical significance in prognosis of anti-tumor therapies: A review. Int Immunopharmacol 2025; 159:114843. [PMID: 40394796 DOI: 10.1016/j.intimp.2025.114843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
In recent years, immunotherapy has gradually become one of the main strategies for cancer treatment, with immune checkpoint inhibitors (ICIs) offering new possibilities for tumor therapy. However, some cancer patients exhibit low responses and resistance to ICIs treatment. T cell exhaustion, a process associated with tumor progression, refers to a subset of T cells that progressively lose effector functions and exhibit increased expression of inhibitory receptors. These exhausted T cells are considered key players in the therapeutic efficacy of immune checkpoint inhibitors. Therefore, understanding the impact of T cell exhaustion on tumor immunotherapy and the underlying mechanisms is critical for improving clinical treatment outcomes. Several elegant studies have provided insights into the prognostic value of exhausted T cells in cancers. In this review, we highlight the process of exhausted T cells and its predictive value in various cancers, as well as the relevant mechanisms behind it, providing new insights into the immunotherapy of cancer.
Collapse
Affiliation(s)
- Anrui Yang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Meng Zhou
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yixuan Gao
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Gynecological Minimal Invasive Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Wang Z, Feng C, Chen G, Cai Q. Astragaloside IV regulates macrophage polarization via the TLR4/NF-κB/STAT3 pathway to inhibit the malignant phenotype of renal clear cell carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04219-3. [PMID: 40343452 DOI: 10.1007/s00210-025-04219-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/23/2025] [Indexed: 05/11/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and aggressive type of kidney cancer. This study aimed to investigate the effect of astragaloside IV (AS-IV) on ccRCC. A variety of cell experimental techniques were used, inducing macrophage polarization, and detecting relevant indicators by flow cytometry, qRT-PCR, immunofluorescence, MTT, wound healing, Transwell, and western blot. A nude mouse xenograft tumor model was constructed for in vivo studies, and siRNA interference technology was used to explore the signaling pathway. The results demonstrated successful macrophage polarization, with AS-IV inhibiting M2 macrophage polarization and promoting the transition from M0 to M1 polarization. Additionally, AS-IV suppressed ccRCC cell proliferation, migration, and invasion, while reversing the malignant effects of M2 macrophages. The study further revealed that AS-IV inhibited M2 polarization through the TLR4/NF-κB/STAT3 signaling pathway. In vivo experiments showed that AS-IV inhibited the growth of ccRCC tumors. This study revealed that AS-IV influences macrophage polarization by regulating the TLR4/NF-κB/STAT3 signaling pathway, thereby inhibiting the malignant phenotype of ccRCC. This finding provides new insights and potential therapeutic strategies for the treatment of ccRCC.
Collapse
Affiliation(s)
- Zhanshi Wang
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Chao Feng
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Guodong Chen
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China
| | - Qi Cai
- Department of Urology, Tongde Hospital of Zhejiang Province, No. 234 Gucui Road, Xihu District, Hangzhou City, Zhejiang Province, 330012, China.
| |
Collapse
|
6
|
Hugaboom MB, Wirth LV, Street K, Ruthen N, Jegede OA, Schindler NR, Shah V, Zaemes JP, Ahmar NE, Matar S, Savla V, Choueiri TK, Denize T, West DJ, McDermott DF, Plimack ER, Sosman JA, Haas NB, Stein MN, Alter R, Bilen MA, Hurwitz ME, Hammers H, Signoretti S, Atkins MB, Wu CJ, Braun DA. Presence of Tertiary Lymphoid Structures and Exhausted Tissue-Resident T Cells Determines Clinical Response to PD-1 Blockade in Renal Cell Carcinoma. Cancer Discov 2025; 15:948-968. [PMID: 39992403 PMCID: PMC12048281 DOI: 10.1158/2159-8290.cd-24-0991] [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: 07/12/2024] [Revised: 01/08/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
SIGNIFICANCE We describe a paradigm wherein combined high TLS and low tissue-resident exhausted CD8+ T cells are required for optimal response to PD-1 blockade in RCC. This analysis identifies key determinants of response to PD-1 blockade in advanced RCC and suggests avenues for future immune modulation through rational combination therapy strategies.
Collapse
Affiliation(s)
- Miya B. Hugaboom
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Lena V. Wirth
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Street
- Division of Biostatistics, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Neil Ruthen
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Opeyemi A. Jegede
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Valisha Shah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob P. Zaemes
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sayed Matar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Varunika Savla
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Destiny J. West
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Jeffrey A. Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Naomi B. Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark N. Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Robert Alter
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mehmet A. Bilen
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael E. Hurwitz
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Hans Hammers
- Department of Internal Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael B. Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David A. Braun
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
Li Y, Li C. ZACN Associated with Poor Prognosis Promotes Proliferation of Kidney Renal Clear Cell Carcinoma Cells by Inhibiting JTC801-Induced Alkaliptosis. Appl Biochem Biotechnol 2025; 197:3346-3362. [PMID: 39937414 DOI: 10.1007/s12010-025-05197-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Alkaliptosis, crucial in various cancers, is a specific form of cell death. This study aims to screen a prognosis-related gene in kidney renal clear cell carcinoma (KIRC) using the alkaliptosis gene set and to investigate the roles of the gene in KIRC and its association with alkaliptosis. Transcriptome and clinical information of KIRC patients were collected from the TCGA-KIRC and GSE29609 database. We detected ZACN levels in normal kidney cells and KIRC cells and assessed the ZACN and JTC801-induced alkaliptosis relationship using immunoblotting and pH measurement. In the alkaliptosis gene set (IKBKB, NFKB1, CA9, CHUK, IKBKG, and RELA), NFKB1, CHUK, and IKBKG exhibited differential expression in TCGA-KIRC. Based on these three genes, two alkaliptosis patterns were identified in TCGA-KIRC. The independent prognostic gene for KIRC, ZACN, was screened. High ZACN in KIRC patients indicated a poor prognosis. ZACN was inversely related to the infiltration of anti-cancer immune cells such as CD4 + T cells, macrophages, and neutrophils, and it regulated the immune checkpoint and gene mutations. Patients characterized by high ZACN levels exhibited a heightened drug sensitivity to ABT737_1910, 5-Fluorouracil_1073, etc. ZACN expression in KIRC cells was increased relative to normal kidney cells and was inhibited in a concentration-dependent manner by JTC801. ZACN overexpression suppressed p-p65/p65 expression, increased expression of CA9, and lowered intracellular pH. In KIRC, ZACN inhibits JTC801-induced alkaliptosis. This study sheds light on a novel mechanism involving ZACN and alkaliptosis in KIRC, offering a promising avenue for further research and potential therapeutic interventions in KIRC management.
Collapse
Affiliation(s)
- Yifan Li
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Can Li
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital of Central South University, No. 139 Renmin Middle Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Elias R, Nirschl T, Rezaee M, Yerrapragada A, Wang S, Cheaib J, Alam R, Patel S, Jing Y, Allaf M, McKean D, Klein AP, Fertig EJ, Baraban E, Ged Y, Yegnasubramanian S, Singla N. Clear-Cell Renal Cell Carcinoma Molecular Subtypes Differ by African and European Genetic Similarity. CANCER RESEARCH COMMUNICATIONS 2025; 5:743-755. [PMID: 40131247 PMCID: PMC12044083 DOI: 10.1158/2767-9764.crc-24-0624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
SIGNIFICANCE Our study shows that AFR genetic similarity correlates with distinct ccRCC molecular subtypes. Further research is needed to disentangle environmental and genetic influences. Identifying these differences underscores the critical importance of including racially and ethnically diverse populations in cancer research to ensure more equitable and sustainable outcomes worldwide for all patients.
Collapse
Affiliation(s)
- Roy Elias
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas Nirschl
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Rezaee
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anirudh Yerrapragada
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shirley Wang
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph Cheaib
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ridwan Alam
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sunil Patel
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuezhou Jing
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mohamad Allaf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David McKean
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alison P. Klein
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elana J. Fertig
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
| | - Ezra Baraban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- inHealth Precision Medicine program, Johns Hopkins Medicine, Baltimore, Maryland
| | - Nirmish Singla
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Jing W, Han M, Wang G, Kong Z, Zhao X, Fu Z, Jiang X, Shi C, Chen C, Zhang J, Zheng Z, Gao J, Sun W, Tang C, Yang Z, Wang Y, Liu Y, Zhao K, Zhu D, Shi B, Jiang X. An in situ engineered chimeric IL-2 receptor potentiates the tumoricidal activity of proinflammatory CAR macrophages in renal cell carcinoma. NATURE CANCER 2025:10.1038/s43018-025-00950-1. [PMID: 40301655 DOI: 10.1038/s43018-025-00950-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 03/11/2025] [Indexed: 05/01/2025]
Abstract
Chimeric antigen receptor macrophage (CAR-M) therapy has shown great promise in solid malignancies; however, the phenotypic re-domestication of CAR-Ms in the immunosuppressive tumor niche restricts their antitumor immunity. We here report an in situ engineered chimeric interleukin (IL)-2 signaling receptor (CSR) for controllably manipulating the proinflammatory phenotype of CAR-Ms, augmenting their sustained tumoricidal immunity. Specifically, our in-house-customized lipid nanoparticles efficiently introduce dual circular RNAs into macrophages to generate CSR-functionalized CAR-Ms. The intracellular inflammatory signaling pathway of CAR-Ms can be stimulated with the IL-2 therapeutic via the synthetic IL-2 receptor, which induces the antitumor phenotype shifting of CAR-Ms. Moreover, hydrogel-mediated combinatory treatment with lipid nanoparticles and IL-2 remodels the immunosuppressive tumor microenvironment and promotes tumor regression in renal carcinoma animal models. In summary, our findings establish that the proinflammatory phenotype of CAR-Ms can be modulated by a synthetic IL-2 receptor, benefiting the antitumor immunotherapy of CAR-Ms with broad application in other solid malignancies.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Maosen Han
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ganyu Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhichao Kong
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaotian Zhao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhipeng Fu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuewen Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chongdeng Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Zhang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zuolin Zheng
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jinxin Gao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weiyi Sun
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunwei Tang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhenmei Yang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kun Zhao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Danqing Zhu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Xinyi Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine; Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
10
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
11
|
Jiang Q, Braun DA, Clauser KR, Ramesh V, Shirole NH, Duke-Cohan JE, Nabilsi N, Kramer NJ, Forman C, Lippincott IE, Klaeger S, Phulphagar KM, Chea V, Kim N, Vanasse AP, Saad E, Parsons T, Carr-Reynolds M, Carulli I, Pinjusic K, Jiang Y, Li R, Syamala S, Rachimi S, Verzani EK, Stevens JD, Lane WJ, Camp SY, Meli K, Pappalardi MB, Herbert ZT, Qiu X, Cejas P, Long HW, Shukla SA, Van Allen EM, Choueiri TK, Churchman LS, Abelin JG, Gurer C, MacBeath G, Childs RW, Carr SA, Keskin DB, Wu CJ, Kaelin WG. HIF regulates multiple translated endogenous retroviruses: Implications for cancer immunotherapy. Cell 2025; 188:1807-1827.e34. [PMID: 40023154 PMCID: PMC11988688 DOI: 10.1016/j.cell.2025.01.046] [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: 01/18/2024] [Revised: 11/14/2024] [Accepted: 01/31/2025] [Indexed: 03/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC), despite having a low mutational burden, is considered immunogenic because it occasionally undergoes spontaneous regressions and often responds to immunotherapies. The signature lesion in ccRCC is inactivation of the VHL tumor suppressor gene and consequent upregulation of the HIF transcription factor. An earlier case report described a ccRCC patient who was cured by an allogeneic stem cell transplant and later found to have donor-derived T cells that recognized a ccRCC-specific peptide encoded by a HIF-responsive endogenous retrovirus (ERV), ERVE-4. We report that ERVE-4 is one of many ERVs that are induced by HIF, translated into HLA-bound peptides in ccRCCs, and capable of generating antigen-specific T cell responses. Moreover, ERV expression can be induced in non-ccRCC tumors with clinical-grade HIF stabilizers. These findings have implications for leveraging ERVs for cancer immunotherapy.
Collapse
Affiliation(s)
- Qinqin Jiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - David A Braun
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Vijyendra Ramesh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nitin H Shirole
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph E Duke-Cohan
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Nicholas J Kramer
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Isabelle E Lippincott
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Kshiti M Phulphagar
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Vipheaviny Chea
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nawoo Kim
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Allison P Vanasse
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | | | | | - Isabel Carulli
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Katarina Pinjusic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yijia Jiang
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Rong Li
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sudeepa Syamala
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Suzanna Rachimi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Eva K Verzani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Jonathan D Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - William J Lane
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Sabrina Y Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | | | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Paloma Cejas
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachet A Shukla
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - L Stirling Churchman
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jennifer G Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | | | | | - Richard W Childs
- Laboratory of Transplantation Immunotherapy, Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Computer Science, Metropolitan College, Boston University, Boston, MA 02215, USA; Section for Bioinformatics, Department of Health Technology, Technical University of Denmark 2800 Lyngby, Denmark.
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA.
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02215, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
12
|
McKinnon MB, Rini BI, Haake SM. Biomarker-informed care for patients with renal cell carcinoma. NATURE CANCER 2025; 6:573-583. [PMID: 40240621 DOI: 10.1038/s43018-025-00942-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
Kidney cancer is a commonly diagnosed cancer in adults, and clear cell renal cell carcinoma (ccRCC) is the most common histological subtype. Immune checkpoint inhibitors have revolutionized care for patients with ccRCC, either as adjuvant therapy or combined with other agents in advanced disease. However, biomarkers to predict therapeutic benefits are lacking. Here, we explore biomarkers that predict therapeutic response in other tumor types and discuss the reasons for their ineffectiveness in ccRCC. We also review emerging predictive and prognostic biomarkers to prioritize in ccRCC, including gene expression signatures.
Collapse
Affiliation(s)
- Mackenzie B McKinnon
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brian I Rini
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Scott M Haake
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Department of Veterans Affairs, Nashville, TN, USA.
| |
Collapse
|
13
|
He S, Sun S, Liu K, Pang B, Xiao Y. Comprehensive assessment of computational methods for cancer immunoediting. CELL REPORTS METHODS 2025; 5:101006. [PMID: 40132544 PMCID: PMC12049729 DOI: 10.1016/j.crmeth.2025.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/23/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Cancer immunoediting reflects the role of the immune system in eliminating tumor cells and shaping tumor immunogenicity, which leaves marks in the genome. In this study, we systematically evaluate four methods for quantifying immunoediting. In colorectal cancer samples from The Cancer Genome Atlas, we found that these methods identified 78.41%, 46.17%, 36.61%, and 4.92% of immunoedited samples, respectively, covering 92.90% of all colorectal cancer samples. Comparison of 10 patient-derived xenografts (PDXs) with their original tumors showed that different methods identified reduced immune selection in PDXs ranging from 44.44% to 60.0%. The proportion of such PDX-tumor pairs increases to 77.78% when considering the union of results from multiple methods, indicating the complementarity of these methods. We find that observed-to-expected ratios highly rely on neoantigen selection criteria and reference datasets. In contrast, HLA-binding mutation ratio, immune dN/dS, and enrichment score of cancer cell fraction were less affected by these factors. Our findings suggest integration of multiple methods may benefit future immunoediting analyses.
Collapse
Affiliation(s)
- Shengyuan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Shangqin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
14
|
Zhang J, Liu G, Wang W. PRSS53 is a potential novel biomarker related to prognosis and immunity in clear cell renal cell carcinoma. Discov Oncol 2025; 16:362. [PMID: 40111561 PMCID: PMC11925835 DOI: 10.1007/s12672-025-02114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE To analyze the expression levels, clinical significance, Immune infiltration and prognostic value of PRSS53 (Protease Serine 53) in clear cell renal cell carcinoma (ccRCC) using bioinformatics methods. METHODS Data on PRSS53 in ccRCC were extracted from databases and platforms, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx), The Gene Expression Omnibus (GEO), Xiantao Academic Tool, Human Protein Atlas (HPA) and so on. We analyzed the relationship between PRSS53 expression and the clinical and pathological characteristics, diagnosis, immune infiltration and prognosis in ccRCC patients. Additionally, immunohistochemical analysis was performed on 9 pairs of ccRCC patient samples. RESULTS PRSS53 was significantly upregulated in ccRCC and was closely associated with the TNM stage and histological grade of ccRCC. Receiver operating characteristic (ROC) curve analysis demonstrated the excellent diagnostic performance of PRSS53 in ccRCC (AUC = 0.928). Patients with high PRSS53 expression exhibited lower overall survival (OS) and disease-specific survival (DSS). Gene set enrichment analysis (GSEA) revealed that PRSS53 is involved in cellular functions such as anchored component of membrane, basement membrane and RNA-binding involved in post-transcriptional gene silencing. Single-sample GSEA (ssGSEA) indicated a positive correlation between PRSS53 expression and T helper cells infiltration levels, and a negative correlation with T gamma delta (Tgd) cell infiltration. PRSS53 was predominantly expressed in renal proximal tubules. The immunohistochemical results and HPA database showed that PRSS53 protein expression was significantly lower in clinical ccRCC tissues compared to normal tissues. CONCLUSION PRSS53 is a new prognostic biomarker and potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Jiajun Zhang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Guocheng Liu
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Wei Wang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China.
- Department of Urology, Yancheng No.1 People's Hospital, Yancheng, 224006, China.
| |
Collapse
|
15
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
16
|
Miyagi H, Yu X, Peak T, Dhillon J, Le C, Wang X, Yoder S, Marchion D, Lu X, Pettaway C, Moran Segura C, Zhang C, Roman Souza G, Yu A, Zemp L, Spiess P, Chahoud J. Progressive T cell exhaustion and predominance of aging tissue associated macrophages with advancing disease stage in penile squamous cell carcinoma. Sci Rep 2025; 15:7703. [PMID: 40044748 PMCID: PMC11882776 DOI: 10.1038/s41598-025-89760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Penile squamous cell carcinoma (PSCC) is a rare malignancy with limited understanding of the tumor immune microenvironment (TIME). The interplay between PSCC and the immune system across disease progression and HPV infection status remains poorly characterized. This study aims to assess the TIME changes from localized to advanced disease and between HPV-positive versus negative tumors to identify potential immune evasion mechanisms in advanced PSCC. scRNA-seq was performed on ten PSCC tissue samples from penile, lymph node and distant metastatic sites with four matched penile and lymph node samples to understand the cellular heterogeneity within PSCC tumors. Analysis of immune cell populations and transcriptional hallmarks were performed stratified by localized (pT1-3, N0) versus advanced (N1-3, M0 or any N, M1) disease states and HPV infection status. We observed significant differences in immune cell infiltration between localized and advanced PSCC disease states and by HPV status. Advanced disease states demonstrated an exhausted immune phenotype, characterized by terminally exhausted CD8+ T cells, M2-like macrophages and hypoxic signature, while localized disease states demonstrated an active innate immune system characterized by increased DCs. HPV-negative tumors displayed low immune cell infiltration while HPV-positive tumors demonstrated an immune exhausted phenotype. These findings offer valuable insights into the evolving PSCC immune landscape, paving the way for the development of potential therapeutic approaches for advanced PSCC.
Collapse
Affiliation(s)
- Hiroko Miyagi
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA.
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Taylor Peak
- Department of Urology, University of Tennessee College of Medicine, Chatanooga, USA
| | - Jasreman Dhillon
- Pathology Department, H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Casey Le
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Sean Yoder
- Molecular Genomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Doug Marchion
- Tissue Core Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xin Lu
- Department of Biological Sciences, University of Notre Dame, Norte Dame, IN, 46556, USA
| | - Curtis Pettaway
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Chaomei Zhang
- Molecular Genomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Gabriel Roman Souza
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA
| | - Alice Yu
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA
| | - Logan Zemp
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA
| | - Philippe Spiess
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA
| | - Jad Chahoud
- Genitourinary Oncology Department, H. Lee Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA.
| |
Collapse
|
17
|
Braun DA, Moranzoni G, Chea V, McGregor BA, Blass E, Tu CR, Vanasse AP, Forman C, Forman J, Afeyan AB, Schindler NR, Liu Y, Li S, Southard J, Chang SL, Hirsch MS, LeBoeuf NR, Olive O, Mehndiratta A, Greenslade H, Shetty K, Klaeger S, Sarkizova S, Pedersen CB, Mossanen M, Carulli I, Tarren A, Duke-Cohan J, Howard AA, Iorgulescu JB, Shim B, Simon JM, Signoretti S, Aster JC, Elagina L, Carr SA, Leshchiner I, Getz G, Gabriel S, Hacohen N, Olsen LR, Oliveira G, Neuberg DS, Livak KJ, Shukla SA, Fritsch EF, Wu CJ, Keskin DB, Ott PA, Choueiri TK. A neoantigen vaccine generates antitumour immunity in renal cell carcinoma. Nature 2025; 639:474-482. [PMID: 39910301 PMCID: PMC11903305 DOI: 10.1038/s41586-024-08507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Personalized cancer vaccines (PCVs) can generate circulating immune responses against predicted neoantigens1-6. However, whether such responses can target cancer driver mutations, lead to immune recognition of a patient's tumour and result in clinical activity are largely unknown. These questions are of particular interest for patients who have tumours with a low mutational burden. Here we conducted a phase I trial (ClinicalTrials.gov identifier NCT02950766) to test a neoantigen-targeting PCV in patients with high-risk, fully resected clear cell renal cell carcinoma (RCC; stage III or IV) with or without ipilimumab administered adjacent to the vaccine. At a median follow-up of 40.2 months after surgery, none of the 9 participants enrolled in the study had a recurrence of RCC. No dose-limiting toxicities were observed. All patients generated T cell immune responses against the PCV antigens, including to RCC driver mutations in VHL, PBRM1, BAP1, KDM5C and PIK3CA. Following vaccination, there was a durable expansion of peripheral T cell clones. Moreover, T cell reactivity against autologous tumours was detected in seven out of nine patients. Our results demonstrate that neoantigen-targeting PCVs in high-risk RCC are highly immunogenic, capable of targeting key driver mutations and can induce antitumour immunity. These observations, in conjunction with the absence of recurrence in all nine vaccinated patients, highlights the promise of PCVs as effective adjuvant therapy in RCC.
Collapse
Affiliation(s)
- David A Braun
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Giorgia Moranzoni
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Vipheaviny Chea
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bradley A McGregor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Eryn Blass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chloe R Tu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison P Vanasse
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juliet Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander B Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nicholas R Schindler
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Yiwen Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuqiang Li
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jackson Southard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven L Chang
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle S Hirsch
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicole R LeBoeuf
- Harvard Medical School, Boston, MA, USA
- Center for Cutaneous Oncology, Dana-Farber Brigham and Women's Cancer Center, Boston, MA, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Oriol Olive
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ambica Mehndiratta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Haley Greenslade
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keerthi Shetty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan Klaeger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Christina B Pedersen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Center for Genomic Medicine, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Matthew Mossanen
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Isabel Carulli
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna Tarren
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joseph Duke-Cohan
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexis A Howard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - J Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bohoon Shim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jon C Aster
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ignaty Leshchiner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, USA
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Lars R Olsen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sachet A Shukla
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Tan H, Cai M, Wang J, Yu T, Xia H, Zhao H, Zhang X. Harnessing Macrophages in Cancer Therapy: from Immune Modulators to Therapeutic Targets. Int J Biol Sci 2025; 21:2235-2257. [PMID: 40083710 PMCID: PMC11900799 DOI: 10.7150/ijbs.106275] [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: 11/03/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
Macrophages, as the predominant phagocytes, play an essential role in pathogens defense and tissue homeostasis maintenance. In the context of cancer, tumor-associated macrophages (TAMs) have evolved into cunning actors involved in angiogenesis, cancer cell proliferation and metastasis, as well as the construction of immunosuppressive microenvironment. Once properly activated, macrophages can kill tumor cells directly through phagocytosis or attack tumor cells indirectly by stimulating innate and adaptive immunity. Thus, the prospect of targeting TAMs has sparked significant interest and emerged as a promising strategy in immunotherapy. In this review, we summarize the diverse roles and underlying mechanisms of TAMs in cancer development and immunity and highlight the TAM-based therapeutic strategies such as inhibiting macrophage recruitment, inhibiting the differentiation reprogramming of TAMs, blocking phagocytotic checkpoints, inducing trained macrophages, as well as the potential of engineered CAR-armed macrophages in cancer therapy.
Collapse
Affiliation(s)
- Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province, China
- General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Meihe Cai
- Department of Traditional Chinese Medicine, Zhushan Renmin Hospital, Zhushan, 442200, China
| | | | - Tao Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Houjun Xia
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huanbin Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Present: Division of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaoyu Zhang
- Department of Gastrointestinal Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
19
|
Chen Y, Jia H, Zhang X, Zhao H, Xiao Y, Li N, Yao Y, Xing X. Disruption of GPSM1/CSF1 signaling reprograms tumor-associated macrophages to overcome anti-PD-1 resistance in colorectal cancer. J Immunother Cancer 2025; 13:e010826. [PMID: 40010765 PMCID: PMC12083360 DOI: 10.1136/jitc-2024-010826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) therapies, particularly anti-PD-1, benefit only a limited subset of colorectal cancer (CRC) patients. G-protein signaling modulator 1 (GPSM1) is implicated in immunity and oncology, yet its role in regulating the CRC tumor microenvironment (TME) and contributing to anti-PD-1 resistance remains poorly understood. METHODS We employed single-cell RNA sequencing and multiplex immunofluorescence on tumor samples from anti-PD-1-resistant CRC patients to evaluate GPSM1 expression and its impact on macrophage polarization. An orthotopic CRC xenograft model in C57BL/6 mice was used to assess the role of GPSM1 in vivo. An in vitro co-culture system, alongside mass cytometry and flow cytometry, explored GPSM1's biological functions within the TME. We further used ChIP-PCR, mass spectrometry, and co-immunoprecipitation to elucidate the mechanisms regulating GPSM1 activity. RESULTS GPSM1 expression was significantly elevated in anti-PD-1-resistant CRC tissues. Enhanced GPSM1 levels promoted anti-PD-1 resistance by driving macrophage polarization toward an immunosuppressive M2 phenotype, facilitating their infiltration into the TME. We identified the deubiquitinase USP9X as a key factor preventing GPSM1 degradation through K63-polyubiquitination. This stabilization of GPSM1 led to MEIS3 nuclear translocation, activating macrophage colony-stimulating factor expression. Importantly, ruxolitinib emerged as a promising GPSM1-targeting candidate, demonstrating improved efficacy in combination with anti-PD-1 therapy in both microsatellite instability-high and microsatellite stable CRC models. CONCLUSIONS Our findings highlight the pivotal role of GPSM1-driven M2 macrophage infiltration in mediating anti-PD-1 resistance in CRC. Targeting GPSM1 offers a novel therapeutic strategy to enhance ICB efficacy, potentially broadening the patient population that may benefit from these therapies.
Collapse
Affiliation(s)
- Yang Chen
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Huiqing Jia
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangyan Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yujing Xiao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Na Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yifan Yao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
20
|
Pourmir I, Benhamouda N, Tran T, Roux H, Pineau J, Gey A, Munoz A, Mabrouk N, Epaillard N, Verkarre V, Vano YA, Tartour E, Oudard S. Soluble TIM-3, likely produced by myeloid cells, predicts resistance to immune checkpoint inhibitors in metastatic clear cell renal cell carcinoma. J Exp Clin Cancer Res 2025; 44:54. [PMID: 39953623 PMCID: PMC11827183 DOI: 10.1186/s13046-025-03293-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Immunotherapies targeting PD-1 and CTLA-4 are key components of the treatment of metastatic clear cell renal cell carcinoma (mccRCC). However, they have distinct safety profiles and resistance to treatment can occur. We assess soluble TIM-3 (sTIM-3) in the plasma of mccRCC patients as a potential theranostic biomarker, as well as its source and biological significance. METHODS We analyzed the association between sTIM-3 and overall survival (OS), tumor response, and common clinical and biological factors in two mccRCC cohorts treated with anti-PD-1 (nivolumab, n = 27), anti-PD-1 or anti-PD-1 + anti-CTLA-4 (nivolumab + ipilimumab - N + I, n = 124). The origin and role of sTIM-3 are studied on tumor and blood samples, using multiplex immunohistochemistry and flow cytometry, as well as analyses of publicly available single-cell transcriptomic (scRNAseq) and mass cytometry data. RESULTS sTIM-3 is significantly elevated in the plasma of treatment-naive mccRCC. It shows distinct associations with survival on anti-PD-1 vs anti-PD-1 + anti-CTLA-4: under nivolumab monotherapy, sTIM-3-high patients have a significantly reduced survival compared to sTIM-3-low patients, while they have similar survival probabilities under N + I. sTIM-3 is independent from other clinical and biological factors. Myeloid immune cells appear as the prominent source of sTIM-3, which may indicate their dysfunctional role in the antitumor immune response. CONCLUSIONS sTIM-3 appears to be a promising biomarker for optimizing treatment strategies in ccRCC as well as a potential therapeutic target, linked with to the immune myeloid compartment. Future investigations are warranted in patients treated with anti-PD-1 + antiangiogenic therapies.
Collapse
Affiliation(s)
- Ivan Pourmir
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Medical Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Thoracic Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| | - Nadine Benhamouda
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Thi Tran
- Université Paris Cité, INSERM U970, PARCC, Paris, France
| | - Hugo Roux
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Joséphine Pineau
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Alain Gey
- Université Paris Cité, INSERM U970, PARCC, Paris, France
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Andyara Munoz
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Nesrine Mabrouk
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Nicolas Epaillard
- Medical Oncology Department, Clinique Kuindo Magnin, Nouméa, Nouvelle Calédonie, France
| | - Virginie Verkarre
- Pathology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, INSERM UMR970, Equipe Labellisée Ligue Contre Le Cancer, Paris, France
| | - Yann-Alexandre Vano
- Medical Oncology Department, Hôpital Foch, Suresnes, France
- Centre de Recherche Des Cordeliers, INSERM1138, Inflammation Complément Et Cancer, Université Paris Cité, Paris, France
| | - Eric Tartour
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Immunology Department, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| | - Stéphane Oudard
- Université Paris Cité, INSERM U970, PARCC, Paris, France.
- Medical Oncology Department, Georges Pompidou European Hospital, CARPEM Cancer Institute, AP-HP Centre, Université Paris Cité, Paris, France.
- Paris - Cardiovascular Research Center, European Georges Pompidou Hospital, 56 Rue Leblanc, Paris, 75015, France.
| |
Collapse
|
21
|
Zhang H, Liu Y, Luo P, Wang L, Jiang A. Re: Tracking Nongenetic Evolution from Primary to Metastatic ccRCC: TRACERx Renal. Eur Urol 2025:S0302-2838(25)00059-4. [PMID: 39920003 DOI: 10.1016/j.eururo.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 01/24/2025] [Indexed: 02/09/2025]
Affiliation(s)
- Hanzhong Zhang
- Department of Urology Changhai Hospital Naval Medical University (Second Military Medical University) Shanghai China
| | - Ying Liu
- Department of Urology Changhai Hospital Naval Medical University (Second Military Medical University) Shanghai China
| | - Peng Luo
- Department of Oncology Zhujiang Hospital Southern Medical University Guangzhou China
| | - Linhui Wang
- Department of Urology Changhai Hospital Naval Medical University (Second Military Medical University) Shanghai China.
| | - Aimin Jiang
- Department of Urology Changhai Hospital Naval Medical University (Second Military Medical University) Shanghai China.
| |
Collapse
|
22
|
Guan B, Chu G, Wang Z, Li J, Yi B. Instance-level semantic segmentation of nuclei based on multimodal structure encoding. BMC Bioinformatics 2025; 26:42. [PMID: 39915737 PMCID: PMC11804060 DOI: 10.1186/s12859-025-06066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Accurate segmentation and classification of cell nuclei are crucial for histopathological image analysis. However, existing deep neural network-based methods often struggle to capture complex morphological features and global spatial distributions of cell nuclei due to their reliance on local receptive fields. METHODS This study proposes a graph neural structure encoding framework based on a vision-language model. The framework incorporates: (1) A multi-scale feature fusion and knowledge distillation module utilizing the Contrastive Language-Image Pre-training (CLIP) model's image encoder; (2) A method to transform morphological features of cells into textual descriptions for semantic representation; and (3) A graph neural network approach to learn spatial relationships and contextual information between cell nuclei. RESULTS Experimental results demonstrate that the proposed method significantly improves the accuracy of cell nucleus segmentation and classification compared to existing approaches. The framework effectively captures complex nuclear structures and global distribution features, leading to enhanced performance in histopathological image analysis. CONCLUSIONS By deeply mining the morphological features of cell nuclei and their spatial topological relationships, our graph neural structure encoding framework achieves high-precision nuclear segmentation and classification. This approach shows significant potential for enhancing histopathological image analysis, potentially leading to more accurate diagnoses and improved understanding of cellular structures in pathological tissues.
Collapse
Affiliation(s)
- Bo Guan
- Key Lab for Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China
| | - Guangdi Chu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Ziying Wang
- Department of Medicine, Qingdao University, Qingdao, 266000, China
| | - Jianmin Li
- Key Lab for Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, 300072, China.
| | - Bo Yi
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
23
|
Gao C, Zhou H, Liang W, Wen Z, Liao W, Xie Z, Liao C, He L, Sun J, Chen Z, Li D, Yuan N, Huang C, Zhang J. Proteome-Wide Association Study for Finding Druggable Targets in Progression and Onset of Parkinson's Disease. CNS Neurosci Ther 2025; 31:e70294. [PMID: 40008429 PMCID: PMC11862824 DOI: 10.1111/cns.70294] [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: 03/13/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE To identify and validate causal protein targets that may serve as potential therapeutic interventions for both the onset and progression of Parkinson's disease (PD) through integrative proteomic and genetic analyses. METHOD We utilized large-scale plasma and brain protein quantitative trait loci (pQTL) datasets from the deCODE Health study and the Religious Orders Study/Rush Memory and Aging Project (ROS/MAP), respectively. Proteome-wide association studies (PWAS) were conducted using the OTTERS framework for plasma proteins and the FUSION tool for brain proteins, examining associations with PD onset and three progression phenotypes: composite, motor, and cognitive. Significant protein associations (FDR-corrected p < 0.05) from PWAS were further validated using summary-based Mendelian randomization (SMR), colocalization analyses, and reverse Mendelian randomization (MR) to establish causality. Phenome-wide Mendelian randomization (PheW-MR) was performed to assess potential side effects across 679 disease traits when targeting these proteins to reduce PD-related phenotype risk by 20%. Additionally, we conducted cellular distribution-based clustering using gene expression data from the Allen Brain Atlas (ABA) to explore the distribution of key proteins across brain regions, constructed protein-protein interaction (PPI) networks via the STRING database to explore interactions among proteins, and evaluated the druggability of identified targets using the DrugBank database to identify opportunities for drug repurposing. RESULT Our analyses identified 25 candidate proteins associated with PD phenotypes, including 16 plasma proteins linked to PD progression (10 cognitive, 4 motor, and 3 composite) and 9 plasma proteins associated with PD onset. Notably, GPNMB was implicated in both plasma and brain tissues for PD onset. PheW-MR revealed predominantly beneficial side effects for the identified targets, with 83.7% of associations indicating positive outcomes and 16.3% indicating adverse effects. Cellular clustering categorized candidate targets into three distinct expression profiles across brain cell types using ABA. PPI network analysis highlighted one key interaction cluster among the proteins for PD cognitive progression and PD onset. Druggability assessment revealed 15 out of 25 proteins had repurposing opportunities for PD treatment. CONCLUSION We have identified 25 causal protein targets associated with the onset and progression of PD, providing new insights into the research and development of treatment strategies for PD.
Collapse
Affiliation(s)
- Chenhao Gao
- Department of Neurology, Henan Provincial People's HospitalZhengzhou University People's HospitalZhengzhouHenanChina
| | - Haobin Zhou
- The First School of Clinical MedicineGuangzhou Medical UniversityGuangzhouChina
| | - Weixuan Liang
- The First School of Clinical MedicineGuangzhou Medical UniversityGuangzhouChina
| | - Zhuofeng Wen
- The Sixth School of Clinical MedicineGuangzhou Medical UniversityGuangzhouChina
| | - Wanzhe Liao
- Nanshan School of Guangzhou Medical UniversityGuangzhouChina
| | - Zhixin Xie
- The Second School of Clinical MedicineGuangzhou Medical UniversityGuangzhouChina
| | - Cailing Liao
- School of PediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Limin He
- The Sixth School of Clinical MedicineGuangzhou Medical UniversityGuangzhouChina
| | - Jingzhang Sun
- School of Cyberspace SecurityHainan UniversityHaikouChina
| | - Zhilin Chen
- Department of Breast SurgeryThe First Affiliated Hospital of Hainan Medical UniversityHaikouChina
| | - Duopin Li
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Naijun Yuan
- School of Traditional Chinese MedicineJinan UniversityGuangzhouChina
| | - Chuiguo Huang
- Department of Medicine and Therapeutics, Prince of Wales HospitalThe Chinese University of Hong KongHong KongChina
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's HospitalZhengzhou University People's HospitalZhengzhouHenanChina
| |
Collapse
|
24
|
Cesana B, Cochet C, Filhol O. New players in the landscape of renal cell carcinoma bone metastasis and therapeutic opportunities. Int J Cancer 2025; 156:475-487. [PMID: 39306698 PMCID: PMC11622000 DOI: 10.1002/ijc.35181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 12/07/2024]
Abstract
Approximately one-third of advanced renal cell carcinoma (RCC) patients develop osteolytic bone metastases, leading to skeletal complications. In this review, we first provide a comprehensive perspective of seminal studies on bone metastasis of RCC describing the main molecular modulators and growth factor signaling pathways most important for the RCC-stimulated osteoclast-mediated bone destruction. We next focus on newer developments revealing with in-depth details, the bidirectional interplay between renal cancer cells and the immune and stromal microenvironment that can through epigenetic reprogramming, profoundly affect the behaviors of transformed cells. Understanding their mechanistic interactions is of paramount importance for advancing both fundamental and translational research. These new investigations into the landscape of RCC-bone metastasis offer novel insights and identify potential avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Beatrice Cesana
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Claude Cochet
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| | - Odile Filhol
- University Grenoble Alpes, INSERM, CEA, IRIG‐Biosanté, UMR 1292GrenobleFrance
| |
Collapse
|
25
|
Xu W, Birch G, Meliki A, Moritz V, Bharadwaj M, Schindler NR, Labaki C, Saliby RM, Dinh K, Horst JT, Sun M, Kashima S, Hugaboom M, Dighe A, Machaalani M, Lee GSM, Hurwitz M, McGregor BA, Hirsch MS, Shukla SA, McDermott DF, Signoretti S, Romee R, Choueiri TK, Braun DA. Progressive natural killer cell dysfunction in advanced-stage clear-cell renal cell carcinoma and association with clinical outcomes. ESMO Open 2025; 10:104105. [PMID: 39813824 PMCID: PMC11783098 DOI: 10.1016/j.esmoop.2024.104105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Natural killer (NK) cells are important contributors to antitumor immunity in clear-cell renal cell carcinoma (ccRCC). However, their phenotype, function, and association with clinical outcomes in ccRCC remain poorly understood. MATERIALS AND METHODS We analyzed single-cell RNA sequencing data from 13 primary tumors, 1 localized tumor extension, and 1 metastasis from ccRCC patients at different clinical stages. For each primary tumor specimen, paired normal kidneys were also analyzed. Differential gene expression analysis was carried out to investigate NK cell phenotypes and to derive a gene expression signature. Gene signatures from NK cell subclusters of interest were used to interrogate bulk transcriptomic datasets and expression with clinical outcomes. Finally, tumor-infiltrating NK cell function (cytokine production and cytotoxicity) was assessed by isolation of live NK cells from ccRCC tissue, co-culture with K562 target cells, and measurement of cytokine production (interferon-γ) and cytotoxicity (CD107a) markers by flow cytometry. RESULTS Single-cell transcriptomic data were analyzed from 13 patients with ccRCC (tumor/normal kidney), resulting in 21 139 NK cells. Clustering analysis revealed six NK cell subsets. Bright-like NK cells were significantly enriched in advanced ccRCC compared with localized ccRCC and normal kidney, expressed markers of tissue residency (ZNF683/Hobit, ITGA1/CD49a, CD9, ITGAE/CD103), and had decreased expression of cytotoxicity genes (GZMB/Granzyme-B, PRF1/perforin). In independent cohorts (The Cancer Genome Atlas ccRCC cohort, CheckMate 025), a gene expression score representing this dysfunctional NK cell phenotype was enriched in advanced ccRCC and was associated with worse overall survival. Functional interrogation of tumor-infiltrating NK cells from ccRCC confirmed that tumor-resident CD49a+CD9+ NK cells had impaired cytotoxicity compared with CD49a-CD9- NK cells. CONCLUSIONS A dysfunctional, tumor-resident NK cell phenotype was enriched among patients with metastatic disease and associated with worse survival in patients with advanced ccRCC across multiple patient cohorts. Restoration of NK cell function (via cytokine stimulation or NK cell engineering) could provide a novel avenue for therapeutic intervention against ccRCC.
Collapse
Affiliation(s)
- W Xu
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - G Birch
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - A Meliki
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - V Moritz
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - M Bharadwaj
- Department of Urology, University of Washington School of Medicine, Seattle, USA
| | - N R Schindler
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - C Labaki
- Harvard Medical School, Boston, USA; Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, USA
| | - R M Saliby
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA; Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - K Dinh
- Dana-Farber Cancer Institute, Boston, USA
| | - J T Horst
- Dana-Farber Cancer Institute, Boston, USA
| | - M Sun
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - S Kashima
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - M Hugaboom
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - A Dighe
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - M Machaalani
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - G-S M Lee
- Dana-Farber Cancer Institute, Boston, USA
| | - M Hurwitz
- Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - B A McGregor
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA
| | - M S Hirsch
- Harvard Medical School, Boston, USA; Department of Pathology, Brigham and Women's Hospital, Boston, USA
| | - S A Shukla
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - D F McDermott
- Harvard Medical School, Boston, USA; Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, USA
| | - S Signoretti
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA; Department of Pathology, Brigham and Women's Hospital, Boston, USA
| | - R Romee
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA.
| | - T K Choueiri
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA.
| | - D A Braun
- Yale Cancer Center, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
26
|
Rinaldi L, Senatore E, Feliciello S, Chiuso F, Insabato L, Feliciello A. Kidney cancer: From tumor biology to innovative therapeutics. Biochim Biophys Acta Rev Cancer 2025; 1880:189240. [PMID: 39674419 DOI: 10.1016/j.bbcan.2024.189240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Renal cell carcinoma (RCC) constitutes the most frequent kidney cancer of the adult population and one of the most lethal malignant tumors worldwide. RCC often presents without early symptoms, leading to late diagnosis. Prognosis varies widely based on the stage of cancer at diagnosis. In the early-stage, localized RCC has a relatively good prognosis, while advanced or metastatic RCC has a poor outcome. Obesity, smoking, genetic mutations and family history are all considered risk factors for RCC, while inherited disorders, such as Tuberous Sclerosis and von Hippel-Lindau syndrome, are causally associated with RCC development. Genetic screening, deep sequencing analysis, quantitative proteomics and immunostaining analysis on RCC tissues, biological fluids and blood samples have been employed to identify novel biomarkers, predisposing factors and therapeutic targets for RCC with important clinical implications for patient treatment. Combined approaches of gene-targeting strategies coupled to a deep functional analysis of cancer cell biology, both in vitro and in appropriate animal models of RCC, significantly contributed to identify and characterize relevant pathogenic mechanisms underlying development and progression of RCC. These studies provided also important cues for the generation of novel target-specific therapeutics that selectively restore deranged cancer cell signalling and dysfunctional immune checkpoints, positively impacting on the survival rate of treated RCC patients. In this review, we will describe the recent discoveries concerning the most relevant pathogenic mechanisms of RCC and will highlight novel therapeutic strategies that interrupt oncogenic pathways and restore immune defences in RCC patients.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Stella Feliciello
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
27
|
Yang W, Di S, Yang Z, Cao J, Fu Q, Ren H, Cheng H, Xie Y, Jia W, Dai X, Yu M, Chen Y, Cui X. One-dimensional nanosonosensitizer boosted multiple branches of immune responses against MHC-deficient immune-evasive urologic tumor. SCIENCE ADVANCES 2025; 11:eado7373. [PMID: 39879294 PMCID: PMC11777198 DOI: 10.1126/sciadv.ado7373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/26/2024] [Indexed: 01/31/2025]
Abstract
Cancer immunotherapies rely on CD8+ cytolytic T lymphocytes (CTLs) in recognition and eradication of tumor cells via antigens presented on major histocompatibility complex class I (MHC-I) molecules. However, we observe MHC-I deficiency in human and murine urologic tumors, posing daunting challenges for successful immunotherapy. We herein report an unprecedented nanosonosensitizer of one-dimensional bamboo-like multisegmented manganese dioxide@manganese-bismuth vanadate (BMMBV) to boost multiple branches of immune responses targeting MHC-I-deficient tumors. BMMBV markedly augments sonodynamic activity contributed by manganese heteroatoms in the lattice of bismuth vanadate with narrowing bandgaps. Under sonoirradiation, BMMBV enhances tumor antigen spreading and emission of adjuvant signals, which potentiate dendritic cell maturation, thereby eliciting high aptitude of CTLs. This therapy substantially up-regulates MHC expression on tumor cells, which are reversely sensitive to CTLs. Alongside, extensive innate immune cells complement the cytolytic activity of CTLs for eliminating mouse urologic tumors. This study offers a reinforced strategy against antigen-loss immune-evasive tumor.
Collapse
Affiliation(s)
- Wei Yang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Sichen Di
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Zihuan Yang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| | - Qingqiao Fu
- School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hui Cheng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, P. R. China
| |
Collapse
|
28
|
Patel L, Kolundzic N, Abedalthagafi M. Progress in personalized immunotherapy for patients with brain metastasis. NPJ Precis Oncol 2025; 9:31. [PMID: 39880875 PMCID: PMC11779815 DOI: 10.1038/s41698-025-00812-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Brain metastasis leads to poor outcomes and CNS injury, significantly reducing quality of life and survival rates. Advances in understanding the tumor immune microenvironment have revealed the promise of immunotherapies, which, alongside surgery, chemotherapy, and radiation, offer improved survival for some patients. However, resistance to immunotherapy remains a critical challenge. This review explores the immune landscape of brain metastases, current therapies, clinical trials, and the need for personalized, biomarker-driven approaches to optimize outcomes.
Collapse
Affiliation(s)
- Lalit Patel
- Department of Pathology and Lab Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nikola Kolundzic
- Department of Women & Children's Health, Faculty of Life Sciences & Medicine, King's College London, London, UK
- REPROCELL Europe Ltd., Glasgow, UK
| | - Malak Abedalthagafi
- Department of Pathology and Lab Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
29
|
Lyu F, Zhong Y, He Q, Xiao W, Zhang X. Identification and validation of prognostic biomarkers in ccRCC: immune-stromal score and survival prediction. BMC Cancer 2025; 25:148. [PMID: 39871215 PMCID: PMC11771106 DOI: 10.1186/s12885-025-13534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is integral to tumor progression. However, its prognostic implications and underlying mechanisms in clear cell renal cell carcinoma (ccRCC) are not yet fully elucidated. This study aims to examine the prognostic significance of genes associated with immune-stromal scores and to explore their underlying mechanisms in ccRCC. METHODS Data from the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) were subjected to analysis to compute immune and stromal scores utilizing the ESTIMATE algorithm. The weighted gene co-expression network analysis (WGCNA) was employed to identify gene modules associated with these scores. Differentially expressed genes were assessed using the limma package. Prognostic biomarkers were subsequently identified through univariate, LASSO, and multivariate Cox regression analyses, culminating in the development of a risk score model. Gene expression was confirmed in ccRCC cell lines (786-O, Caki-1) and tumor tissues. Functional assays, such as wound healing and Transwell assays, were employed to evaluate tumor invasion and migration. The prognostic accuracy was assessed through ROC curve analysis, and a nomogram integrating risk scores with clinical variables was constructed. Analyses of immune infiltration, human leukocyte antigen (HLA) expression, immune checkpoint expression, immunophenoscore (IPS), tumor immune dysfunction and exclusion (TIDE) scores, and responses to six targeted therapies were conducted across different risk groups. RESULTS Twelve critical prognostic markers, including CAPRIN1, CXCR3, FERMT3, HAPLN3, HBP1, MACF1, MPEG1, OSCAR, STAT1, UBA7, VAMP1, and VSIG4, were identified. The risk score model exhibited a high degree of predictive accuracy for survival outcomes in ccRCC. Immune profiling revealed significant differences in the TME between risk groups, with high-risk patients displaying elevated expression of HLA and immune checkpoints. Drug sensitivity analyses suggested that high-risk patients had a better response to erlotinib, temsirolimus, axitinib, and sunitinib, whereas low-risk patients demonstrated greater sensitivity to pazopanib. Variability in immunotherapy responsiveness between groups was observed based on IPS and TIDE analyses. CONCLUSION This study highlights the prognostic value and TME-related mechanisms of immune-stromal score signatures in ccRCC, developing a risk score model and nomogram for predicting patient prognosis.
Collapse
Affiliation(s)
- Fang Lyu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuxin Zhong
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qingliu He
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
30
|
Maurer K, Park CY, Mani S, Borji M, Raths F, Gouin KH, Penter L, Jin Y, Zhang JY, Shin C, Brenner JR, Southard J, Krishna S, Lu W, Lyu H, Abbondanza D, Mangum C, Olsen LR, Lawson MJ, Fabani M, Neuberg DS, Bachireddy P, Glezer EN, Farhi SL, Li S, Livak KJ, Ritz J, Soiffer RJ, Wu CJ, Azizi E. Coordinated immune networks in leukemia bone marrow microenvironments distinguish response to cellular therapy. Sci Immunol 2025; 10:eadr0782. [PMID: 39854478 DOI: 10.1126/sciimmunol.adr0782] [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: 06/13/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025]
Abstract
Understanding how intratumoral immune populations coordinate antitumor responses after therapy can guide treatment prioritization. We systematically analyzed an established immunotherapy, donor lymphocyte infusion (DLI), by assessing 348,905 single-cell transcriptomes from 74 longitudinal bone marrow samples of 25 patients with relapsed leukemia; a subset was evaluated by both protein- and transcriptome-based spatial analysis. In acute myeloid leukemia (AML) DLI responders, we identified clonally expanded ZNF683+ CD8+ cytotoxic T lymphocytes with in vitro specificity for patient-matched AML. These cells originated primarily from the DLI product and appeared to coordinate antitumor immune responses through interaction with diverse immune cell types within the marrow microenvironment. Nonresponders lacked this cross-talk and had cytotoxic T lymphocytes with elevated TIGIT expression. Our study identifies recipient bone marrow microenvironment differences as a determinant of an effective antileukemia response and opens opportunities to modulate cellular therapy.
Collapse
Affiliation(s)
- Katie Maurer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cameron Y Park
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Shouvik Mani
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Mehdi Borji
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Yinuo Jin
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Jia Yi Zhang
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Crystal Shin
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - James R Brenner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jackson Southard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachi Krishna
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Wesley Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Haoxiang Lyu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Domenic Abbondanza
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL 60637, USA
| | - Chanell Mangum
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lars Rønn Olsen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | | | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Pavan Bachireddy
- Department of Hematopoietic Biology & Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elham Azizi
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| |
Collapse
|
31
|
Song X, Jiao J, Qin J, Zhang W, Qin W, Ma S. Single-cell transcriptomics reveals the heterogeneity and function of mast cells in human ccRCC. Front Immunol 2025; 15:1494025. [PMID: 39840068 PMCID: PMC11747552 DOI: 10.3389/fimmu.2024.1494025] [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: 09/10/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction The role of mast cells (MCs) in clear cell renal carcinoma (ccRCC) is unclear, and comprehensive single-cell studies of ccRCC MCs have not yet been performed. Methods To investigate the heterogeneity and effects of MCs in ccRCC, we studied single-cell transcriptomes from four ccRCC patients, integrating both single-cell sequencing and bulk tissue sequencing data from online sequencing databases, followed by validation via spatial transcriptomics and multiplex immunohistochemistry (mIHC). Results We identified four MC signature genes (TPSB2, TPSAB1, CPA3, and HPGDS). MC density was significantly greater in ccRCC tissues than in normal tissues, but MC activation characteristics were not significantly different between ccRCC and normal tissues. Activated and resting MCs were defined as having high and low expression of MC receptors and mediators, respectively, whereas proliferating MCs had high expression of proliferation-related genes. The overall percentage of activated MCs in ccRCC tissues did not change significantly but shifted toward a more activated subpopulation (VEGFA+ MCs), with a concomitant decrease in proliferative MCs (TNF+ MCs) and resting MCs. An analysis of the ratio of TNF+/VEGFA+ MCs in tumors revealed that MCs exerted antitumor effects on ccRCC. However, VEGFA+MC was produced in large quantities in ccRCC tissues and promoted tumor angiogenesis compared with adjacent normal tissues, which aroused our concern. In addition, MC signature genes were associated with a better prognosis in the KIRC patient cohort in the TCGA database, which is consistent with our findings. Furthermore, the highest level of IL1B expression was observed in macrophages in ccRCC samples, and spatial transcriptome analysis revealed the colocalization of VEGFA+ MCs with IL1B+ macrophages at the tumor-normal interface. Discussion In conclusion, this study revealed increased MC density in ccRCC. Although the proportion of activated MCs was not significantly altered in ccRCC tissues compared with normal tissues, this finding highlights a shift in the MC phenotype from CTSGhighMCs to more activated VEGFA+MCs, providing a potential therapeutic target for inhibiting ccRCC progression.
Collapse
Affiliation(s)
- Xiyu Song
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jiayang Qin
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shuaijun Ma
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Xijing Innovation Research Institute, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
32
|
Figiel S, Bates A, Braun DA, Eapen R, Eckstein M, Manley BJ, Milowsky MI, Mitchell TJ, Bryant RJ, Sfakianos JP, Lamb AD. Clinical Implications of Basic Research: Exploring the Transformative Potential of Spatial 'Omics in Uro-oncology. Eur Urol 2025; 87:8-14. [PMID: 39227262 DOI: 10.1016/j.eururo.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
New spatial molecular technologies are poised to transform our understanding and treatment of urological cancers. By mapping the spatial molecular architecture of tumours, these platforms uncover the complex heterogeneity within and around individual malignancies, offering novel insights into disease development, progression, diagnosis, and treatment. They enable tracking of clonal phylogenetics in situ and immune-cell interactions in the tumour microenvironment. A whole transcriptome/genome/proteome-level spatial analysis is hypothesis generating, particularly in the areas of risk stratification and precision medicine. Current challenges include reagent costs, harmonisation of protocols, and computational demands. Nonetheless, the evolving landscape of the technology and evolving machine learning applications have the potential to overcome these barriers, pushing towards a future of personalised cancer therapy, leveraging detailed spatial cellular and molecular data. PATIENT SUMMARY: Tumours are complex and contain many different components. Although we have been able to observe some of these differences visually under the microscope, until recently, we have not been able to observe the genetic changes that underpin cancer development. Scientists are now able to explore molecular/genetic differences using approaches such as "spatial transcriptomics" and "spatial proteomics", which allow them to see genetic and cellular variation across a region of normal and cancerous tissue without destroying the tissue architecture. Currently, these technologies are limited by high associated costs, and a need for powerful and complex computational analysis workflows. Future advancements and results through these new technologies may assist patients and their doctors as they make decisions about treating their cancer.
Collapse
Affiliation(s)
- Sandy Figiel
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Anthony Bates
- Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - David A Braun
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Renu Eapen
- Department of Genitourinary Oncology & Division of Cancer Surgery, Peter MacCallum Cancer Centre, The University of Melbourne, Victoria, Australia
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg & Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Brandon J Manley
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Matthew I Milowsky
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tom J Mitchell
- Early Detection Centre, University of Cambridge, Cambridge, UK
| | - Richard J Bryant
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - John P Sfakianos
- Department of Urology, Ichan School of Medicine at the Mount Sinai Hospital, New York, NY, USA
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
33
|
Yoshikawa T, Yanagita M. Single-Cell Analysis Provides New Insights into the Roles of Tertiary Lymphoid Structures and Immune Cell Infiltration in Kidney Injury and Chronic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:40-54. [PMID: 39097168 DOI: 10.1016/j.ajpath.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024]
Abstract
Chronic kidney disease (CKD) is a global health concern with high morbidity and mortality. Acute kidney injury (AKI) is a pivotal risk factor for the progression of CKD, and the rate of AKI-to-CKD progression increases with aging. Intrarenal inflammation is a fundamental mechanism underlying AKI-to-CKD progression. Tertiary lymphoid structures (TLSs), ectopic lymphoid aggregates formed in nonlymphoid organs, develop in aged injured kidneys, but not in young kidneys, with prolonged inflammation and maladaptive repair, which potentially exacerbates AKI-to-CKD progression in aged individuals. Dysregulated immune responses are involved in the pathogenesis of various kidney diseases, such as IgA nephropathy, lupus nephritis, and diabetic kidney diseases, thereby deteriorating kidney function. TLSs also develop in several kidney diseases, including transplanted kidneys and renal cell carcinoma. However, the precise immunologic mechanisms driving AKI-to-CKD progression and development of these kidney diseases remain unclear, which hinders the development of novel therapeutic approaches. This review aims to describe recent findings from single-cell analysis of cellular heterogeneity and complex interactions among immune and renal parenchymal cells, which potentially contribute to the pathogenesis of AKI-to-CKD progression and other kidney diseases, highlighting the mechanisms of formation and pathogenic roles of TLSs in aged injured kidneys.
Collapse
Affiliation(s)
- Takahisa Yoshikawa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan.
| |
Collapse
|
34
|
Yao Y, Liu Y, Lu B, Ji G, Wang L, Dong K, Zhao Z, Lyu D, Wei M, Tu S, Lyu X, Li Y, Huang R, Zhou W, Xu G, Pan X, Cui X. Construction and validation of a regulatory T cells-based classification of renal cell carcinoma: an integrated bioinformatic analysis and clinical cohort study. Cell Oncol (Dordr) 2024:10.1007/s13402-024-01030-9. [PMID: 39714755 DOI: 10.1007/s13402-024-01030-9] [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] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
PURPOSE Renal cell carcinoma (RCC), exhibiting remarkable heterogeneity, can be highly infiltrated by regulatory T cells (Tregs). However, the relationship between Treg and the heterogeneity of RCC remains to be explored. METHODS We acquired single-cell RNA-seq profiles and 537 bulk RNA-seq profiles of TCGA-KIRC cohort. Through clustering, monocle2 pseudotime and prognostic analyses, we identified Treg states-related prognostic genes (TSRPGs), then constructing the RCC Treg states-related prognostic classification (RCC-TSC). We also explored its prognostic significance and multi-omics landmarks. Additionally, we utilized correlation analysis to establish regulatory networks, and predicted candidate inhibitors. More importantly, in Xinhua cohort of 370 patients with kidney neoplasm, we used immunohistochemical (IHC) staining for classification, then employing statistical analyses including Chi-square tests and multivariate Cox proportional hazards regression analysis to explore its clinical relevance. RESULTS We defined 44 TSRPGs in four different monocle states, and identified high immune infiltration RCC (HIRC, LAG3+, Mki67+) as the highly exhausted subtype with the worst prognosis in RCC-TSC (p < 0.001). BATF-LAG3-immune cells axis might be its underlying metastasis-related mechanism. Immunotherapy and inhibitors including sunitinib potentially conferred best therapeutic effects for HIRC. Furthermore, we successfully validated HIRC subtype as an independent prognostic factor within the Xinhua cohort (OS, HR = 16.68, 95% CI = 1.88-148.1, p = 0.011; PFS, HR = 4.43, 95% CI = 1.55-12.6, p = 0.005). CONCLUSION Through integrated bioinformatics analysis and a large-sample retrospective clinical study, we successfully established RCC-TSC and a diagnostic kit, which could stratify RCC patients with different prognosis and to guide personalized treatment.
Collapse
Affiliation(s)
- Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Donghao Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Maodong Wei
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Siqi Tu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xukun Lyu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuanan Li
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Guofeng Xu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
35
|
Huang T, Peng Y, Liu R, Ma B, Chen J, Wei W, Zhong W, Liu Y, Guo S, Han H, Zhou F, Zhang Z, He L, Dong P. Prognostic significance of immune evasion-related genes in clear cell renal cell carcinoma immunotherapy. Int Immunopharmacol 2024; 142:113106. [PMID: 39288623 DOI: 10.1016/j.intimp.2024.113106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) represents a prevalent malignancy of the urinary system. Despite the integration of immune checkpoint inhibitors (ICIs) into the treatment paradigm for advanced RCC, resistance to immunotherapy has emerged as a pivotal determinant impacting the clinical outlook of ccRCC. Accumulating evidence underscores the pivotal role of immune evasion-related genes and pathways in enabling tumor escape from host immune surveillance, consequently influencing patients' responsiveness to immunotherapy. Nonetheless, the clinical relevance of immune evasion-related genes in ccRCC patients undergoing immunotherapy remains inadequately understood. In this study, we aggregated RNA sequencing and clinical data from ccRCC patients across three cohorts: the Cancer Genome Atlas (TCGA), CheckMate cohorts, and the JAVELIN Renal 101 trial. Leveraging a curated immune evasion-related gene set from Lawson et al., we employed the LASSO algorithm and Cox regression analysis to identify eight genes (LPAR6, RGS5, NFYC, PCDH17, CENPW, CNOT8, FOXO3, SNRPB) significantly associated with immune therapy prognosis (HR, 3.57; 95 % CI, 2.38-5.35; P<0.001). A predictive algorithm developed utilizing these genes exhibited notable accuracy in forecasting patients' progression-free survival in the training set (AUC, 0.835). Furthermore, stratification of patients by risk score revealed discernible differences in immunotherapy response and tumor microenvironment. In summary, we present a prognostic model intricately linked with immune status and treatment response. For ccRCC patients undergoing immunotherapy, this approach holds promise in aiding clinical decision-making by providing more precise and tailored treatment recommendations.
Collapse
Affiliation(s)
- Tingxuan Huang
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yulu Peng
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ruiqi Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Binglei Ma
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Junlin Chen
- The School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wensu Wei
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Weifeng Zhong
- Department of Urology, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Yang Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shengjie Guo
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Hui Han
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Fangjian Zhou
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Zhiling Zhang
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| | - Liru He
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Pei Dong
- Department of Urology Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| |
Collapse
|
36
|
Xie D, Han Z, Wang Y, Shi H, Wu X, Wu J, Dai Y. Integrative analysis of bulk and single-cell RNA sequencing reveals sphingolipid metabolism and immune landscape in clear cell renal cell carcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:5391-5404. [PMID: 39230203 DOI: 10.1002/tox.24319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 09/05/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by its aggressive behavior and complex molecular heterogeneity, posing significant challenges for treatment and prognostication. This study offers a comprehensive analysis of ccRCC by leveraging both bulk and single-cell RNA sequencing data, with a specific aim to unravel the complexities of sphingolipid metabolism and the intricate dynamics within the tumor microenvironment (TME). By examining ccRCC samples sourced from public databases, our investigation delves deep into the genetic and transcriptomic landscape of this cancer type. Employing advanced analytical techniques, we have identified pivotal patterns in gene expression and cellular heterogeneity, with a special focus on the roles and interactions of various immune cells within the TME. Significantly, our research has unearthed insights into the dynamics of sphingolipid metabolism in ccRCC, shedding light on its potential implications for tumor progression and strategies for immune evasion. A novel aspect of this study is the development of a risk score model designed to enhance prognostic predictions for ccRCC patients, which is currently pending external validation to ascertain its clinical utility. Despite its contributions, the study is mindful of its limitations, including a reliance on observational data from public sources and a primary focus on RNA sequencing data, which may constrain the depth and generalizability of the findings. The study does not encompass critical aspects, such as protein expression, posttranslational modifications, and comprehensive metabolic profiles. Moreover, its retrospective design underscores the necessity for future prospective studies to solidify these preliminary conclusions. Our findings illuminate the intricate interplay between genetic alterations, sphingolipid metabolism, and immune responses in ccRCC. This research not only enhances our understanding of the molecular foundations of ccRCC but also paves the way for the development of targeted therapies and personalized treatment modalities. The study underlines the importance of cautious interpretation of results and champions ongoing research using diverse methodologies to thoroughly comprehend and effectively combat this formidable cancer type.
Collapse
Affiliation(s)
- Dongdong Xie
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Zhitao Han
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Yu Wang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Haoyu Shi
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiang Wu
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Jiaqing Wu
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| |
Collapse
|
37
|
Wang M, Yao F, Chen N, Wu T, Yan J, Du L, Zeng S, Du C. The advance of single cell transcriptome to study kidney immune cells in diabetic kidney disease. BMC Nephrol 2024; 25:412. [PMID: 39550562 PMCID: PMC11568691 DOI: 10.1186/s12882-024-03853-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent microvascular complication of diabetes mellitus and a primary cause of end-stage renal disease (ESRD). Increasing studies suggest that immune cells are involved in regulating renal inflammation, which contributes to the progression of DKD. Compared with conventional methods, single-cell sequencing technology is more developed technique that has advantages in resolving cellular heterogeneity, parallel multi-omics studies, and discovering new cell types. ScRNA-seq helps researchers to analyze specifically gene expressions, signaling pathways, intercellular communication as well as their regulations in various immune cells of kidney biopsy and urine samples. It is still challenging to investigate the function of each cell type in the pathophysiology of kidney due to its complex and heterogeneous structure and function. Here, we discuss the application of single-cell transcriptomics in the field of DKD and highlight several recent studies that explore the important role of immune cells including macrophage, T cells, B cells etc. in DKD through scRNA-seq analyses. Through combing the researches of scRNA-seq on immune cells in DKD, this review provides novel perspectives on the pathogenesis and immune therapeutic strategy for DKD.
Collapse
Affiliation(s)
- Mengjia Wang
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
| | - Fang Yao
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Ning Chen
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ting Wu
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Jiaxin Yan
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China
| | - Linshan Du
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shijie Zeng
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China
| | - Chunyang Du
- Department of Pathology, Key Laboratory of Kidney Diseases of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China.
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
38
|
Kippenberger M, Schönberg G, Kaczorowski A, Schneider F, Böning S, Sun A, Schwab C, Görtz M, Schütz V, Stenzinger A, Hohenfellner M, Duensing A, Duensing S. Immune landscape of renal cell carcinoma with metastasis to the pancreas. Urol Oncol 2024; 42:373.e9-373.e17. [PMID: 38981801 DOI: 10.1016/j.urolonc.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024]
Abstract
INTRODUCTION Clear cell Renal Cell Carcinoma (ccRCC) has a poor prognosis once metastatic. However, certain metastatic sites have been reported to have a different impact on the patient prognosis. For example, patients with pancreatic metastases have a much more favorable prognosis than those with metastases to other organs. The biological basis for this observation remains poorly understood. The aim of this study was to characterize the immune landscape of pancreatic metastases and the corresponding primary tumors in order to identify possible immunological features that correlate with disease biology. PATIENTS AND METHODS A detailed assessment of immune cell populations was performed using a total of 1,700 microscopic images from ccRCCs from 11 patients, their corresponding pancreatic metastases and ccRCCs from 10 patients without pancreatic metastases. Tumor specimens were stained for CD45, CD8, CD163 and FOXP3 and the densities of the respective immune cells were assessed semiquantitatively in the intratumoral and extratumoral compartment. Multispectral imaging was performed in selected tumors. RESULTS We found that pancreatic metastases show the lowest intratumoral infiltration with CD8+ cytotoxic T lymphocytes of all tumor specimens analyzed. The frequency of CD8+ lymphocytes was on 1.9 fold lower in pancreatic metastases (median density 8.3 cells per field of view [FOV] = 1.23 mm2) when compared to the corresponding primary tumor (15.6 cells per FOV, P = 0.0002) and more than 3-fold lower when compared to ccRCCs without pancreatic metastases (27.2 cells per FOV, P = 0.0012). There was also a significantly reduced intratumoral infiltration with immunosuppressive FOXP3+ lymphocytes in pancreatic metastases (2.6 cells per FOV, P = 0.009) and corresponding primary tumors (2 cells per FOV, P = 0.028) when compared to ccRCCs without pancreatic metastases (5.6 cells per FOV). CONCLUSIONS In this proof-of-concept study, we show that pancreatic metastases of ccRCC present with unique immunological features including a low intratumoral density of CD8+ and FOXP3+ lymphocytes. The low counts of CD8+ and FOXP3+ lymphocytes may reflect less aggressive features of ccRCC with pancreatic metastasis that may result in a more favorable patient prognosis.
Collapse
Affiliation(s)
- Maximilian Kippenberger
- Department of Urology, Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Gita Schönberg
- Department of Urology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Adam Kaczorowski
- Department of Urology, Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Schneider
- Department of Urology, Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sarah Böning
- Department of Urology, Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Angela Sun
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Constantin Schwab
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | | | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Anette Duensing
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA; Department of Urology, Precision Oncology of Urological Malignancies, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, Molecular Urooncology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
39
|
Mantovani A, Marchesi F, Di Mitri D, Garlanda C. Macrophage diversity in cancer dissemination and metastasis. Cell Mol Immunol 2024; 21:1201-1214. [PMID: 39402303 PMCID: PMC11528009 DOI: 10.1038/s41423-024-01216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Invasion and metastasis are hallmarks of cancer. In addition to the well-recognized hematogenous and lymphatic pathways of metastasis, cancer cell dissemination can occur via the transcoelomic and perineural routes, which are typical of ovarian and pancreatic cancer, respectively. Macrophages are a universal major component of the tumor microenvironment and, in established tumors, promote growth and dissemination to secondary sites. Here, we review the role of tumor-associated macrophages (TAMs) in cancer cell dissemination and metastasis, emphasizing the diversity of myeloid cells in different tissue contexts (lungs, liver, brain, bone, peritoneal cavity, nerves). The generally used models of lung metastasis fail to capture the diversity of pathways and tissue microenvironments. A better understanding of TAM diversity in different tissue contexts may pave the way for tailored diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.
- William Harvey Research Institute, Queen Mary University, London, UK.
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| |
Collapse
|
40
|
Stransky LA, Gao W, Schmidt LS, Bi K, Ricketts CJ, Ramesh V, James A, Difilippantonio S, Ileva L, Kalen JD, Karim B, Jeon A, Morgan T, Warner AC, Turan S, Unite J, Tran B, Choudhari S, Zhao Y, Linn DE, Yun C, Dhandapani S, Parab V, Pinheiro EM, Morris N, He L, Vigeant SM, Pignon JC, Sticco-Ivins M, Signoretti S, Van Allen EM, Linehan WM, Kaelin WG. Toward a CRISPR-based mouse model of Vhl-deficient clear cell kidney cancer: Initial experience and lessons learned. Proc Natl Acad Sci U S A 2024; 121:e2408549121. [PMID: 39365820 PMCID: PMC11474080 DOI: 10.1073/pnas.2408549121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
CRISPR is revolutionizing the ability to do somatic gene editing in mice for the purpose of creating new cancer models. Inactivation of the VHL tumor suppressor gene is the signature initiating event in the most common form of kidney cancer, clear cell renal cell carcinoma (ccRCC). Such tumors are usually driven by the excessive HIF2 activity that arises when the VHL gene product, pVHL, is defective. Given the pressing need for a robust immunocompetent mouse model of human ccRCC, we directly injected adenovirus-associated viruses (AAVs) encoding sgRNAs against VHL and other known/suspected ccRCC tumor suppressor genes into the kidneys of C57BL/6 mice under conditions where Cas9 was under the control of one of two different kidney-specific promoters (Cdh16 or Pax8) to induce kidney tumors. An AAV targeting Vhl, Pbrm1, Keap1, and Tsc1 reproducibly caused macroscopic ccRCCs that partially resembled human ccRCC tumors with respect to transcriptome and cell of origin and responded to a ccRCC standard-of-care agent, axitinib. Unfortunately, these tumors, like those produced by earlier genetically engineered mouse ccRCCs, are HIF2 independent.
Collapse
Affiliation(s)
- Laura A. Stransky
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Wenhua Gao
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Laura S. Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Kevin Bi
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - Vijyendra Ramesh
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Amy James
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Simone Difilippantonio
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Albert Jeon
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Sevilay Turan
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Joanne Unite
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Bao Tran
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Sulbha Choudhari
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | - Yongmei Zhao
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | | | - Changhong Yun
- Pharmacokinetics, Merck & Co., Inc., Boston, MA02115
| | | | - Vaishali Parab
- Pharmacokinetics, Merck & Co., Inc., South San Francisco, CA94080
| | | | - Nicole Morris
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lixia He
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Sean M. Vigeant
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Jean-Christophe Pignon
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
| | - Maura Sticco-Ivins
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Eliezer M. Van Allen
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - William G. Kaelin
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
41
|
Maksimovic S, Boscolo NC, La Posta L, Barrios S, Moussa MJ, Gentile E, Pesquera PI, Li W, Chen J, Gomez JA, Basi A, Burks JK, Alvarez-Breckenridge C, Gao J, Campbell MT, Dondossola E. Antiangiogenic Tyrosine Kinase Inhibitors have Differential Efficacy in Clear Cell Renal Cell Carcinoma in Bone. CANCER RESEARCH COMMUNICATIONS 2024; 4:2621-2637. [PMID: 39248577 PMCID: PMC11459607 DOI: 10.1158/2767-9764.crc-24-0304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney neoplasm; bone metastasis (BM) develops in 35% to 40% of metastatic patients and results in substantial morbidity and mortality, as well as medical costs. A key feature of ccRCC is the loss of function of the von Hippel-Lindau protein, which enhances angiogenesis via vascular endothelial growth factor release. Consequently, antiangiogenic tyrosine kinase inhibitors (TKI) emerged as a treatment for ccRCC. However, limited data about their efficacy in BM is available, and no systematic comparisons have been performed. We developed mouse models of bone and lung ccRCC tumors and compared their anticancer efficacy, impact on mouse survival, and mechanisms of action, including effects on tumor cells and both immune and nonimmune (blood vessels and osteoclasts) bone stromal components. This approach elucidates the efficacy of TKIs in ccRCC bone tumors to support rational interrogation and development of therapies. SIGNIFICANCE TKIs showed different efficacy in synchronous bone and lung metastases and did not eradicate tumors as single agents but induced extensive reprogramming of the BM microenvironment. This resulted in a significant decrease in neoangiogenic blood vessels, bone remodeling, and immune cell infiltration (including CD8 T cells) with altered spatial distribution.
Collapse
Affiliation(s)
- Stefan Maksimovic
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Nina C. Boscolo
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ludovica La Posta
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Bioengineering, Rice University, Houston, Texas.
| | - Mohammad Jad Moussa
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Emanuela Gentile
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Pedro I. Pesquera
- Division of Surgery, Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Wenjiao Li
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jianfeng Chen
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Javier A. Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Akshay Basi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Jianjun Gao
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
42
|
Malik S, Sureka N, Ahuja S, Aden D, Zaheer S, Zaheer S. Tumor-associated macrophages: A sentinel of innate immune system in tumor microenvironment gone haywire. Cell Biol Int 2024; 48:1406-1449. [PMID: 39054741 DOI: 10.1002/cbin.12226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
The tumor microenvironment (TME) is a critical determinant in the initiation, progression, and treatment outcomes of various cancers. Comprising of cancer-associated fibroblasts (CAF), immune cells, blood vessels, and signaling molecules, the TME is often likened to the soil supporting the seed (tumor). Among its constituents, tumor-associated macrophages (TAMs) play a pivotal role, exhibiting a dual nature as both promoters and inhibitors of tumor growth. This review explores the intricate relationship between TAMs and the TME, emphasizing their diverse functions, from phagocytosis and tissue repair to modulating immune responses. The plasticity of TAMs is highlighted, showcasing their ability to adopt either protumorigenic or anti-tumorigenic phenotypes based on environmental cues. In the context of cancer, TAMs' pro-tumorigenic activities include promoting angiogenesis, inhibiting immune responses, and fostering metastasis. The manuscript delves into therapeutic strategies targeting TAMs, emphasizing the challenges faced in depleting or inhibiting TAMs due to their multifaceted roles. The focus shifts towards reprogramming TAMs to an anti-tumorigenic M1-like phenotype, exploring interventions such as interferons, immune checkpoint inhibitors, and small molecule modulators. Noteworthy advancements include the use of CSF1R inhibitors, CD40 agonists, and CD47 blockade, demonstrating promising results in preclinical and clinical settings. A significant section is dedicated to Chimeric Antigen Receptor (CAR) technology in macrophages (CAR-M cells). While CAR-T cells have shown success in hematological malignancies, their efficacy in solid tumors has been limited. CAR-M cells, engineered to infiltrate solid tumors, are presented as a potential breakthrough, with a focus on their development, challenges, and promising outcomes. The manuscript concludes with the exploration of third-generation CAR-M technology, offering insight into in-vivo reprogramming and nonviral vector approaches. In conclusion, understanding the complex and dynamic role of TAMs in cancer is crucial for developing effective therapeutic strategies. While early-stage TAM-targeted therapies show promise, further extensive research and larger clinical trials are warranted to optimize their targeting and improve overall cancer treatment outcomes.
Collapse
Affiliation(s)
- Shaivy Malik
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, Jamia Hamdard, New Delhi, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| |
Collapse
|
43
|
Sobottka B, Vetter V, Banaei-Esfahani A, Nowak M, Lorch A, Sirek A, Mertz KD, Brunelli M, Berthold D, de Leval L, Kahraman A, Koelzer VH, Moch H. Immune Phenotype-Genotype Associations in Primary Clear Cell Renal Cell Carcinoma and Matched Metastatic Tissue. Mod Pathol 2024; 37:100558. [PMID: 38969270 DOI: 10.1016/j.modpat.2024.100558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
Adjuvant immunotherapy has been recently recommended for patients with metastatic clear cell renal cell carcinoma (ccRCC), but there are no tissue biomarkers to predict treatment response in ccRCC. Potential predictive biomarkers are mainly assessed in primary tumor tissue, whereas metastases (METs) remain understudied. To explore potential differences between genomic alterations and immune phenotypes in primary tumors and their matched METs, we analyzed primary tumors (PTs) of 47 ccRCC patients and their matched distant METs by comprehensive targeted parallel sequencing, whole-genome copy number variation analysis, determination of microsatellite instability, and tumor mutational burden. We quantified the spatial distribution of tumor-infiltrating CD8+ T cells and coexpression of the T-cell-exhaustion marker thymocyte selection-associated high mobility group box (TOX) by digital immunoprofiling and quantified tertiary lymphoid structures. Most METs were pathologically "cold." Inflamed, pathologically "hot" PTs were associated with decreased disease-free survival, worst for patients with high levels of CD8+TOX+ T cells. Interestingly, inflamed METs showed a relative increase in exhausted CD8+TOX+ T cells and increased accumulative size of tertiary lymphoid structures compared with PTs. Integrative analysis of molecular and immune phenotypes revealed BAP1 and CDKN2A/B deficiency to be associated with an inflamed immune phenotype. Our results highlight the distinct spatial distribution and differentiation of CD8+ T cells at metastatic sites, and the association of an inflamed microenvironment with specific genomic alterations.
Collapse
Affiliation(s)
- Bettina Sobottka
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Viola Vetter
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Amir Banaei-Esfahani
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anja Lorch
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Andrej Sirek
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland; Institute of Medical Genetics and Pathology, University Hospital of Basel, Basel, Switzerland
| | - Matteo Brunelli
- Università di Verona, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Dominik Berthold
- Department of Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Abdullah Kahraman
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; School of Life Sciences Fachhochschule Nordwestschweiz, Institute for Chemistry and Bioanalytics, Muttenz, Switzerland
| | - Viktor Hendrik Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Institute of Medical Genetics and Pathology, University Hospital of Basel, Basel, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Huang Z, Chen T, Li W, He W, Liu S, Wu Z, Li B, Yuan Y, Qiu J. Atezolizumab and bevacizumab plus transarterial chemoembolization and hepatic arterial infusion chemotherapy for patients with high tumor burden unresectable hepatocellular carcinoma: A multi-center cohort study. Int Immunopharmacol 2024; 139:112711. [PMID: 39029233 DOI: 10.1016/j.intimp.2024.112711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Though atezolizumab plus bevacizumab (A+B) offer promise for unresectable hepatocellular carcinoma (uHCC) treatment, the response rate remains suboptimal. Our previous studies highlighted the potential of transarterial chemoembolization (TACE) when combined with FOLFOX-based hepatic arterial infusion chemotherapy (HAIC) in HCC treatment. This study aims to evaluate the safety and efficacy of A+B plus TACE-HAIC for high tumor burden uHCC (HTB-uHCC). METHODS This three-center retrospective study involved 82 HTB-uHCC patients administered with TACE-HAIC followed by A+B. We characterized HTB-uHCC patients as those surpassing the up-to-11 criteria, exhibiting VP 3-4, or presenting extrahepatic metastases. The primary outcomes were the objective response rate (ORR) and progression-free survival (PFS). Secondary outcomes encompassed the incidence of treatment-related adverse events (TRAEs) and overall survival (OS). RESULTS Employing the mRECIST criteria, the ORR was 62.2 %, wherein 18 (22.0 %) patients achieved complete response, 33 (40.2 %) demonstrated partial response, 21 (25.6 %) maintained stable disease, and 10 (12.2 %) exhibited disease progression. Impressively, 11 (13.4 %) patients were converted to resectable HCC and underwent curative hepatectomy. The median PFS was 10.1 months (95 % CI, 8.4 to NA), and the median OS was still pending. At the one-year mark, the OS and PFS rates were 92.8 % (95 % CI, 86.1 to 100.0) and 42.9 % (95 % CI, 31.3 to 58.7), respectively. 79 (96.3 %) experienced TRAEs, and 39 (47.6 %) had grade 3-4 TRAEs, though no treatment-related death was recorded. CONCLUSIONS The findings underscore the potential of the A+B and TACE-HAIC combined treatment for HTB-uHCC patients, marking it as a viable therapeutic option, given its potent efficacy and tolerable safety profile.
Collapse
Affiliation(s)
- Zhenkun Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tiejun Chen
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Wenbin Li
- Department of Biliopancreatic Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Wei He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shaoru Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zongfeng Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China; Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
45
|
Wang B, Wei Z, Xu M, Shu H, Fan Z. Identification of key ferroptosis genes and subtypes in kidney renal clear cell carcinoma. Discov Oncol 2024; 15:492. [PMID: 39331243 PMCID: PMC11436560 DOI: 10.1007/s12672-024-01363-9] [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: 04/26/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Tumour immunity is highly important for the occurrence and development of tumours, and many cancers are resistant to ferroptosis. This study aims to explore the relationship between ferroptosis-related genes (FRGs) and the immunological characteristics of kidney renal clear cell carcinoma (KIRC). We obtained RNA-seq profiles and clinical data of KIRC patients from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and identified CD44 and GLRX5 as the key FRGs involved in KIRC immune infiltration through Spearman's correlation analysis. Based on the expression of CD44 and GLRX5, the consensus clustering algorithm was used to classify the TCGA-KIRC samples into two clusters. A nomogram was constructed to evaluate the prognosis of KIRC patients. ESTIMATE, CIBERSORT, and single-sample gene set enrichment analysis (ssGSEA) were performed to evaluate immune infiltration between the two clusters. A weighted gene co-expression network analysis (WGCNA) was used to identify the most relevant genes to the clusters and immunity. Then, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed. The external dataset GSE53757 was used to validate the immunological features between the two clusters. Cluster 2 patients had more active immune infiltration and might be more sensitive to immunotherapy; Cluster 2 patients also had a worse prognosis and might be at a more advanced stage of KIRC. We identified key ferroptosis-related genes and subgroups involved in the immune infiltration of KIRC, which is highly important for exploring the molecular mechanisms and treatments of KIRC.
Collapse
Affiliation(s)
- Biao Wang
- Department of Urology, The Central Hospital of Xiaogan, Xiaogan, 432000, Hubei, China
| | - Zhuo Wei
- Department of Urology, The Central Hospital of Xiaogan, Xiaogan, 432000, Hubei, China
| | - Man Xu
- Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Hui Shu
- Department of Urology, The Central Hospital of Xiaogan, Xiaogan, 432000, Hubei, China.
| | - Zheqi Fan
- Department of Urology, The Central Hospital of Xiaogan, Xiaogan, 432000, Hubei, China.
| |
Collapse
|
46
|
Heidari-Foroozan M, Rezalotfi A, Rezaei N. The molecular landscape of T cell exhaustion in the tumor microenvironment and reinvigoration strategies. Int Rev Immunol 2024; 43:419-440. [PMID: 39257319 DOI: 10.1080/08830185.2024.2401352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/31/2023] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
Immunotherapy has emerged as a promising therapeutic approach for cancer treatment by harnessing the immune system to target cancer cells. However, the efficacy of immunotherapy is hindered by the tumor microenvironment (TME), comprising regulatory T cells (Tregs), macrophages, myeloid-derived suppressor cells (MDSCs), neutrophils, soluble factors (TGF-β, IL-35, IL-10), and hypoxia. These components interact with inhibitory receptors (IRs) on T cells, leading to alterations in T cell transcriptomes, epigenomes, and metabolism, ultimately resulting in T cell exhaustion and compromising the effectiveness of immunotherapy. T cell exhaustion occurs in two phases: pre-exhaustion and exhaustion. Pre-exhausted T cells exhibit reversibility and distinct molecular properties compared to terminally exhausted T cells. Understanding these differences is crucial for designing effective interventions. This comprehensive review summarizes the characteristics of pre-exhausted and exhausted T cells and elucidates the influence of TME components on T cell activity, transcriptomes, epigenomes, and metabolism, ultimately driving T cell exhaustion in cancer. Additionally, potential intervention strategies for reversing exhaustion are discussed. By gaining insights into the mechanisms underlying T cell exhaustion and the impact of the TME, this review aims to inform the development of innovative approaches for combating T cell exhaustion and enhancing the efficacy of immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Mahsa Heidari-Foroozan
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alaleh Rezalotfi
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Zhang Z, Xiao Y, Zhao S, Liu J, Zeng J, Xiao F, Liao B, Shan X, Zhu H, Guo H. FAM109B plays a tumorigenic role in low-grade gliomas and is associated with tumor-associated macrophages (TAMs). J Transl Med 2024; 22:833. [PMID: 39256832 PMCID: PMC11389277 DOI: 10.1186/s12967-024-05641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Family with sequence similarity 109, member B (FAM109B) is involved in endocytic transport and affects genetic variation in brain methylation. It is one of the important genes related to immune cell-associated diseases. In the tumor immune system, methylation can regulate tumor immunity and influence the maturation and functional response of immune cells. Whether FAM109B is involved in tumor progression and its correlation with the tumor immune microenvironment has not yet been disclosed. METHODS A comprehensive pan-cancer analysis of FAM109B expression, prognosis, immunity, and TMB was conducted. The expression, clinical features, and prognostic value of FAM109B in low-grade gliomas (LGG) were evaluated using TCGA, CGGA, and Gravendeel databases. The expression of FAM109B was validated by qRT-PCR, immunohistochemistry (IHC), and Western blotting (WB). The relationship between FAM109B and methylation, Copy Number Variation (CNV), prognosis, immune checkpoints (ICs), and common chemotherapy drug sensitivity in LGG was explored through Cox regression, Kaplan-Meier curves, and Spearman correlation analysis. FAM109B levels and their distribution were studied using the TIMER database and single-cell analysis. The potential role of FAM109B in gliomas was further investigated through in vitro and in vivo experiments. RESULTS FAM109B was significantly elevated in various tumor types and was associated with poor prognosis. Its expression was related to aggressive progression and poor prognosis in low-grade glioma patients, serving as an independent prognostic marker for LGG. Glioma grade was negatively correlated with FAM109B DNA promoter methylation. Immune infiltration and single-cell analysis showed significant expression of FAM109B in tumor-associated macrophages (TAMs). The expression of FAM109B was closely related to gene mutations, immune checkpoints (ICs), and chemotherapy drugs in LGG. In vitro studies showed increased FAM109B expression in LGG, closely related to cell proliferation. In vivo studies showed that mice in the sh-FAM109B group had slower tumor growth, slower weight loss, and longer survival times. CONCLUSIONS FAM109B, as a novel prognostic biomarker for low-grade gliomas, exhibits specific overexpression in TAMs and may be a potential therapeutic target for LGG patients.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Yao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Siyi Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Jun Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Jie Zeng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Xuesong Shan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
- Institute of Neuroscience, Nanchang University, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China.
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
- Institute of Neuroscience, Nanchang University, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China.
| |
Collapse
|
48
|
Zheng X, Sun R, Wei T. Immune microenvironment in papillary thyroid carcinoma: roles of immune cells and checkpoints in disease progression and therapeutic implications. Front Immunol 2024; 15:1438235. [PMID: 39290709 PMCID: PMC11405226 DOI: 10.3389/fimmu.2024.1438235] [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: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of primary thyroid cancer. Despite the low malignancy and relatively good prognosis, some PTC cases are highly aggressive and even develop refractory cancer in the thyroid. Growing evidence suggested that microenvironment in tumor affected PTC biological behavior due to different immune states. Different interconnected components in the immune system influence and participate in tumor invasion, and are closely related to PTC metastasis. Immune cells and molecules are widely distributed in PTC tissues. Their quantity and proportion vary with the host's immune status, which suggests that immunotherapy may be a very promising therapeutic modality for PTC. In this paper, we review the role of immune cells and immune checkpoints in PTC immune microenvironment based on the characteristics of the PTC tumor microenvironment.
Collapse
Affiliation(s)
- Xun Zheng
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruonan Sun
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tao Wei
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Jiang A, Liu W, Liu Y, Hu J, Zhu B, Fang Y, Zhao X, Qu L, Lu J, Liu B, Qi L, Cai C, Luo P, Wang L. DCS, a novel classifier system based on disulfidptosis reveals tumor microenvironment heterogeneity and guides frontline therapy for clear cell renal carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:263-279. [PMID: 39281723 PMCID: PMC11401502 DOI: 10.1016/j.jncc.2024.06.003] [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: 05/29/2023] [Revised: 04/26/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Background Emerging evidence suggests that cell deaths are involved in tumorigenesis and progression, which may be treated as a novel direction of cancers. Recently, a novel type of programmed cell death, disulfidptosis, was discovered. However, the detailed biological and clinical impact of disulfidptosis and related regulators remains largely unknown. Methods In this work, we first enrolled pancancer datasets and performed multi-omics analysis, including gene expression, DNA methylation, copy number variation and single nucleic variation profiles. Then we deciphered the biological implication of disulfidptosis in clear cell renal cell carcinoma (ccRCC) by machine learning. Finally, a novel agent targeting at disulfidptosis in ccRCC was identified and verified. Results We found that disulfidptosis regulators were dysregulated among cancers, which could be explained by aberrant DNA methylation and genomic mutation events. Disulfidptosis scores were depressed among cancers and negatively correlated with epithelial mesenchymal transition. Disulfidptosis regulators could satisfactorily stratify risk subgroups in ccRCC, and a novel subtype, DCS3, owning with disulfidptosis depression, insensitivity to immune therapy and aberrant genome instability were identified and verified. Moreover, treating DCS3 with NU1025 could significantly inhibit ccRCC malignancy. Conclusion This work provided a better understanding of disulfidptosis in cancers and new insights into individual management based on disulfidptosis.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Junyi Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xuenan Zhao
- Center for Translational Medicine, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Juan Lu
- Vocational Education Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bing Liu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Chen Cai
- Department of Special Clinic, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
50
|
Zhu Q, Yang Y, Zeng Y, Chen K, Zhang Q, Wang L, Huang Y, Jian S. The significance of CD8 + tumor-infiltrating lymphocytes exhaustion heterogeneity and its underlying mechanism in diffuse large B-cell lymphoma. Int Immunopharmacol 2024; 137:112447. [PMID: 38909497 DOI: 10.1016/j.intimp.2024.112447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/25/2024]
Abstract
CD8+ tumor-infiltrating lymphocytes (TILs) exhaustion is a major barrier to effective tumor control in diffuse large B-cell lymphoma (DLBCL) and may consist of heterogeneous populations with different functional states. We profiled the CD8+TILs exhaustion heterogeneity and explored its clinical significance as well as the underlying mechanism through single-cell RNA sequencing (n = 7), bulk RNA sequencing (n = 3300), immunohistochemistry (n = 116), and reverse transcription-quantitative polymerase chain reaction (n = 95), and somatic mutation data (n = 48). Our results demonstrated that exhausted CD8+TILs in DLBCL were composed of progenitor and terminal states characterized by CCL5 and TUBA1B, respectively. High terminally exhausted CD8+TILs indicated an immunosuppressive tumor microenvironment, activated B-cell-like subtype, inferior prognosis, and poor response to immune checkpoint blockade therapy in DLBCL. Our study further demonstrated that the CD39/A2AR-related signaling may be the potential pathway that promoted the transition of progenitor toward terminally exhausted CD8+TILs in DLBCL. Furthermore, the CD39/A2AR-related pathway in DLBCL may be regulated by BATF and STAT3 in exhausted CD8+TILs, and MYD88 mutation in tumor cells. Our study highlights CD8+TILs exhaustion heterogeneity and its possible regulatory mechanism provides a novel prognostic indicator and can facilitate the optimization of individualized immunotherapy.
Collapse
Affiliation(s)
- Qiqi Zhu
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pathology, Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Yiming Yang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Key Lab of Process Analysis and Control of Sichuan Universities, Yibin University, Yibin, Sichuan 644000, China
| | - Yi Zeng
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Kexin Chen
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Qiaoyu Zhang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Li Wang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pathology, Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Yifan Huang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pathology, Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Shunhai Jian
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pathology, Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|