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
|
Liu Y, Dong K, Yao Y, Lu B, Wang L, Ji G, Zhang H, Zhao Z, Yang X, Huang R, Zhou W, Pan X, Cui X. Construction and validation of renal cell carcinoma tumor cell differentiation-related prognostic classification (RCC-TCDC): an integrated bioinformatic analysis and clinical study. Ann Med 2025; 57:2490830. [PMID: 40248945 PMCID: PMC12010653 DOI: 10.1080/07853890.2025.2490830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a heterogeneous malignancy with diverse gene expression patterns, molecular landscapes, and differentiation characteristics of tumor cells. It is imperative to develop molecular RCC classification based on tumor cell differentiation for precise risk stratification and personalized therapy. METHODS We obtained scRNA-seq profiles from GSE159115 and bulk RNA-seq profiles from TCGA-KIRC cohort. We then performed scRNA-seq cluster analysis, monocle2 pseudotime analysis, and prognostic analysis to obtain tumor cell differentiation-related prognostic genes (TCDGs). Subsequently, we conducted consensus clustering to construct the RCC tumor cell differentiation-related prognostic classification (RCC-TCDC) and implemented prognostic and multi-omics analyses. Moreover, we utilized Lasso regression to help develop a multivariable prognostic model. In addition, we performed correlation analysis and Cmap algorithm for regulatory network establishment and candidate inhibitor prediction. We eventually included 370 kidney neoplasm patients in Xinhua cohort to undergo immunohistochemical staining and scoring for classification and comprehensive statistical analyses, including Chi-square tests, Kaplan-Meier survival analyses, and multivariable Cox regression analysis . RESULTS 32 TCDGs were identifiedand RCC-TCDC was constructed to classify TCGA-KIRC patients into RCC-low differentiation (RCC-LD) (S100A11+ SH3BGRL3+, high risk), RCC-moderate differentiation (TSPAN7+, medium risk), and RCC-high differentiation (RCC-HD) (AQP1+ NPR3+, low risk). Notably, RCC-LD was validated as anindependent risk factor for both OS (p = 0.015, HR = 14.0, 95%CI = 1.67-117.8) and PFS (p = 0.010, HR = 4.0, 95%CI = 1.39-11.7) of RCC patients in Xinhua cohort, taking RCC-HD as reference. CONCLUSIONS We constructed and validated a robust molecular classification system, RCC-TCDC, elucidating three distinct RCC subtypes.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingnan Lu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo Ji
- Department of Pathology, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wang Zhou
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Shaikh S, Zhao X, Wagner RT, Pan X, Hlady RA, Wang L, Ho TH, Robertson KD. Deciphering the interplay between SETD2 mediated H3K36me3 and RNA N6-methyladenosine in clear cell renal cell carcinoma (ccRCC). Epigenetics 2025; 20:2456418. [PMID: 39874221 PMCID: PMC11776469 DOI: 10.1080/15592294.2025.2456418] [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: 08/25/2024] [Revised: 12/21/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
RNA N6-methyladenosine (m6A) plays diverse roles in RNA metabolism and its deregulation contributes to tumor initiation and progression. Clear cell renal cell carcinoma (ccRCC) is characterized by near ubiquitous loss of VHL followed by mutations in epigenetic regulators PBRM1, SETD2, and BAP1. Mutations in SETD2, a histone H3 lysine 36 trimethylase (H3K36me3), are associated with reduced survival, greater metastatic propensity, and metabolic reprogramming. While m6A and H3K36me3 deregulation are separately implicated in renal tumorigenesis, H3K36me3 may participate directly in m6A targeting, but the m6A-H3K36me3 interplay has not been investigated in the context of ccRCC. Using RCC-relevant SETD2 isogenic knockout and rescue cell line models, we demonstrate a dynamic redistribution of m6A in the SETD2 depleted transcriptome, with a subset of transcripts involved in metabolic reprogramming demonstrating SETD2 dependent m6A and expression level changes. Using a panel of six histone modifications we show that m6A redistributes to regions enriched in gained active enhancers upon SETD2 inactivation. Finally, we demonstrate a reversal of transcriptomic programs involved in SETD2 loss mediated metabolic reprogramming, and reduced cell viability through pharmacologic inhibition or genetic ablation of m6A writer METTL3 specific to SETD2 deficient cells. Thus, targeting m6A may represent a novel therapeutic vulnerability in SETD2 mutant ccRCC.
Collapse
Affiliation(s)
- Shafiq Shaikh
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Xia Zhao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ryan T. Wagner
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Xiaoyu Pan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ryan A. Hlady
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Thai H. Ho
- Division of Hematology and Oncology, Medical University of South Carolina, Charleston, SC, USA
| | - Keith D. Robertson
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
4
|
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
|
5
|
Xu L, Wang S, Li D, Yang B, Zhang J, Ran L, Liu S, Zhang Q, Li B. Dual targeting of ENPP3 and SIRPα with a bispecific antibody enhances macrophage-mediated immunity in renal cell carcinoma. Biochem Biophys Res Commun 2025; 769:151955. [PMID: 40349459 DOI: 10.1016/j.bbrc.2025.151955] [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: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Renal cell carcinoma (RCC) remains a therapeutic challenge despite recent immunotherapy advances. We identified ENPP3 and SIRPα as significantly overexpressed in RCC tissues with positive correlation and prognostic relevance. Based on these findings, we developed a novel bispecific antibody simultaneously targeting tumor-associated ENPP3 and macrophage checkpoint SIRPα. The ENPP3-SIRPα bispecific antibody demonstrated specific binding to both targets and effectively blocked CD47-SIRPα interaction in vitro. In vivo this bispecific approach exhibited superior anti-tumor efficacy compared to monotherapies or their combination as separate agents. Mechanistic studies confirmed that the therapeutic effect was macrophage-dependent, with enhanced phagocytosis of tumor cells. Importantly, the bispecific antibody maintained a favorable safety profile with no significant hematological abnormalities observed during treatment. These findings demonstrate that simultaneous targeting of ENPP3 and SIRPα represents a promising immunotherapeutic strategy for RCC, combining tumor-specific targeting with immune checkpoint inhibition while mitigating potential toxicities associated with systemic SIRPα blockade.
Collapse
MESH Headings
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/therapy
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/therapy
- Humans
- Receptors, Immunologic/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/metabolism
- Macrophages/immunology
- Macrophages/drug effects
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/pharmacology
- Cell Line, Tumor
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- Mice
- Immunotherapy
- Phagocytosis/drug effects
- Female
Collapse
Affiliation(s)
- Lijun Xu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shanlong Wang
- The Second Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Dan Li
- General Psychiatric Department, Henan Rongkang Hospital, Luoyang, Henan, China
| | - Bowen Yang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Junhan Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Longchao Ran
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shujian Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Qi Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Bingyu Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan, China; The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
6
|
Lou Y, Dong C, Jiang Q, He Z, Yang S. Protein succinylation mechanisms and potential targeted therapies in urinary disease. Cell Signal 2025; 131:111744. [PMID: 40090556 DOI: 10.1016/j.cellsig.2025.111744] [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: 12/13/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
Succinylation is a relatively common post-translational modification. It occurs in the cytoplasm, mitochondria, and the nucleus, where its essential precursor, succinyl-CoA, is present, allowing for the modification of non-histone and histone proteins. In normal cells, succinylation levels are carefully regulated to sustain a dynamic balance, necessitating the involvement of various regulatory mechanisms, including non-enzymatic reactions, succinyltransferases, and desuccinylases. Among these regulatory factors, sirtuin 5, the first identified desuccinylase, plays a significant role and has been extensively researched. The level of succinylation has a significant effect on multiple metabolic pathways, including the tricarboxylic acid cycle, redox balance, and fatty acid metabolism. Dysregulated succinylation can contribute to the progression or exacerbation of various urinary diseases. Succinylation predominantly affects disease progression by altering the expression of key genes and modulating the activity of enzymes involved in vital metabolic processes. Desuccinylases primarily affect enzymes associated with Warburg's effect, thereby affecting the energy supply of tumor cells, while succinyltransferases can regulate gene transcription to alter cell phenotype, thereby involving the development of urinary diseases. Considering these effects, targeting succinylation-related enzymes to regulate metabolic pathways or gene expression may offer a promising therapeutic strategy for treating urinary diseases.
Collapse
Affiliation(s)
- Yuanquan Lou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Caitao Dong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Qinhong Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China.
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
7
|
Hashemi M, Gholamrezaie H, Ziyaei F, Asadi S, Naeini ZY, Salimian N, Enayat G, Sharifi N, Aliahmadi M, Rezaie YS, Khoushab S, Rahimzadeh P, Miri H, Abedi M, Farahani N, Taheriazam A, Nabavi N, Entezari M. Role of lncRNA PVT1 in the progression of urological cancers: Novel insights into signaling pathways and clinical opportunities. Cell Signal 2025; 131:111736. [PMID: 40081549 DOI: 10.1016/j.cellsig.2025.111736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Urologic malignancies, encompassing cancers of the kidney, bladder, and prostate, represent approximately 25 % of all cancer cases. Recent advances have enhanced our understanding of PVT1's crucial functions. Long noncoding RNAs influence both the onset and development of cancer, as well as epigenetic alterations. Recent findings have focused on PVT1's mechanism of action across several malignancies, particularly urologic cancers. Understanding the various functions of PVT1 linked to cancer is necessary for the development of cancer detection and treatment when PVT1 is dysregulated. Furthermore, recent advancements in genomic and epigenetic research have elucidated the complex regulatory networks that control PVT1 expression. Comprehending the intricate role of PVT1 Understanding the complex function of PVT1 in urologic cancers has substantial clinical implications. Here, we summarize some of the most recent findings about the carcinogenic effects of PVT1 signaling pathways and the possible treatment strategies for urological malignancies that target these pathways.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Ziyaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Yousefian Naeini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology,Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Salimian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Golnaz Enayat
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Sharifi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran,Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Weng B, Braaten M, Lehn J, Morrissey R, Asghar MS, Silberstein P, Abdul Jabbar AB, Mathews A, Tauseef A, Mirza M. Survival and treatment of stage IV renal cell carcinoma in academic vs non-academic medical centers. World J Nephrol 2025; 14:103923. [DOI: 10.5527/wjn.v14.i2.103923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/22/2025] [Accepted: 02/08/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is treated with surgical resection as the gold standard, as it is notoriously resistant to systemic therapy. Advancements with targeted therapies contribute to declining mortality, but metastatic RCC (mRCC) survival remains poor. One possible factor is treatment at academic centers, which employ advanced providers and novel therapies. This study compared outcomes of mRCC in patients treated at academic/research facilities compared to those treated at non-academic centers.
AIM To compare survival outcomes of mRCC and their various etiologies between academic and non-academic centers.
METHODS The National Cancer Database was used to identify mRCC patients including all histology subtypes and stage IV disease. Descriptive statistics and Kaplan-Meier curves measured survival outcomes for user file facility types sorted into a binary academic/research and non-academic research variable. Multivariate logistic regression and Cox proportional hazard testing generated odds ratio and hazard ratio. Data was analyzed using Statistical Package for the Social Sciences version 29.0 using a significance level of P < 0.05.
RESULTS Overall, academic facility patients experienced greater 5-year and 10-year overall survival than non-academic facility patients. Treatment at non-academic facilities was associated with increased odds of death that persisted even after controlling for age, tumor size, sex, and distance traveled to treatment center. In comparison, non-academic facility patients also experienced greater risk of hazard.
CONCLUSION Patients with mRCC treated at academic/research facilities experienced increased survival compared to patients treated at non-academic facilities, were more likely to be younger, carry private insurance, and come from a large metropolitan area. They also were significantly more likely to receive surgery and adjuvant immunotherapy.
Collapse
Affiliation(s)
- Bob Weng
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Marco Braaten
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Jenna Lehn
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Reid Morrissey
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Muhammad Sohaib Asghar
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Peter Silberstein
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Ali Bin Abdul Jabbar
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Abraham Mathews
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Abubakar Tauseef
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| | - Mohsin Mirza
- Department of Internal Medicine, Creighton University School of Medicine, Omaha, NE 68178, United States
| |
Collapse
|
9
|
Chatterjee S, Paul N, Das A, Bank S, Bankura B, Sarkar K, Saha S, Malakar S, Choudhury S, Ghosh S, Das M. Investigating the association of VHL gene variants with disease risk and clinicopathological outcomes in ccRCC patients from West Bengal, India. Urol Oncol 2025; 43:394.e9-394.e22. [PMID: 39809638 DOI: 10.1016/j.urolonc.2024.12.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a prevalent and aggressive malignancy, with the von Hippel-Lindau (VHL) gene playing a critical role in its pathogenesis. However, the association between VHL gene variants and sporadic ccRCC risk remains unexplored in the Indian population. This study aimed to investigate the somatic and germline variants of the VHL gene in sporadic ccRCC patients from West Bengal, India, and their association with disease risk and clinicopathological parameters. METHODS A total of 210 ccRCC patients and 255 ethnicity-matched healthy controls were enrolled. Genomic DNA from blood and tissue samples was analyzed using PCR-based Sanger sequencing. The association of VHL variants with ccRCC risk was assessed using Chi-square tests. The impact of genetic variants on patient clinicopathological features and overall survival was evaluated using Kaplan-Meier survival analysis and Cox proportional hazards models. RESULTS We identified twenty-three single nucleotide variants (SNVs) in the VHL gene, including 3 novel variants, OR250433 T > G, OR125589 C > T and OQ627404 G > C. The intronic variant rs61758376 G > C and 3'UTR variant rs1642742 A > G were significantly associated with an increased risk of ccRCC (OR = 1.676, P = 0.0074; OR = 1.735, P = 0.0171, respectively). The rs1642742 GG genotype was also significantly associated with larger tumor size (P < 0.05) and advanced tumor stage (pT4). Kaplan-Meier analysis indicated poorer overall survival for patients with the rs1642742 GG genotype (log-rank P = 0.029). CONCLUSION Our study is the first to document the association of VHL gene variants with sporadic ccRCC risk and clinical outcomes in the Indian population. The identified variants, particularly rs61758376 and rs1642742, could serve as potential biomarkers for ccRCC susceptibility and prognosis.
Collapse
Affiliation(s)
| | - Nirvika Paul
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Anwesha Das
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Sarbashri Bank
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Biswabandhu Bankura
- Multidisciplinary Research Unit, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Kunal Sarkar
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Soumen Saha
- Department of Urology, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Subhajit Malakar
- Department of Urology, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Sunirmal Choudhury
- Department of Urology, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Sudakshina Ghosh
- Department of Zoology, Vidyasagar College for Women, Kolkata, West Bengal, India
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| |
Collapse
|
10
|
Deng W, Wu L, Chen L, Wang K, Lin N, Zhu L, Chen J. Development of B7-H3 targeted CAR-T cells for renal cell carcinoma therapy: in vitro and in vivo efficacy. Clin Transl Oncol 2025; 27:2667-2678. [PMID: 39560834 DOI: 10.1007/s12094-024-03792-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: 08/21/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE This study aims to develop chimeric antigen receptor (CAR)-T cells specifically targeting B7-H3-expressing renal cell carcinoma (RCC) and to evaluate the feasibility of B7-H3 CAR-T therapy for RCC. METHODS We analyzed B7-H3 expression in RCC using bioinformatics approaches and confirmed it in tissues and cell lines through immunohistochemical staining and Western blot analysis. A lentiviral vector containing a B7-H3 specific CAR was constructed and transfected into human T cells, with CAR expression verified by flow cytometry. Cytotoxic efficacy was evaluated in co-culture experiments, measuring the production of interferon-gamma (IFN-γ), interleukin-2 (IL-2), granzyme B, and lactate dehydrogenase (LDH) release. Xenograft models in nude mice were used to evaluate tumor growth inhibition by B7-H3 CAR-T cells. RESULTS B7-H3 was significantly expressed in RCC and associated with poor prognosis. Elevated levels of B7-H3 expression were validated in both RCC tissues and cell lines. A B7-H3-specific CAR-T cell was developed, achieving a CAR transduction efficiency of 39.85%, as assessed by flow cytometry. In vitro co-culture assays demonstrated that the CAR-T cells exhibited substantial cytotoxic activity against RCC cell lines, with this activity positively correlating with the effector-to-target ratio. Furthermore, the secretion levels of IFN-γ, IL-2, granzyme B, and LDH were significantly increased compared to the control groups. In vivo experiments further confirmed that B7-H3 CAR-T cells significantly inhibited tumor growth. CONCLUSION The current study suggests that B7-H3 CAR-T cells exhibit significant efficacy in targeting and eliminating RCC cells, indicating a promising cellular immunotherapy approach for RCC treatment.
Collapse
Affiliation(s)
- Wenyi Deng
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Lvying Wu
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Liuyan Chen
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China
| | - Kuanyin Wang
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China
- Department of Urology, the Second Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Na Lin
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China
| | - Lingfeng Zhu
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China.
- Department of Urology, the Second Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China.
| | - Jin Chen
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, 368 Yehai Avenue, Haikou, 570311, Hainan, China.
- Minimally Invasive Urology and Translational Medicine Center, Fuzhou First General Hospital Affiliated With Fujian Medical University, 190th Dadao Road, Fuzhou, 350009, Fujian, China.
| |
Collapse
|
11
|
Zheng X, Liu Y, Yang Z, Tian Y. Metabolic reprogramming and immune microenvironment profiling in clear cell renal cell carcinoma: implications for prognosis, targeted therapy, and drug resistance. Discov Oncol 2025; 16:850. [PMID: 40397318 DOI: 10.1007/s12672-025-02401-w] [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: 01/26/2025] [Accepted: 04/15/2025] [Indexed: 05/22/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent form of kidney cancer, distinguished by intricate interactions between metabolic reprogramming, immune microenvironment dynamics, and genetic mutations. In this detailed investigation, we analyzed the ccRCC cohort from The Cancer Genome Atlas (TCGA) alongside 81 metabolic signaling pathways from the KEGG database. By utilizing Gene Set Variation Analysis (GSVA), we performed hierarchical clustering of patients based on their metabolic pathway activity profiles, identifying three distinct clusters with notable differences in pathway activity and survival outcomes. Cluster 1 displayed high metabolic activity and more favorable survival outcomes, while Cluster 3 was characterized by low metabolic activity and poorer prognosis. Clinical comparisons revealed significant disparities in gender, histological stage, and survival status, with Cluster 3 exhibiting a higher proportion of patients at advanced stages and those who had passed away. Genetically, Cluster 1 showed the highest mutation burden, with prominent mutations in genes such as VHL and PBRM1. Biological process analysis indicated that pathways like organic carboxylic acid metabolism and ATP synthesis were upregulated in Cluster 1 but suppressed in Cluster 3. Machine learning models (GBM, CoxBoost, and LASSO regression) enabled the identification of four pivotal genes-BCAT1, IL4I1, ACADM, and ACADSB-which were subsequently used to construct a multifactorial Cox regression model. This model successfully stratified patients into high- and low-risk groups, correlating with marked differences in immune activities. The high-risk group showed elevated expression of chemokines, TNF, and HLA molecules. Drug sensitivity analysis suggested that AKT inhibitor III was more effective in the low-risk cohort, while Bortezomib might be more beneficial for high-risk patients. Additionally, a clinical prediction model integrating risk scores and clinical factors demonstrated strong predictive power for patient survival. Methylation profiling of the core genes via the UALCAN platform revealed distinct epigenetic signatures in ccRCC, providing deeper insight into the disease's molecular mechanisms. This study contributes to a more comprehensive understanding of ccRCC and proposes valuable directions for personalized treatment strategies and enhanced patient management.
Collapse
Affiliation(s)
- Xiao Zheng
- Department of Nephrology, Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of China Three Gorges University, Yichang, 443002, Hubei, China
| | - Yongqiang Liu
- Department of Nephrology, Gezhouba Central Hospital of Sinopharm, The Third Clinical Medical College of China Three Gorges University, Yichang, 443002, Hubei, China
| | - Zixin Yang
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Yanhua Tian
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
12
|
Yue D, Zheng M. Proteomics and succinylation modification characterization in clear cell renal cell carcinoma. Discov Oncol 2025; 16:835. [PMID: 40394288 DOI: 10.1007/s12672-025-02737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most lethal form of kidney cancer, with a significant number of patients experiencing tumor progression. Succinylation modification is a novel post-translational modification (PTM) that refers to modifying a protein with a succinyl group, which most frequently happens to lysine residues. Recent studies have revealed that abnormal succinylation, altered protein activity, dysfunctional roles in energy metabolism, and subsequent epigenetic modifications are linked to the onset and progression of conditions like inflammation, cancer, and other diseases. No studies have offered a comprehensive analysis of succinylation modification in ccRCC or clarified the mechanisms by which this modification operates within disease progression. In this study, we applied quantitative proteomics and succinylation modification omics to extensively examine the global proteome and succinylation modification changes in ccRCC tissues. Using high-throughput liquid chromatography-mass spectrometry, we identified 4801 lysine succinylation modification sites across 1274 proteins in ccRCC and adjacent non-cancerous tissues. Additionally, 434 succinylation sites within 328 proteins displayed significant differential modification in ccRCC (fold change (FC) > 1.5 or p < 0.05). Notably, the succinylated proteins were primarily associated with energy metabolism pathways, including fatty acid elongation, glyoxylate and dicarboxylate metabolism, the tricarboxylic acid cycle, and oxidative phosphorylation, and were predominantly located within the mitochondria. This study is the first to present a global proteomic profile and a detailed succinylation modification landscape in ccRCC. These findings introduce new potential approaches for treating ccRCC by reversing abnormal succinylation modifications.
Collapse
Affiliation(s)
- Dong Yue
- Department of Urology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Miao Zheng
- Department of Operating Room, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
13
|
Wang K, Shen X, Wu J, Bi Q, Gao Z, Sun Z, Wang W. Fibrogenesis-driven tumor progression in clear cell renal cell carcinoma: prognostic, therapeutic implications and the dual role of neuropilin-1. Cancer Cell Int 2025; 25:179. [PMID: 40380175 PMCID: PMC12082889 DOI: 10.1186/s12935-025-03801-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/24/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the predominant subtype of renal cancer, with a poor prognosis driven by therapy resistance and a propensity for recurrence. Tumor microenvironment (TME)-associated fibrosis accelerates disease progression by fostering immune evasion. Neuropilin-1 (NRP1), a key mediator in fibrotic signaling and cancer biology, has been implicated in these processes. However, the genetic correlation between fibrogenesis and ccRCC remains largely unexplored, necessitating a focused analysis of fibrogenesis-related genes (FRGs) to identify novel prognostic markers and therapeutic strategies. METHODS This study utilized an integrative bioinformatics framework to identify prognosis-associated fibrogenesis-related genes (pFRGs) and applied non-negative matrix factorization (NMF) to stratify ccRCC patients based on fibrotic signatures. A machine learning-derived prognostic model was developed to categorize patients into high-risk and low-risk groups, with tumor microenvironment (TME) features analyzed across these subgroups. The pro-tumorigenic role of NRP1 via the TGF-β/SMAD signaling pathway was validated in vitro and in vivo. RESULTS Twelve pFRGs were identified, with elevated expression correlating with reduced survival. NMF revealed two ccRCC subtypes with different fibrotic and immune profiles. The high-fibrosis subtype showed worse survival and a pro-tumorigenic TME. The risk model demonstrated robust predictive performance (AUCs: 0.738, 0.731, 0.711 for 1-, 2-, and 3-year survival). High-risk patients, marked by immune dysfunction, exhibited worse survival but greater immunotherapy sensitivity. Among the pFRGs, NRP1 was upregulated in ccRCC, and paradoxically associated with favorable prognosis in TCGA, primarily due to stromal enrichment. In vitro and in vivo experiments confirmed that NRP1 promotes ccRCC proliferation, migration, and invasion by enhancing TGF-β/SMAD-driven epithelial-mesenchymal transition (EMT). CONCLUSION Fibrosis is a critical driver of ccRCC progression, linking fibrogenesis-related genes to poor prognosis, immune suppression, and tumor aggressiveness. NRP1 was identified as a central regulator of fibrosis-induced tumor progression through the TGF-β/SMAD signaling pathway. Combining NRP1 inhibition with anti-fibrotic therapies presents a potential strategy for enhancing therapeutic outcomes in ccRCC.
Collapse
Affiliation(s)
- Kai Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Urology, Linyi People's Hospital, Linyi, Shandong, China
| | - Xihao Shen
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiyue Wu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qing Bi
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zihao Gao
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zejia Sun
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Wei Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Chen R, Tang T, Han J, Li S, Liu W, Deng H, Jian T, Fu Z. Temporal trends of the disease burden of renal cell carcinoma from 1992 to 2019 in the US: a population-based analysis. Cancer Causes Control 2025:10.1007/s10552-025-02007-1. [PMID: 40358845 DOI: 10.1007/s10552-025-02007-1] [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: 03/16/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE Significant advances in the management, in particular the treatment, of renal cell carcinoma (RCC) has have been made over the years. However, it is not clear whether these advances reduce the disease burden of RCC at the population level. METHODS Using data from the Surveillance, Epidemiology, and End Results database, we estimated the temporal trends of RCC incidence, incidence-based mortality (IBM), and survival rates in the United States (US) from 1992 to 2019. RESULTS From 2008 to 2019, the incidence increased slowly at 1.1% annually (95% CI: 0.6% to 1.5%). The overall IBM rate of RCC increased by 6.8% per year (95% CI: - 1.1% to 15.3%) between 1994 and 1997, plateaued between 1997 and 2015, and then decreased nonsignificantly after 2015. During the study period, the overall Five year survival rate of RCC continuously increased from 53.69 in 1992 to 72.90% in 2014, with the best improvement observed for RCC patients with distant disease. However, we projected that, given the current trends, the incidence of RCC in the US will continue to increase from 6.92 per 100,000 in 2015-2019 to 9.59 per 100,000 in 2040-2044. CONCLUSION Over the years, the mortality of RCC has been decreased reducing at the US population level mainly because the considerably significantly improved survival of RCC patients at all stages through the advances in treatment. However, the overall incidence of RCC is continuously increasing, indicating that more effective preventive strategies should be developed to reduce the disease burden of RCC.
Collapse
Affiliation(s)
- Ruyan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tian Tang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jianglong Han
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Si Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenmin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiyu Deng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tingting Jian
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhenming Fu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
15
|
Liang H, Bai Y, Bao W, Wei L, Xie P. Solitary Radius Metastasis as the First Presentation of Renal Cell Carcinoma Revealed by Bone Scintigraphy. Clin Nucl Med 2025:00003072-990000000-01711. [PMID: 40357627 DOI: 10.1097/rlu.0000000000005951] [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: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025]
Abstract
Solitary radius metastasis as the initial presentation of renal cell carcinoma is extremely rare. Here we present the bone scintigraphy findings of a 72-year-old man who complained of left wrist pain for 6 months. The 99mTc-MDP bone scintigraphy demonstrated intense ring-like MDP uptake in the distal left radius. Finally, solitary radius metastasis from renal cell carcinoma was confirmed.
Collapse
Affiliation(s)
| | - Yanbin Bai
- Hand Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, PR China
| | | | | | | |
Collapse
|
16
|
Leng X, Zhou C, Wu J, Zheng H, Wang J, Li Q, Huang Y, Liu J. The relationship between renal cell carcinoma pathological types and perirenal fat area. BMC Cancer 2025; 25:841. [PMID: 40340924 PMCID: PMC12060561 DOI: 10.1186/s12885-025-14164-2] [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: 01/19/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
INTRODUCTION To explore whether there is a relationship between perirenal fat area (PFA) and the pathological types of renal cell carcinoma (RCC). METHODS Two hundred ninety-seven cases of RCC patients were included in our study, which is a retrospective analysis. Based on pathological type, we divided the 297 RCC patients into two groups: the clear cell renal cell carcinoma (ccRCC) group (236 cases) and the non-clear cell renal cell carcinoma (non-ccRCC) group (61 cases). Computed tomography (CT) images at the renal vein level were used to measure PFA. A multivariate logistic regression model was employed to examine the connection between various pathological types of RCC and PFA. RESULTS Significant differences were observed between ccRCC and non-ccRCC patients in PFA (P = 0.007), contralateral PFA (P = 0.011), weight (P = 0.002), BMI (P < 0.001), pathological stage 1 (P = 0.010), and pathological stage 2 (P = 0.002). To study the link between pathological subtypes and PFA, a multivariate logistic regression model was employed. Stratifying patients by tumor location in the kidney, the multivariate logistic regression analysis showed that when the tumor is located outside the polar lines of the kidney (OPLK), for every 1 cm2 increase in PFA, the probability of developing ccRCC increases by 5% [1.05 (1.01, 1.10) P = 0.0153]. Furthermore, after stratifying patients by tumor location and pathological stage, it was found that in T1 stage patients with tumors located OPLK, for every 1 cm2 increase in PFA, the probability of developing ccRCC increases by 6% [1.06 (1.01, 1.11) P = 0.0300]. CONCLUSION When the tumor is located OPLK in T1 stage patients, PFA is positively correlated with ccRCC. Perirenal adipose tissue may be a risk factor for ccRCC.
Collapse
Affiliation(s)
- Xin Leng
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jiulong Wu
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Hongfang Zheng
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Jianliang Wang
- Department of Radiology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Qiaoxing Li
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Jianhu Liu
- Department of Urology, The First People's Hospital of Kunshan, Suzhou, 215300, China.
| |
Collapse
|
17
|
Wang Y, Lu J, Lin B, Chen J, Lin F, Zheng Q, Xue X, Wei Y, Chen S, Xu N. Integrated analysis of MIOX gene in prognosis of clear-cell renal cell carcinoma. Cell Death Dis 2025; 16:368. [PMID: 40341358 PMCID: PMC12062366 DOI: 10.1038/s41419-025-07698-7] [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: 10/26/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is a highly aggressive malignancy that originates in the kidney. It often exhibits a limited response or can be refractory to a wide range of anti-cancer therapies, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors. Ferroptosis is a form of oxidative, iron-dependent cell death characterized by lipid peroxidation. Targeting ferroptosis may offer a promising alternative therapeutic strategy for cancer cells that are resistant to existing treatments. The impact of ferroptosis-related genes on the prognosis of ccRCC patients is still not fully understood. In this study, we identified 30 differentially expressed ferroptosis-related genes in ccRCC samples compared to normal tissues using data from The Cancer Genome Atlas (TCGA). Lasso regression analyses, along with Kaplan-Meier analysis, were conducted to identify genes associated with prognosis. Based on scRNA-seq and spatial transcriptome analysis, we identified specificity of MIOX in ccRCC. Furthermore, MIOX demonstrated the highest significance, highlighting its independent prognostic value as a single gene in ccRCC. Our findings suggest that MIOX could serve as potential targets for therapeutic interventions in ccRCC.
Collapse
Affiliation(s)
- Yiqiu Wang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayi Lu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bohan Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayin Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qingshui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xueyi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaohao Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
18
|
Mao J, Li W, Sun X, Fu B, He J, Yan C, Zhu J, Zhang Z, Mao J, Hong Z, Tang Q, Liu Z, Li P, Zhang Y, Wang R. RPF-Net: A multimodal model for the postoperative UISS risk stratification of non-metastatic ccRCC based on CT and whole-slide images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 268:108836. [PMID: 40381420 DOI: 10.1016/j.cmpb.2025.108836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND OBJECTIVES Postoperative non-metastatic clear cell renal cell carcinoma (nccRCC) patients face the risk of tumor recurrence and metastasis. However, prognosis assessment for nccRCC remains time-consuming and subjective. In the current diagnostic landscape, computed tomography (CT) images provide macro-scale anatomical information, and whole-slide images (WSIs) offer micro-scale details that are inaccessible to CT imaging. To address this gap, the study proposes a multimodal approach that leverages both CT and WSI data to develop an automated model for postoperative risk stratification in nccRCC. METHODS This study proposes a multimodal model named the Radiology-Pathology Fusion Network (RPF-Net), which employs self-attention, graph-attention, and dynamic attention fusion mechanisms to integrate CT images and WSIs for classifying nccRCC patients into low-risk and intermediate-high-risk groups per the University of California, Los Angeles, Integrated Staging System (UISS) criteria. The proposed model is divided into three steps. First, the ResNet-50 and 3D ResNet-50 are used as feature extractors to respectively extract representative feature maps from WSIs and CT images. Second, a dual-branch module is designed to extract global and local features of the WSIs. Finally, a multilayer dynamic attention fusion (MDAF) module is developed to facilitate cross-modal feature interaction and predict the risk stratification results. RESULTS The area under the curve (AUC), accuracy, precision, and F1 Score of the RPF-Net on the internal validation set are 0.949±0.013, 0.894±0.019, 0.895±0.020, and 0.894±0.019, respectively. Furthermore, the RPF-Net shows robust generalization, achieving an AUC of 0.901 on the external validation set and 0.924 on the public dataset. CONCLUSIONS The RPF-Net models the diagnostic process of multimodal data and shows strong generalization and excellent performance. This model may be a potential tool to facilitate clinical risk stratification and management for postoperative nccRCC patients.
Collapse
Affiliation(s)
- Jiahui Mao
- Medical College, Guizhou University, Guiyang, 550025, China; Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Wuchao Li
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Xinhuan Sun
- CETC Big Data Research Institute Co., Ltd., Guizhou 550022, China; National Engineering Research Center of Big Data Application to Improvement of Governance Capacity, Guizhou 550022, China
| | - Bangkang Fu
- Medical College, Guizhou University, Guiyang, 550025, China; Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Junjie He
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Chongzhe Yan
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Jianguo Zhu
- Department of Urinary, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Zhuxue Zhang
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Jiahui Mao
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, 200082, China
| | - Zhangxin Hong
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Qi Tang
- Medical College, Guizhou University, Guiyang, 550025, China
| | - Zhen Liu
- Medical College, Guizhou University, Guiyang, 550025, China
| | - Pinhao Li
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550002, China.
| | - Yan Zhang
- Department of Imaging, Affiliated Hospital of Guizhou Medical University, Guiyang, 550002, China.
| | - Rongpin Wang
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
19
|
Xiao Z, Wang Y, Pan D, Liu X, Gan J, Huang L, Feng Y. USP3 promotes clear cell renal cell carcinoma progression by stabilizing MYC and enhancing glycolysis. Biochim Biophys Acta Gen Subj 2025; 1869:130801. [PMID: 40164288 DOI: 10.1016/j.bbagen.2025.130801] [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: 12/17/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of renal malignancy, and the deubiquitinase USP3 has been implicated as a critical factor in tumor biology. However, the precise mechanisms by which USP3 contributes to ccRCC progression remain unclear. This study investigates the role of USP3 in ccRCC and elucidates its underlying molecular mechanisms. Data from TCGA and GTEx databases showed elevated USP3 expression in ccRCC tissues and cell lines compared to normal renal tissues. Further analysis using qPCR and Western blot confirmed this upregulation in ccRCC cell lines. Functional assays revealed that silencing USP3 significantly impaired cell proliferation, migration, and invasion, while promoting apoptosis. Additionally, co-immunoprecipitation assays demonstrated an interaction between USP3 and MYC, with subsequent ubiquitination assays showing that USP3 regulates MYC stability. USP3 depletion also led to alterations in glycolysis-related gene expression, which could be partially reversed by MYC overexpression. These findings suggest that USP3 modulates ccRCC progression by stabilizing MYC, highlighting its potential as a therapeutic target in ccRCC treatment.
Collapse
Affiliation(s)
- Zhiliang Xiao
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yuan Wang
- Guangzhou Medical University, Guangzhou 511436, China
| | - Dehua Pan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Xin Liu
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Jin Gan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Liang Huang
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yan Feng
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China.
| |
Collapse
|
20
|
Hu P, Chen D, Yu J, Mi H. Comprehensive Whole-Course Management Strategy for Recurrent Renal Cell Carcinoma: Case Report and Literature Review. Clin Case Rep 2025; 13:e70418. [PMID: 40321230 PMCID: PMC12045782 DOI: 10.1002/ccr3.70418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/27/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Renal cell carcinoma, a highly recurrent and metastatic malignancy. Drawing upon a decade-long expertise in managing recurrent cases, we advocate the adoption of Programmed Death-1 (PD-1) inhibitors in conjunction with Axitinib when monotherapy with Axitinib fails to restrain tumor metastasis, thereby empowering doctors with an efficacious treatment option.
Collapse
Affiliation(s)
- PengNan Hu
- Department of UrologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Di Chen
- Department of UrologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jun Yu
- Department of UrologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Hua Mi
- Department of UrologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
21
|
Alocha H, Rajab I, Khalid N, Sabah I, Dahmas AM, Mahajna R. Atypical presentation of renal cell carcinoma with lactic acidosis: case report and literature review. J Surg Case Rep 2025; 2025:rjaf290. [PMID: 40331018 PMCID: PMC12052241 DOI: 10.1093/jscr/rjaf290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/20/2025] [Indexed: 05/08/2025] Open
Abstract
Renal cell carcinoma is often difficult to diagnose early due to its nonspecific clinical presentation, which extends beyond the classic triad of flank pain, hematuria, and a flank mass. Recognizing alternative indicators, such as microscopic hematuria and lactic acidosis, can aid in early detection. A 77-year-old male with diabetes, varicose veins, and tobacco use presented with weakness, nausea, dyspnea, and poor appetite. He exhibited somnolence, confusion, transaminitis, elevated alkaline phosphatase, and severe lactic acidosis. Urinalysis revealed microscopic hematuria. Imaging showed bilateral pleural effusions and a right hepatic lesion. A computed tomography scan identified a large renal mass invading the renal vein, inferior vena cava, and right atrium. The patient developed deep vein thromboses and underwent radical nephrectomy, but succumbed postoperatively. This case highlights renal cell carcinoma's potential for atypical presentations, emphasizing the importance of early recognition and comprehensive diagnostic approaches to improve outcomes.
Collapse
Affiliation(s)
- Henry Alocha
- Department of Internal Medicine, St. Joseph’s University Medical Center, 703 Main Street, Paterson, Passaic County, NJ 07503, United States
| | - Islam Rajab
- Department of Internal Medicine, St. Joseph’s University Medical Center, 703 Main Street, Paterson, Passaic County, NJ 07503, United States
| | - Noman Khalid
- Department of Internal Medicine, St. Joseph’s University Medical Center, 703 Main Street, Paterson, Passaic County, NJ 07503, United States
| | - Ibraheem Sabah
- Department of Internal Medicine, St. Joseph’s University Medical Center, 703 Main Street, Paterson, Passaic County, NJ 07503, United States
| | - Abdelfattah M Dahmas
- Department of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus Street, New Campus, Nablus, West Bank 00970, Palestine
| | - Rawda Mahajna
- Department of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus Street, New Campus, Nablus, West Bank 00970, Palestine
| |
Collapse
|
22
|
Li X, Wu M, Chen G, Ma W, Chen Y, Ding Y, Dong P, Ding W, Zhang L, Yang L, Gan W, Li D. The Role of HADHB in Mitochondrial Fatty Acid Metabolism During Initiation of Metastasis in ccRCC. Mol Carcinog 2025; 64:923-935. [PMID: 39991877 DOI: 10.1002/mc.23898] [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: 09/11/2024] [Revised: 01/07/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
The initiation and progression of clear cell renal cell carcinoma (ccRCC) are closely linked to significant metabolic alterations. Specifically, lipid metabolism alterations and their association with the high invasiveness in ccRCC require further investigation. After conducting RNA-sequencing (RNA-seq), we discovered that Hydroxyacyl-CoA Dehydrogenase Trifunctional Multienzyme Complex Subunit Beta (HADHB) was significantly downregulated in the highly invasive ccRCC cell line. It was found that the expression of HADHB in ccRCC tumor tissues was lower than that in paracancer tissues, which is associated with poor patient prognosis. Subsequently, we confirmed that highly invasive ccRCC exhibited an increased lipid accumulation due to the suppression of mitochondrial fatty acid transport and enhanced conversion of fatty acids to triglycerides within cancer cells. Specifically, the downregulation of HADHB inhibited mitochondrial fatty acid β-oxidation (FAO) in cancer cells, leading to partial impairment of mitochondrial function and decreased ATP production. However, this trade-off involving the reduction of a high-yield ATP production conferred an advantage by reducing reactive oxygen species (ROS) generation within cancer cells, thereby protecting them from oxidative stress and enhancing their invasive potential. Furthermore, the downregulation of HADHB promoted epithelial-mesenchymal transition (EMT) and angiogenesis in cancer cells, accelerating the progression of ccRCC and endowing ccRCC cells with metastatic capabilities.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Mengmeng Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Guijuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Wenliang Ma
- Department of Urology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yi Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Yibing Ding
- Translational Medicine Core Facilities, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Ping Dong
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Weidong Ding
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Luqing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Lei Yang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Weidong Gan
- Department of Urology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-embryology, Medical School, Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Robledo XG, García-Perdomo HA. Neoadjuvant systemic therapy in managing locally advanced renal cancer before surgery A systematic review and meta-analysis. Can Urol Assoc J 2025; 19:E199-E204. [PMID: 40209186 DOI: 10.5489/cuaj.8901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
INTRODUCTION We aimed to estimate the effectiveness and safety of neoadjuvant systemic therapy in locally advanced renal tumor patients who undergo a nephrectomy in terms of survival, tumor response, and surgical feasibility. METHODS We included clinical trials, quasi-experimental studies, and cohort studies providing data on the use of neoadjuvant systemic therapy in managing locally advanced renal cancer before surgery in adult patients. The primary outcomes were cancer-specific survival (CSS), overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS). We performed a meta-analysis of proportions in R and assessed the risk of bias with the MINORS tool. RESULTS Nine studies were included for qualitative synthesis and seven for meta-analysis. All non-randomized studies assessed had a low risk of bias against the stated objective by clearly stating their purpose. Likewise, most studies had a low risk of bias in the consecutive inclusion of patients, contemporary groups, and equivalence. Change in tumor size ranged from -50% to +7.9%. Partial response was achieved in 26% (95% confidence interval 15-42). CONCLUSIONS Neoadjuvant therapy in locally advanced renal tumors ≥T3 or N1 has shown positive results regarding clinical tumor regression in about one-third of the patients. It is feasible and safe in this high-risk population.
Collapse
Affiliation(s)
- Ximena Guzman Robledo
- Division of Urology/Uro-oncology, Department of Surgery, School of Medicine, Universidad del Valle, Cali, Colombia
| | - Herney Andrés García-Perdomo
- Division of Urology/Uro-oncology, Department of Surgery, School of Medicine, Universidad del Valle, Cali, Colombia
| |
Collapse
|
24
|
Osorio L, Grazioso TP, de Velasco G, Etxaniz O, Pérez-Gracia JL, Pinto Á, Durán I, Grande E, Garcia PB, Lázaro M, Rodriguez L, Villalobos ML, García L, Cuellar A, Solís-Hernández MP, Pernaut C, Rodríguez-Moreno JF, Rodriguez-Antona C, García-Donas J. Retrospective study assessing the role of the androgen receptor in clear cell renal cell cancer patients treated with VEGFR inhibitors in monotherapy. Clin Transl Oncol 2025; 27:2241-2255. [PMID: 39365364 DOI: 10.1007/s12094-024-03652-9] [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: 06/14/2024] [Accepted: 07/27/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND PURPOSE Despite that incorporating antiangiogenic in combination with immune-checkpoint inhibitors as the standard first-line treatment for advanced clear cell renal cell cancer (ccRCC) yields promising outcomes, these regimens often lead to significant toxicity. However, a subgroup of patients has shown responsiveness to VEGFR tyrosine-kinase inhibitors (TKIs) in monotherapy, leading to the question of whether employing combination therapies can significantly enhance overall survival in all patients over monotherapy. Thus, we aim to identify gene expression signatures that can predict TKI response within subpopulations that might benefit from single-agent therapies, to minimize unnecessary exposure to combination therapies and their associated toxicities, as well as to discover new potential therapeutic targets to improve ccRCC treatment. Based on prior data, the androgen receptor (AR) might meet both conditions. PATIENTS AND METHODS We evaluated the association between AR expression, assessed through NanoString® technology-derived mRNA counts, and the clinical outcomes of 98 ccRCC patients treated with first-line antiangiogenics and determined its association with other genes implicated in ccRCC tumorigenesis. RESULTS Higher AR-expression correlates significantly with better prognosis and survival based on the MSKCC risk score, and longer PFS. Furthermore, we have identified a gene set signature associated with AR-overexpression and several genes involved in angiogenesis and transcriptional targets of the hypoxia-inducible factor, a cornerstone of ccRCC. CONCLUSIONS AR-overexpression and its association with other genes could favor a transcriptomic signature set to aid in identifying patients suitable for TKI in monotherapy, rather than aggressive combinations, enhancing thus, precision and personalized therapeutic decisions.
Collapse
Affiliation(s)
- Lucia Osorio
- Servicio de Urología, Urología Hospitalaria, Hospital HM La Rosaleda, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
| | - Tatiana P Grazioso
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain
| | | | - Olatz Etxaniz
- Grupo de Investigación Aplicada en Oncología de Badalona (B·ARGO), Hospital Germá Trials I Pujol, Barcelona, Spain
| | | | - Álvaro Pinto
- Medical Oncology Department, Hospital Universitario La Paz - IdiPAZ, Madrid, Spain
| | - Ignacio Durán
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | - Juan Francisco Rodríguez-Moreno
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain
| | - Cristina Rodriguez-Antona
- Pharmacogenomics and Tumor Biomarkers Group, Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM) CSIC/UAM, Madrid, Spain.
- Grupo de Cáncer Endocirno Hereditario, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, Spain.
| | - Jesús García-Donas
- Instituto de Investigación Sanitaria HM Hospitales (IISHM), Madrid, Spain.
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain.
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo CEU, Madrid, Spain.
| |
Collapse
|
25
|
Ding C, Ma L, Liang Y, Zhang Z, Wu Q, Lyu J, Su L. Gastrointestinal adverse events associated with Lenvatinib versus Lenvatinib plus Pembrolizumab: A pharmacovigilance study in FDA adverse event reporting system. Sci Rep 2025; 15:15047. [PMID: 40301541 PMCID: PMC12041505 DOI: 10.1038/s41598-025-99773-4] [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: 05/26/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
This study aimed to empirically analyze gastrointestinal adverse events associated with Lenvatinib monotherapy and its combination with Pembrolizumab using FDA FAERS data (January 2015-December 2023), focusing on risk profiles, temporal patterns, and influencing factors. Proportional disproportionality analysis (ROR, PRR, BCPNN, EBGM) evaluated drug-AE associations. Kaplan-Meier curves characterized temporal distributions, while Wilcoxon rank-sum test compared median time-to-onset between regimens. Univariate logistic regression identified independent risk factors. A total of 291 severe gastrointestinal AEs reports were included. The gastrointestinal system had the most positive AE signals in both treatment groups. Perforation events showed strong positive signals in both regimens, while haemorrhage and fistula events were unique positive signals in the lenvatinib monotherapy group. In contrast, colitis and pancreatitis positive signals were more common in the combination therapy group. Most gastrointestinal AEs in both groups occurred within the first month of treatment. The monotherapy group had a significantly shorter median onset time than the combination therapy group (27 days vs. 38 days, P = 0.003). Logistic regression indicated that female sex (OR = 0.195, P = 0.022) and low-dose medication (OR = 0.240, P = 0.049) were independent protective factors for gastrointestinal AEs in the monotherapy group. This first comprehensive comparison reveals distinct gastrointestinal toxicity profiles: monotherapy predisposes to acute bleeding/fistulas, while combination therapy increases delayed tumor-related complications. Intensive monitoring during the first treatment month and gender/dosage-adjusted prevention strategies are recommended. These findings provide evidence-based insights for optimizing safety management of targeted-immunotherapy combinations.
Collapse
Affiliation(s)
- Chufeng Ding
- Department of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Lin Ma
- Department of Pharmacy, School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pharmacy, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Yankun Liang
- Department of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Zhenpo Zhang
- Department of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Qimin Wu
- Department of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Ling Su
- Department of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
26
|
Oya M, Yasuoka S, Tokudome T, Minegishi T, Hamada M, Ozaki M, Maekawa S, Ito Y. Adverse events of hepatic function disorder in Japanese patients with radically unresectable or metastatic renal cell carcinoma treated with pembrolizumab plus axitinib: a post-marketing surveillance study. Int J Clin Oncol 2025:10.1007/s10147-025-02708-2. [PMID: 40299253 DOI: 10.1007/s10147-025-02708-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/18/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Post-marketing surveillance focusing on hepatic function disorder was requested owing to its higher incidence in the pembrolizumab plus axitinib group than in the sunitinib group in KEYNOTE-426. We aimed to evaluate the prevalence and risk factors of adverse events (AEs) of hepatic function disorder in patients with unresectable/metastatic renal cell carcinoma (RCC) treated with pembrolizumab plus axitinib in real-world clinical practice in Japan. METHODS Patients were observed for 9 months after starting treatment with pembrolizumab plus axitinib. RESULTS In total, 193 patients were included in the safety analysis set (median age, 70 years). Most patients did not have a history of hepatic function disorder before starting treatment (96.4%, 186/193). The median treatment period was 27.1 weeks. At the 9-month data cut-off, 62.2% (120/193) of patients discontinued treatment, the most common reason being any AE in 31.1% (60/193). The incidence of AEs of hepatic function disorder was 30.1% (58/193) for any grade and 15.0% (29/193) for grade ≥ 3. Most AEs of hepatic function disorder occurred within 3 months from starting treatment. AEs of hepatic function disorder were the reason for discontinuation of pembrolizumab in 9.3% (18/193) of patients; axitinib, 7.3% (14/193); and both pembrolizumab and axitinib, 5.2% (10/193). No background factors were identified as being associated with the occurrence of AEs of hepatic function disorder. CONCLUSION There were no new safety signals for AEs of hepatic function disorder, and the incidence was consistent with that reported in KEYNOTE-426, in Japanese patients with radically unresectable/metastatic RCC treated with pembrolizumab plus axitinib.
Collapse
Affiliation(s)
- Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Yasuoka
- Oncology Medical Affairs, MSD K.K., 1‑13‑12 Kudan‑kita, Chiyoda‑ku, Tokyo, 102‑8667, Japan
| | - Takuto Tokudome
- Oncology Medical Affairs, MSD K.K., 1‑13‑12 Kudan‑kita, Chiyoda‑ku, Tokyo, 102‑8667, Japan.
| | - Toshihiko Minegishi
- Oncology Medical Affairs, MSD K.K., 1‑13‑12 Kudan‑kita, Chiyoda‑ku, Tokyo, 102‑8667, Japan
| | | | | | | | - Yuichiro Ito
- Oncology Medical Affairs, MSD K.K., 1‑13‑12 Kudan‑kita, Chiyoda‑ku, Tokyo, 102‑8667, Japan
| |
Collapse
|
27
|
Song H, Wang G, Gao G, Xia H, Jiao L, Wu K. A Systematic Analysis of Expression and Function of RAS GTPase-Activating Proteins (RASGAPs) in Urological Cancers: A Mini-Review. Cancers (Basel) 2025; 17:1485. [PMID: 40361412 PMCID: PMC12071082 DOI: 10.3390/cancers17091485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
The RAS signaling pathway is one of the most commonly dysregulated pathways in urological cancers. This pathway can be regulated by RASGAPs, which catalyze the hydrolysis of RAS-GTP to RAS-GDP. As such, the loss of RASGAPs can promote the activation of the RAS signaling pathway. Dysregulation of RASGAPs significantly contributes to the progression of urological cancers, including prostate cancer, bladder cancer, and renal cell carcinoma. Furthermore, alterations in RASGAP expression may influence sensitivity to chemotherapy, radiotherapy, and targeted therapies, suggesting their potential as therapeutic targets. Despite the challenges involved, a deeper understanding of the complexity of the RAS signaling network, along with the evolution of personalized medicine, holds promise for delivering more precise and effective treatment options targeting RASGAPs in urological cancers.
Collapse
Affiliation(s)
- Hao Song
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Guojing Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Guoqiang Gao
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Huayu Xia
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Lianying Jiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China;
| | - Kaijie Wu
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| |
Collapse
|
28
|
Zhou J, Xu Z, Yu Y, Zhu B, Xing Q. FOXM1 could serve as a bridge mediating prognosis and immunity for clear cell renal cell carcinoma via single-cell and bulk RNA-sequencing. Discov Oncol 2025; 16:626. [PMID: 40293585 PMCID: PMC12037465 DOI: 10.1007/s12672-025-02438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND In the development of several cancers, the Forkhead Box M1 (FOXM1) is crucial. The relationship between the immune system and FOXM1 in renal cell carcinoma (ccRCC), which has been verified by bulk RNA sequencing and scRNA sequencing, is the primary subject of this research. METHOD Publicly available data related to FOXM1 and ccRCC were extracted from The Cancer Genome Atlas (TCGA) database. The impact of FOXM1 on the prognosis of ccRCC was examined using Cox regression analysis. Results were verified by immunohistochemistry and quantitative real-time PCR (qRT-PCR). Additionally, single-cell sequencing data were analyzed. RESULTS When compared to para-carcinoma tissues, the expression of FOXM1 was considerably higher in ccRCC tissues. Patients with elevated FOXM1 expression had lower survival rates. FOXM1 may be a standalone prognostic factor for ccRCC, according to results of univariate and multivariate Cox regression studies. Reduced FOXM1 expression was linked to higher immunotherapy sensitivity, according to immunocorrelation analysis. This suggests FOXM1 may mediate immunotherapy resistance in ccRCC. Additionally, FOXM1 showed strong associations with tumor mutation load, microsatellite instability, and antitumor immunity. These results imply FOXM1 may regulate antitumor immunity in the ccRCC microenvironment. Consistent results from immunohistochemistry, PCR, and single-cell RNA sequencing confirmed upregulated FOXM1 expression in ccRCC. CONCLUSIONS According to the findings, FOXM1 might be used as a stand-alone prognostic biomarker for ccRCC. Moreover, FOXM1 has exhibited robust correlations with microsatellite instability, tumor mutation burden, immune response, and immunotherapy efficacy. FOXM1 may promote ccRCC pathogenesis partly by suppressing antitumor immunity and mediating immunotherapy resistance.
Collapse
Affiliation(s)
- Jianhua Zhou
- Department of Urology, Affiliated Hospital of Nantong University, No. 20 West Temple Road, Nantong, 226001, Jiangsu, China
| | - Zhuxian Xu
- Department of Urology, Affiliated Hospital of Nantong University, No. 20 West Temple Road, Nantong, 226001, Jiangsu, China
| | - Yang Yu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Bingye Zhu
- Department of Urology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), No. 881 Yonghe Road, Nantong, 226001, Jiangsu, China.
| | - Qianwei Xing
- Department of Urology, Affiliated Hospital of Nantong University, No. 20 West Temple Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
29
|
Netti GS, De Luca F, Camporeale V, Khalid J, Leccese G, Troise D, Sanguedolce F, Stallone G, Ranieri E. Liquid Biopsy as a New Tool for Diagnosis and Monitoring in Renal Cell Carcinoma. Cancers (Basel) 2025; 17:1442. [PMID: 40361369 PMCID: PMC12070982 DOI: 10.3390/cancers17091442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Renal cell carcinoma (RCC) presents a significant diagnostic challenge, particularly in small renal masses. The search for non-invasive screening methods and biomarkers has directed research toward liquid biopsy, which focuses on microRNAs (miRNAs), exosomes, and circulating tumor cells (CTCs). miRNAs are small non-coding RNA molecules that show considerable dysregulation in RCC, and they have potential for both diagnostic and prognostic applications. Research has highlighted their utility on biofluids, such as plasma, serum, and urine, in detecting RCC and characterizing its subtypes. Promising miRNA signatures have been associated with overall survival, suggesting their potential importance in the management of RCC. Exosomes, which carry a variety of molecular components, including miRNAs, are emerging as valuable biomarkers, whereas CTCs, released from primary tumors into the bloodstream, provide critical information on cancer progression. However, translation of these findings into clinical practice requires additional validation and standardization through large-scale studies and robust evidence. Although there are currently no approved diagnostic tests for RCC, the future potential of liquid biopsy in monitoring, treatment decision-making, and outcome prediction in patients with this disease is significant. This review examined and discussed recent developments in liquid biopsy for RCC, assessing both the strengths and limitations of these approaches for managing this disease.
Collapse
Affiliation(s)
- Giuseppe Stefano Netti
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Federica De Luca
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Valentina Camporeale
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Javeria Khalid
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Giorgia Leccese
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Dario Troise
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Unit of Nephrology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Francesca Sanguedolce
- Unit of Pathology, Department of Clinical and Experimental Medicine, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Giovanni Stallone
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Unit of Nephrology, Dialysis and Transplantation, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology, Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
- Center for Research and Innovation in Medicine (CREATE), Department of Medical and Surgical Sciences, University of Foggia–University Hospital “Policlinico Riuniti”, Viale Luigi Pinto, 71122 Foggia, Italy
| |
Collapse
|
30
|
Li L, Bai J, Wen X, Zeng X. Adverse reactions of four multi-targeted tyrosine kinase inhibitors: a descriptive analysis of the WHO-VigiAccess database. Front Pharmacol 2025; 16:1585862. [PMID: 40331199 PMCID: PMC12052882 DOI: 10.3389/fphar.2025.1585862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Background The introduction of multi-targeted tyrosine kinase inhibitors (MTKIs) such as axitinib, lenvatinib, sorafenib, and sunitinib has greatly broadened the available treatment options for Renal Cell Carcinoma (RCC). The study aims to compare the nature of the adverse reactions associated with these four MTKIs to identify which medication poses the least risk for personalized patient management, thus enabling more accurate clinical drug oversight. Methods Employing a retrospective descriptive analysis methodology, this research concentrated on four commercially available MTKIs. Reports pertaining to these medications were sourced from the WHO-VigiAccess database. The data gathering process involved collecting comprehensive information on various parameters, such as age demographics, gender, and the geographical distribution of patients associated with the ADR reports. Furthermore, the study explored disease systems and symptoms that were documented alongside the adverse reactions, as outlined in the annual ADR reports produced by the WHO. To assess the relationship between these four MTKIs and the linked AEs, both the Proportional Reporting Ratio (PRR) and the Reported Odds Ratio (ROR) were utilized. Results At the time of the search, a total of 123,818 AEs associated with the four MTKIs had been documented in the VigiAccess database. The common ADRs for these four MTKIs include diarrhoea, fatigue, death, hypertension, nausea, asthenia, weight decreased, and vomiting. Gastrointestinal disorders and general disorders and administration site conditions emerged as the SOCs with the highest number of adverse signals, both ranking first in terms of frequency. The elevated ROR (1.08) and PRR (1.06) values associated with gastrointestinal disorders in patients treated with sorafenib suggest a higher incidence of such adverse events compared to those observed with axitinib, lenvatinib, and sunitinib. Conclusion Recent comparative observational research suggests that the ADR reports submitted to the WHO and the FDA for these medications highlight both common and specific ADRs. It is essential for clinical practitioners to develop personalized treatment strategies that consider the adverse effects linked to different medications, alongside the unique circumstances of their patients, thus encouraging the responsible use of these MTKIs.
Collapse
Affiliation(s)
- Lijun Li
- Department of Pharmacy, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Clinical Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, the Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Jiayu Bai
- Department of rehabilitation medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xuelong Wen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xuefan Zeng
- Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Zheng Y, Zhang TN, Hao PH, Yang N, Du Y. Histone deacetylases and their inhibitors in kidney diseases. Mol Ther 2025:S1525-0016(25)00300-4. [PMID: 40263937 DOI: 10.1016/j.ymthe.2025.04.026] [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: 01/27/2025] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Histone deacetylases (HDACs) have emerged as key regulators in the pathogenesis of various kidney diseases. This review explores recent advancements in HDAC research, focusing on their role in kidney development and their critical involvement in the progression of chronic kidney disease (CKD), acute kidney injury (AKI), autosomal dominant polycystic kidney disease (ADPKD), and diabetic kidney disease (DKD). It also discusses the therapeutic potential of HDAC inhibitors in treating these conditions. Various HDAC inhibitors have shown promise by targeting specific HDAC isoforms and modulating a range of biological pathways. Their protective effects include modulation of apoptosis, autophagy, inflammation, and fibrosis, underscoring their broad therapeutic potential for kidney diseases. However, further research is essential to improve the selectivity of HDAC inhibitors, minimize toxicity, overcome drug resistance, and enhance their pharmacokinetic properties. This review offers insights to guide future research and prevention strategies for kidney disease management.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Peng-Hui Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yue Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
32
|
Szyk P, Czarczynska-Goslinska B, Ziegler-Borowska M, Larrosa I, Goslinski T. Sorafenib-Drug Delivery Strategies in Primary Liver Cancer. J Funct Biomater 2025; 16:148. [PMID: 40278256 PMCID: PMC12027913 DOI: 10.3390/jfb16040148] [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: 03/02/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Current primary liver cancer therapies, including sorafenib and transarterial chemoembolization, face significant limitations due to chemoresistance caused by impaired drug uptake, altered metabolism, and other genetic modulations. These challenges contribute to relapse rates of 50-80% within five years. The need for improved treatment strategies (adjuvant therapy, unsatisfactory enhanced permeability and retention (EPR) effect) has driven research into advanced drug delivery systems, including targeted nanoparticles, biomaterials, and combinatory approaches. Therefore, this review evaluates recent advancements in primary liver cancer pharmacotherapy, focusing on the potential of drug delivery systems for sorafenib and its derivatives. Approaches such as leveraging Kupffer cells for tumor migration or utilizing smaller NPs for inter-/intracellular delivery, address EPR limitations. Biomaterials and targeted therapies focusing on targeting have demonstrated effectiveness in increasing tumor-specific delivery, but clinical evidence remains limited. Combination therapies have emerged as an interesting solution to overcoming chemoresistance or to broadening therapeutic functionality. Biomimetic delivery systems, employing blood cells or exosomes, provide methods for targeting tumors, preventing metastasis, and strengthening immune responses. However, significant differences between preclinical models and human physiology remain a barrier to translating these findings into clinical success. Future research must focus on the development of adjuvant therapy and refining drug delivery systems to overcome the limitations of tumor heterogeneity and low drug accumulation.
Collapse
Affiliation(s)
- Piotr Szyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Beata Czarczynska-Goslinska
- Chair and Department of Pharmaceutical Technology, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| | - Marta Ziegler-Borowska
- Department of Biomedical Chemistry and Polymer Science, Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland;
| | - Igor Larrosa
- Department of Chemistry, University of Manchester, Chemistry Building, Oxford Road, Manchester M13 9PL, UK;
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
33
|
Guo W, Wang S, Yang Z, Dong Y, Xia Z, Xue W, Zhang C. SAP30 promotes clear cell renal cell carcinoma proliferation and inhibits apoptosis through the MT1G axis. Eur J Med Res 2025; 30:306. [PMID: 40247376 PMCID: PMC12007153 DOI: 10.1186/s40001-025-02440-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Sin3A-associated protein 30 (SAP30) is a crucial component of the SIN/HDAC histone deacetylase complex and acts as a scaffold that facilitates target gene binding. SAP30 is highly expressed in various tumours; however, its role in renal cell carcinoma (RCC) remains unclear. In our study, we observed the upregulation of SAP30 in clear cell renal cell carcinoma (ccRCC) tissues, and its elevated expression was correlated with a poor prognosis. Previous research has suggested that SAP30 may influence the growth, proliferation, and apoptosis of RCC cells. Gene Ontology (GO) analysis of the downstream regulatory targets of SAP30 revealed that SAP30 suppressed the expression of MT1G, a protein that binds to p53. Mechanistically, SAP30 inhibited MT1G transcription, thereby impairing the function of MT1G in delivering zinc ions to p53, which diminished p53 activity. Moreover, reduced MT1G levels attenuated the inhibitory effect of MT1G on MDM2, further destabilizing p53. Consequently, this cascade promoted RCC progression. In conclusion, our findings indicate that SAP30 inhibits the p53 pathway through MT1G suppression, suggesting that SAP30 and MT1G are potential prognostic markers and therapeutic targets for RCC.
Collapse
Affiliation(s)
- Wei Guo
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Shuwen Wang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zitong Yang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yu Dong
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Zhinan Xia
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wei Xue
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Cheng Zhang
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
34
|
Huang R, Kee L, Gont A, Meens J, Ferens FG, Irwin MS, Ailles L, Yuzwa SA, Robinson CM, Ohh M. Comparative single-cell transcriptomic profiling of patient-derived renal carcinoma cells in cellular and animal models of kidney cancer. FEBS Open Bio 2025. [PMID: 40241258 DOI: 10.1002/2211-5463.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer that often displays resistance to conventional cancer therapies, including chemotherapy and radiation therapy. Targeted treatments, including immunotherapies and small molecular inhibitors, have been associated with improved outcomes. However, variations in the patient response and the development of resistance suggest that more models that better recapitulate the pathogenesis and metastatic mechanisms of ccRCC are required to improve our understanding and disease management. Here, we examined the transcriptional landscapes of in vitro cell culture as well as in vivo orthotopic and metastatic NOD/SCID-γ mouse models of ccRCC using a single patient-derived RCC243 cell line to allow unambiguous comparison between models. In our mouse model assays, RCC243 cells formed metastatic tumors, and all tumors retained clear cell morphology irrespective of model type. Notably, gene expression profiles differed markedly between the RCC243 tumor models-cell culture, orthotopic tumors, and metastatic tumors-suggesting an impact of the experimental model system and whether the tumor was orthotopic or metastatic. Furthermore, we found conserved prognostic markers between RCC243 tumor models and human ccRCC patient datasets, and genes upregulated in metastatic RCC243 were associated with worse patient outcomes.
Collapse
Affiliation(s)
- Richard Huang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Lynn Kee
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Alexander Gont
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Fraser G Ferens
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Meredith S Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Claire M Robinson
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Michael Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Department of Biochemistry, University of Toronto, Canada
| |
Collapse
|
35
|
Chatterjee S, Paul N, Das A, Bank S, Bankura B, Yadav RP, Sarkar K, Saha S, Malakar S, Choudhury S, Ghosh S, Das M. Molecular dynamics reveal potential effects of novel VHL variants on VHL-Elongin C binding in ccRCC patients from Eastern India. Sci Rep 2025; 15:13022. [PMID: 40234555 PMCID: PMC12000512 DOI: 10.1038/s41598-025-95875-1] [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] [Received: 06/05/2024] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
Renal cell carcinoma (RCC) is the one of the most fatal and frequent form of urological malignancy worldwide. The von Hippel-Lindau (VHL) tumour suppressor gene is a critical component of the VHL-Cullin2-ElonginB/C (VCB) complex that regulates the ubiquitin-mediated proteasomal degradation of proteins with mutations consistently associated with the development of clear cell renal cell carcinoma (ccRCC). Despite extensive investigations conducted worldwide, there is a notable lack of data concerning VHL mutations in sporadic ccRCC patients from India. Our study aimed to investigate the sporadic VHL mutations within the tumours of 210 ccRCC patients without a familial history of VHL disease. We extracted genomic DNA from tumour and adjacent normal tissues, PCR amplified and sequenced the VHL gene. In silico tools were used assess the damaging potential of missense variants on pVHL structure and stability. Protein-protein docking and protein flexibility molecular docking simulation study were employed to study the interaction between wild-type and mutated VHL models with Elongin C. Sequence analysis revealed seven novel missense mutations in patient tumour tissues p.(Val170Phe), p.(Arg69Cys), p.(Phe76Leu), p.(Glu173Asp), p.(Leu201Val), p.(His208Leu), p.(Arg205Pro). I-Mutant 2.0 indicated these mutations reduced pVHL stability (ΔΔG < -0.5 kcal/mol). Protein Flexibility-Molecular Dynamic (MD) Simulation study indicated that mutations weaken the interaction of VHL with Elongin C, with V170F showing the most significant reduction in binding quality and stability. In conclusion, this study introduces novel genetic data from an understudied population and highlights the impact of VHL mutations on its interaction with Elongin C. These findings contribute to our understanding of the molecular basis of VHL-related pathologies and may guide future therapeutic strategies targeting these interactions.
Collapse
Affiliation(s)
- Srilagna Chatterjee
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Nirvika Paul
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Anwesha Das
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Sarbashri Bank
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Biswabandhu Bankura
- Multidisciplinary Research Unit, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Ravi Prakash Yadav
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Kunal Sarkar
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India
| | - Soumen Saha
- Department of Urology, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Subhajit Malakar
- Department of Urology, Institute of Postgraduate Medical Education and Research, Kolkata, West Bengal, India
| | - Sunirmal Choudhury
- Department of Urology, Calcutta Medical College and Hospital, Kolkata, West Bengal, India
| | - Sudakshina Ghosh
- Department of Zoology, Vidyasagar College for Women, 39 Sankar Ghosh Lane, Kolkata, 700006, India
| | - Madhusudan Das
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
36
|
Karakulak T, Zajac N, Bolck HA, Bratus-Neuenschwander A, Zhang Q, Qi W, Basu D, Oltra TC, Rehrauer H, von Mering C, Moch H, Kahraman A. Heterogeneous and novel transcript expression in single cells of patient-derived clear cell renal cell carcinoma organoids. Genome Res 2025; 35:698-711. [PMID: 40107723 PMCID: PMC12047245 DOI: 10.1101/gr.279345.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Splicing is often dysregulated in cancer, leading to alterations in the expression of canonical and alternatively spliced isoforms. We used the multiplexed arrays sequencing (MAS-seq) protocol of PacBio to sequence full-length transcripts in patient-derived organoid (PDO) cells of clear cell renal cell carcinoma (ccRCC). The sequencing revealed a heterogeneous dysregulation of splicing across 2599 single ccRCC cells. The majority of novel transcripts could be removed with stringent filtering criteria. The remaining 31,531 transcripts (36.6% of the 86,182 detected transcripts on average) were previously uncharacterized. In contrast to known transcripts, many of the novel transcripts have cell-specific expression. Novel transcripts common to ccRCC cells belong to genes involved in ccRCC-related pathways, such as hypoxia and oxidative phosphorylation. A novel transcript of the ccRCC-related gene nicotinamide N-methyltransferase is validated using PCR. Moreover, >50% of novel transcripts possess a predicted complete protein-coding open reading frame. An analysis of the most dominant transcript-switching events between ccRCC and non-ccRCC cells shows many switching events that are cell- and sample-specific, underscoring the heterogeneity of alternative splicing events in ccRCC. Overall, our study elucidates the intricate transcriptomic architecture of ccRCC, underlying its aggressive phenotype and providing insights into its molecular complexity.
Collapse
Affiliation(s)
- Tülay Karakulak
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Natalia Zajac
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Functional Genomics Center Zurich, ETH, 8057 Zurich, Switzerland
| | - Hella Anna Bolck
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
- Centre for AI, School of Engineering, Zurich University of Applied Sciences (ZHAW), 8400 Winterthur, Switzerland
| | | | - Qin Zhang
- Functional Genomics Center Zurich, ETH, 8057 Zurich, Switzerland
| | - Weihong Qi
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Functional Genomics Center Zurich, ETH, 8057 Zurich, Switzerland
| | - Debleena Basu
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | | | - Hubert Rehrauer
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Functional Genomics Center Zurich, ETH, 8057 Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, 8091 Zurich, Switzerland
| | - Abdullah Kahraman
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland;
- School for Life Sciences, Institute for Chemistry and Bioanalytics, University of Applied Sciences Northwestern Switzerland, 4132 Muttenz, Switzerland
| |
Collapse
|
37
|
Oki R, Takemura K, Urasaki T, Fujiwara R, Numao N, Yonese J, Miura Y, Yuasa T. Prevailing challenges in personalized treatment for metastatic renal cell carcinoma: a narrative review. Expert Rev Anticancer Ther 2025:1-13. [PMID: 40210604 DOI: 10.1080/14737140.2025.2491647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
INTRODUCTION The management of metastatic renal cell carcinoma (mRCC) has advanced with recent therapies, yet optimizing treatment remains challenging due to disease heterogeneity and the growing number of options. Integrating systemic and local treatments requires a multidisciplinary approach to improve outcomes. AREA COVERED This review summarizes recent developments in treatment for mRCC. Upfront immuno-oncology (IO)-based combinations have improved survival, though concerns about overtreatment and toxicity persist. While the role of cytoreductive nephrectomy (CN) has declined to some extent, it may still benefit well-selected patients. Metastasis-directed therapies, including metastasectomy and stereotactic radiotherapy, provide prognostic value, particularly for oligometastatic lesions or brain metastases. Comprehensive genomic profiling (CGP) holds promise for personalized treatment but is currently limited by the lack of actionable mutations and predictive biomarkers. EXPERT OPINION A personalized, multimodal approach is essential for optimizing mRCC management. Careful patient selection is key to balancing the benefits of treatment with the risks of toxicity. While CN and metastasis-directed therapies remain useful in select cases, advancing individualized care requires the development of validated biomarkers and broader application of CGP.
Collapse
Affiliation(s)
- Ryosuke Oki
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kosuke Takemura
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuya Urasaki
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Fujiwara
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noboru Numao
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuji Miura
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Yuasa
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
38
|
Ge Q, Meng J, Wang Z, Anwaier A, Lu J, Tian X, Wang Y, Yang J, Zhang H, Ye D, Xu W. Spatially segregated APOE + macrophages restrict immunotherapy efficacy in clear cell renal cell carcinoma. Theranostics 2025; 15:5312-5336. [PMID: 40303328 PMCID: PMC12036886 DOI: 10.7150/thno.109097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Immunotherapy has revolutionized cancer treatment and holds great potential for them, including metastatic clear cell renal cell carcinoma (ccRCC). However, immune resistance remains a major obstacle, limiting its efficacy and durability. Understanding the mechanisms of immune tolerance in the tumor microenvironment (TME) is pivotal for overcoming these challenges and enhancing therapeutic outcomes. Methods: Over 2000 samples, including a real-world cohort of 230 advanced ccRCC patients treated with immune checkpoint blockade (ICB) were analyzed. Single-cell RNA sequencing data from 13 tumor regions were categorized into ICB-exposed, ICB-resistant, and ICB-responsive groups. Multiple robust algorithms and multiplex immunofluorescence were used to explore TME composition and macrophage heterogeneity. Spatial communication dynamics were further investigated. In vitro experiments were performed to evaluate the impact of SPP1 on 786-O and 769-P cells. Co-culture experiments with THP-1-derived macrophages, followed by Western blot, flow cytometry, and functional assays, were performed to investigate SPP1-mediated macrophage polarization and its impact on tumor progression. Results: The results revealed an elevated presence of Apolipoprotein E (APOE)+ macrophages in ICB-resistant ccRCC. Notably, higher APOE+ macrophage proportion indicated shorter prognosis and worse response to ICB (P < 0.001). Elevated expression of CCAAT Enhancer Binding Protein Delta (CEBPD) was markedly linked to several immunosuppressive pathways, hindering T cell recruitment, promoting exhaustion, ultimately diminishing poorer prognosis and worse ICB efficacy. Meanwhile, upregulated Secreted Phosphoprotein 1 (SPP1) significantly enhances the proliferation, clonal formation, and migration of ccRCC cells. Tumor-derived SPP1. Additionally, SPP1 signaling from malignant cells appeared to recruit APOE+ macrophages to tumor margins, and promotes macrophage polarization into APOE+ M2-like macrophages. In the vicinity of the tumor, these APOE+ macrophages shape immunosuppressive TME by releasing abundant TGF-β signals, limiting anti-tumor effector T cells activity in ICB-resistant tumors, and contributing to tumor progression. Conclusion: This study reveals the critical role of APOE+ macrophages in promoting immune suppression and resistance to ICB therapy in ccRCC. By promoting T cell exhaustion and immunosuppressive signaling, particularly via localized TGF-β, these spatially segregated macrophages undermine treatment efficacy. Targeting APOE+ macrophages, especially in conjunction with ICB, presents a promising strategy to overcome immune resistance and enhance outcomes for ccRCC patients.
Collapse
Affiliation(s)
- Qintao Ge
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, 230022, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, 230022, P.R. China
| | - Zhongyuan Wang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jiahe Lu
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Jianfeng Yang
- Department of Urology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, P.R. China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center; Center; Department of Oncology, Shanghai Medical College; Qingdao Institute of Life Sciences, Fudan University, Shanghai, 200032, P.R. China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, P.R. China
| |
Collapse
|
39
|
Lv X, Liu J, Islam K, Ruan J, He C, Chen P, Huang C, Wang H, Dhar A, Moness M, Shi D, Murphy S, Zhao X, Yang S, Montoute I, Polakkattil A, Chung A, Ruiz E, Carbajal B, Padavala A, Chen L, Hua G, Chen X, Davis JS, Wang C. Hyperactivated YAP1 is essential for sustainable progression of renal clear cell carcinoma. Oncogene 2025:10.1038/s41388-025-03354-8. [PMID: 40210757 DOI: 10.1038/s41388-025-03354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025]
Abstract
The most notable progress in renal clear cell carcinoma (ccRCC) in the past decades is the introduction of drugs targeting the VHL-HIF signaling pathway-associated angiogenesis. However, mechanisms underlying the development of VHL mutation-independent ccRCC are unclear. Here we provide evidence that the disrupted Hippo-YAP signaling contributes to the development of ccRCC independent of VHL alteration. We found that YAP1 and its primary target genes are frequently upregulated in ccRCC and the upregulation of these genes is associated with unfavorable patient outcomes. Research results derived from our in vitro and in vivo experimental models demonstrated that, under normoxic conditions, hyperactivated YAP1 drives the expression of FGFs to stimulate the proliferation of tumor and tumor-associated endothelial cells in an autocrine/paracrine manner. When rapidly growing cancer cells create a hypoxic environment, hyperactivated YAP1 in cancer cells induces the production of VEGF, which promotes the angiogenesis of tumor-associated endothelial cells, leading to improved tumor microenvironment and continuous tumor growth. Our study indicates that hyperactivated YAP1 is essential for maintaining ccRCC progression, and targeting the dual role of hyperactivated YAP1 represents a novel strategy to improve renal carcinoma therapy.
Collapse
Affiliation(s)
- Xiangmin Lv
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiyuan Liu
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kazi Islam
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinpeng Ruan
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunbo He
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peichao Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cong Huang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anjali Dhar
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Dartmouth College, Hanover, NH, USA
| | - Madelyn Moness
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Davie Shi
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Savannah Murphy
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingeng Zhao
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Siyi Yang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Montoute
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Aneeta Polakkattil
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andie Chung
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Ruiz
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Brianna Carbajal
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Stem cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Alekhya Padavala
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guohua Hua
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Binzaqr S, Kryza D, Giraudet AL, Bernhard JC, Gross-Goupil M, Yacoub M, Margue G, Hindié E, Morgat C. Prostate-specific membrane antigen (PSMA) expression in primary and metastatic renal cell cancer (UroCCR-65 study). EJNMMI Res 2025; 15:38. [PMID: 40205264 PMCID: PMC11981970 DOI: 10.1186/s13550-025-01232-8] [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: 11/08/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) has been shown to be overexpressed in the neo-vasculature of renal cancers. However, studies investigating the pattern of PSMA expression in primary RCC and RCC metastases according to metastatic sites are rare. 44 frozen samples of RCC, 19 primaries (9 clear cell (cc) RCC, 7 papillary (pap) RCC, and 3 chromophobe (ch) RCC) and 25 (24 samples have ccRCC histology and one is unclassified) unpaired metastases (8 from adrenals, 8 from bones, 2 from lungs, 2 from liver and 5 others (1 lymph node, 1 pancreas, 1 brain, 1 gallbladder and 1 muscle)), were available from the UroCCR project (NCT03293563). PSMA expression was assessed by autoradiography using [177Lu]Lu-PSMA-617 as binding agent and the specific binding (total binding-non-specific binding) was calculated and expressed as a percentage of total binding. A patient suffering from metastatic ccRCC was also administered [68Ga]Ga-PSMA-11 to evaluate PSMA expression. RESULTS The mean specific binding was 28.9 ± 40.4% for primary renal cancer and 65.0 ± 38.9% for metastasis. Regarding histology, high PSMA expression was depicted in 33.3% of ccRCC, 33.3% of chRCC and 57.1% of papRCC. PSMA was more frequently expressed in primary samples of papRCC histology with renal capsule invasion (p = 0.0286). A higher PSMA-specific binding and a higher number of samples with high PSMA-expression were depicted in metastatic samples. Bone metastasis showed lower binding than other metastatic sites combined (p = 0.0005). The patient suffering from metastatic ccRCC showed high [68Ga]Ga-PSMA-11 uptake on known distant metastases and additional site uncovered. CONCLUSION PSMA showed high expression in metastases of ccRCC. CLINICAL TRIAL REGISTRATION NCT, NCT03293563, prospectively registered September 20, 2017, http://www. CLINICALTRIALS gov .
Collapse
Affiliation(s)
- Salma Binzaqr
- Department of Nuclear Medicine, University Hospital of Bordeaux, Bordeaux, F-33076, France
| | - David Kryza
- Univ. Lyon - Université Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, Villeurbanne, F-69100, France
- Léon Bérard Comprehensive Cancer center, Nuclear medicine department LUMEN, 15 rue Gabriel Sarrazin, cedex 08, Lyon, F-69373, France
| | - Anne-Laure Giraudet
- Univ. Lyon - Université Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS, Villeurbanne, F-69100, France
| | - Jean Christophe Bernhard
- Department of Urology, University Hospital of Bordeaux, Bordeaux, F-33076, France
- I. Care Bordeaux, University of Bordeaux, Bordeaux, F-33076, France
| | - Marine Gross-Goupil
- Department of Urology, University Hospital of Bordeaux, Bordeaux, F-33076, France
| | - Mokrane Yacoub
- Department of Pathology, University Hospital of Bordeaux, Bordeaux, F-33076, France
| | - Gaelle Margue
- Department of Urology, University Hospital of Bordeaux, Bordeaux, F-33076, France
- I. Care Bordeaux, University of Bordeaux, Bordeaux, F-33076, France
| | - Elif Hindié
- Department of Nuclear Medicine, University Hospital of Bordeaux, Bordeaux, F-33076, France
- University of Bordeaux, UMR CNRS 5287, INCIA, Talence, F-33400, France
- Institut Universitaire de France (IUF), Paris, F-75000, France
| | - Clément Morgat
- Department of Nuclear Medicine, University Hospital of Bordeaux, Bordeaux, F-33076, France.
- University of Bordeaux, UMR CNRS 5287, INCIA, Talence, F-33400, France.
| |
Collapse
|
41
|
Yang Z, Wan J, Zhang X, Mei J, Hao H, Liu S, Yi Y, Jiang M, He Y. Baicalin reduces sunitinib-induced cardiotoxicity in renal carcinoma PDX model by inhibiting myocardial injury, apoptosis and fibrosis. Front Pharmacol 2025; 16:1563194. [PMID: 40264678 PMCID: PMC12011809 DOI: 10.3389/fphar.2025.1563194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Sunitinib (SU), a multi-targeted tyrosine kinase inhibitor, has anticancer function but its clinical use is often limited by cardiovascular complications. Baicalin (BA) has demonstrated various pharmacological activities including antioxidant, anti-inflammatory and antiviral properties, but its potential roles in SU-induced cardiotoxicity have not been reported. In this study, we aimed to investigate the effect of BA in SU-induced cardiotoxicity in vivo by using renal carcinoma patient-derived xenograft (PDX) model. Female Nod Scid mice with renal carcinoma PDX were treated with vehicle, SU (50 mg/kg/d), BA (100 mg/kg/d), or BA combined with SU for 6 weeks. The tumor volume and weight of tumor-bearing mice were measured, and cardiovascular functions were evaluated by testing the Heart index and blood biochemical indicators, and by hematoxylin and eosin (H&E), Masson and Tunel staining. The results showed that SU therapy and combination therapy effectively inhibited the growth of renal tumors. Combination therapy inhibited SU-induced increase of creatine kinase (CK) and lactate dehydrogenase (LDH), and ameliorated the heart parameters. Moreover, BA effectively protected SU-induced cardiac dysfunction by decreasing injury, apoptosis, and fibrosis. Collectively, our results demonstrate that BA can be as a potential cardioprotective approach for cardiovascular complications during SU regimen.
Collapse
Affiliation(s)
- Zefu Yang
- Cardiovascular Medicine Department of Nanhai District People’s Hospital, Foshan, Guangdong, China
- Cardiovascular Medicine Department of The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Jianping Wan
- Electrophysiology Department of The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Xinjin Zhang
- Cardiovascular Center, Affiliated Hospital of Yunnan University, Kunming, China
| | - Jiaqi Mei
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hua Hao
- Department of Pathology, Yangpu District Central Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Sibo Liu
- The Queen MARY school, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yun Yi
- Center of Biobank, Nanchang University Second Affiliated Hospital, Jiangxi Medical College, Nanchang, China
| | - Meixiu Jiang
- The Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang, China
- Nanchang Royo Biotechnology, Nanchang, China
| |
Collapse
|
42
|
Guo Y, Lin Z, Zhou Z, Zhang W, Mao S, Shan Z, Wu P, Yao X. Oncogenic and immunological functions of USP35 in pan-cancer and its potential as a biomarker in kidney clear cell carcinoma. BMC Cancer 2025; 25:617. [PMID: 40188027 PMCID: PMC11972461 DOI: 10.1186/s12885-025-13964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Ubiquitin-specific protease 35 (USP35) has gained attention as a regulator in cancer progression. However, its specific role in kidney clear cell carcinoma (KIRC) remains unclear. METHODS USP35 expression in KIRC tumor and normal tissues was evaluated using TCGA data. Correlations between USP35 expression, clinical parameters, and survival outcomes were examined. Functional enrichment analyses were performed to explore the pathways associated with USP35 expression. Immune-related analyses were conducted to assess the effect of USP35 on immune cell recruitment and neoantigen presentation. Drug sensitivity analyses were used to identify potential therapeutic agents targeting USP35. RESULTS USP35 was significantly overexpressed in KIRC tumor tissues compared to normal tissues, and its high expression correlated with advanced disease stages and poor survival outcomes. Gene set enrichment analysis revealed that high USP35 expression was associated with oncogenic pathways, including glycerophospholipid and linoleic acid metabolism, while low expression linked to nitrogen and purine metabolism. USP35 also modulated immune responses, affecting immune cell recruitment and neoantigen presentation, suggesting a role in immune evasion. Drug sensitivity analysis showed that high USP35 expression correlated with increased sensitivity to paclitaxel, bosutinib, and lapatinib. In vitro knockdown of USP35 significantly reduced KIRC cell proliferation, migration, and epithelial-mesenchymal transition (EMT), further supporting its role in tumor progression. CONCLUSION USP35 is overexpressed in KIRC and associated with poor prognosis, likely promoting tumor progression through oncogenic pathways and immune modulation. Its correlation with drug sensitivity positions USP35 as a potential therapeutic target, warranting further investigation into its mechanistic functions and therapeutic applications.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Kidney Neoplasms/immunology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/mortality
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/drug therapy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Ubiquitin-Specific Proteases/genetics
- Ubiquitin-Specific Proteases/metabolism
- Gene Expression Regulation, Neoplastic
- Prognosis
- Cell Line, Tumor
- Cell Proliferation
- Epithelial-Mesenchymal Transition
Collapse
Affiliation(s)
- Yadong Guo
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Ziyou Lin
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zijing Zhou
- Laboratory of Ruijin Hospitalaffiliated to, Wuxi Branchaq, Shanghai Jiaotong University School of Medicine, Wuxi, Jiangsu, China
| | - Wentao Zhang
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Mao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zezhi Shan
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Pengfei Wu
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Xudong Yao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Liu J, Zhang X, Lv R, Zhang X, Wang R, Zeng X. Predictive value of extracellular volume fraction determined using enhanced computed tomography for pathological grading of clear cell renal cell carcinoma: a preliminary study. Cancer Imaging 2025; 25:49. [PMID: 40186299 PMCID: PMC11969730 DOI: 10.1186/s40644-025-00866-0] [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: 01/16/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE To explore the potential of using the extracellular volume fraction (ECV), measured through enhanced computed tomography (CT), as a tool for determining the pathological grade of clear cell renal cell carcinoma (ccRCC). METHODS This retrospective study, approved by the institutional review board, included 65 patients (median age: 58.40 ± 10.84 years) who were diagnosed with ccRCC based on the nucleolar grading of the International Society of Urological Pathology (ISUP). All patients underwent preoperative abdominal enhanced CT between January 2022 and August 2024. CT features from the unenhanced, corticomedullary, nephrographic, and delayed phases were analyzed, and the extracellular volume fraction (ECV) of ccRCC was calculated by measuring CT values from regions of interest in both the unenhanced and nephrographic phases. Statistical significance was evaluated for differences in these parameters across the four ISUP grades. Additionally, diagnostic efficiency was assessed using receiver operating characteristic (ROC) curve analysis. RESULTS The ECV showed significant differences across the four ISUP grades of ccRCC, its potential as an important predictor of high-grade ccRCC (P = 0.035). The ROC curve analysis indicated that ECV exhibited the highest diagnostic efficacy for assessing the lower- and higher- pathological grade of ccRCC, with an area under the ROC curve of 0.976. The optimal diagnostic threshold for ECV was determined to be 41.64%, with a sensitivity of 91.31% and a specificity of 97.62%. CONCLUSIONS ECV derived from enhanced CT has the potential to function as an in vivo biomarker for distinguishing between lower- and higher-grade ccRCC. This quantitative measure provides diagnostic value that extends beyond traditional qualitative CT features, offering a more precise and objective assessment of tumor grade.
Collapse
Affiliation(s)
- Jian Liu
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing & Cognitive Intelligence, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Ministry of Education, Guizhou University, No. 2708, Huaxi Avenue, Guiyang, 550025, Guizhou, China
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xunlan Zhang
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Rui Lv
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xiaoyong Zhang
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Rongpin Wang
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xianchun Zeng
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
44
|
Sun Z, Wang Y, Zheng C, Xiao L, Zang Y, Fang L, Cui X, Chang M, Sun Q, Li W, Ren J. NAT10 promotes the progression of clear cell renal cell carcinoma by regulating ac4C acetylation of NFE2L3 and activating AKT/GSK3β signaling pathway. Cell Death Dis 2025; 16:235. [PMID: 40169553 PMCID: PMC11962090 DOI: 10.1038/s41419-025-07528-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of renal cell carcinoma, and the tumour growth and metastasis of ccRCC are related to prognosis. N4-acetylcytidine (ac4C) is one of the major modifications of RNA and is known to be mediated by N-acetyltransferase 10 (NAT10). The role of NAT10 in cancer is gradually being revealed, although the role of NAT10-mediated RNA ac4C modification in ccRCC has not been reported. In this study, NAT10 was found to be upregulated in ccRCC tissues and associated with a poor prognosis in patients. HIF-1α activated NAT10 expression at the transcriptional level. CCK8, EdU, Transwell and scratch assays after NAT10 knockdown or overexpression showed that NAT10 promoted cell proliferation and migration. The results of subcutaneous xenograft and caudal vein injection showed that NAT10 promoted tumour growth and metastasis in vivo, while Remodelin inhibited tumour growth. The acRIP-seq, RIP, RNA stability and dual luciferase reporter experiments showed that NAT10 activated ac4C acetylation of NFE2L3 mRNA and promoted NFE2L3 mRNA stability. The ChIP-seq results showed that NFE2L3 regulated the expression of LASP1 and thus activated the AKT/GSK3β signalling pathway. In summary, our results suggest that NAT10 mediates ac4C acetylation of NFE2L3 mRNA, promotes its mRNA stability, regulates the LASP1-AKT/GSK3β/β-catenin axis and promotes the progression of renal clear cell carcinoma.
Collapse
Affiliation(s)
- Zenghui Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Yuqiong Wang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Chao Zheng
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Lixiang Xiao
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Liang Fang
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Xixi Cui
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Mingjie Chang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Qiyu Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Wenjuan Li
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China.
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China.
| |
Collapse
|
45
|
Assal RA, Rashwan HH, Zakaria ZI, Sweillam JH, Fouda YM, Abdelhamid AM, Youness RA. Deciphering the mysteries of MEG3 LncRNA and its implications in genitourinary cancers. Front Oncol 2025; 15:1519103. [PMID: 40242248 PMCID: PMC12000830 DOI: 10.3389/fonc.2025.1519103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/28/2025] [Indexed: 04/18/2025] Open
Abstract
Maternally expressed gene 3 (MEG3), a long non-coding RNA, plays a pivotal role in various biological processes, including tumorigenesis. Aberrant expression of MEG3 has been implicated in several cancers, including genitourinary malignancies. This comprehensive review explores the multifaceted functions of MEG3 in the context of genitourinary cancers through unravelling the molecular mechanisms underlying the influence of MEG3 on cellular proliferation, apoptosis, invasion, and metastasis. Additionally, we discuss the potential clinical implications of MEG3 as a biomarker and therapeutic target in genitourinary cancers. By unraveling the intricate role of MEG3 in these biological processes, this review aims to contribute to the development of novel strategies for the diagnosis and treatment of genitourinary malignancies.
Collapse
Affiliation(s)
- Reem A. Assal
- Department of Pharmacology and Toxicology, Heliopolis University for Sustainable Development (HU), Cairo, Egypt
| | - Hannah H. Rashwan
- Bioinformatics Group, Center for Informatics Science (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Zeina I. Zakaria
- Faculty of Biology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jana H. Sweillam
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University, Cairo, Egypt
| | - Yasmine M. Fouda
- Faculty of Medicine, Al-Kasr Al Ainy, Cairo University, Cairo, Egypt
| | | | - Rana A. Youness
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University, Cairo, Egypt
| |
Collapse
|
46
|
Montalvan L, Bernhard JC, Bensalah K, Paparel P, Bigot P, Doumerc N, Olivier J, Bruyère F, Ingels A, Rouprêt M, Audenet F, Lebacle C, Long JA, Durand X, Waeckel T, Durand M, Lang H, Pignot G, Cussenot O, Charles T, Tambwe R, Xylinas E, Boissier R, Patard JJ, Beauval JB, Mallet R, Rouget B, Nouhaud FX. Effect of obesity on histological type of renal tumor: The Uroccr study 69. THE FRENCH JOURNAL OF UROLOGY 2025; 35:102884. [PMID: 40185313 DOI: 10.1016/j.fjurol.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Renal cell carcinomas (RCC) are increasing worldwide and obesity is one of its recognized causal factor. Obesity thus could be differently associated with the different histologic types of renal tumors and also impact prognosis and patient care. PURPOSE The primary objective was to evaluate an association between obesity and histological type of RCC. The secondary objective was to assess if there was an effect of obesity on tumor characteristics. METHODS A multicenter comparative descriptive study was carried out in nearly 30 French hospitals. Histological features of operated RCC of patients included between 2007 and 2020 were compared according to their BMI. Obesity was defined as a BMI≥30kg/m2, with a P<0.05. RESULTS In total, 6749 RCC were analyzed, including 1687 (25%) from obese patients. This population was more frequently diabetic (61% versus 41%, P<0.001) and hypertensive (27% versus 11%, P<0.001). Regarding histological evaluation, the obese group presented clearer cell carcinoma. Other types presented odd ratios below 1 on multivariate analysis, P<0.01 except for type II papillary RCC. Tumor size (pT) and nuclear ISUP grade were both lower when BMI was above 30kg/m2: 72.4% of size pT 1-2 versus 68.5% (P=0.026) and 50% of nuclear ISUP grade 1-2 versus 43.3% for the non-obese group (P<0.0001). There was no difference in survival due to an insufficient number of events. CONCLUSION In this large cohort study, we report novel data on a positive association between obesity and histological type of RCC, in particular clear cell RCC. LEVEL OF EVIDENCE: 3
Collapse
|
47
|
Shiota M, Nemoto S, Ikegami R, Tanegashima T, Blas L, Miyake H, Takahashi M, Oya M, Tsuchiya N, Masumori N, Kobayashi K, Obara W, Shinohara N, Fujimoto K, Nozawa M, Ohba K, Ohyama C, Hashine K, Akamatsu S, Motoshima T, Mita K, Gotoh M, Tatarano S, Fujisawa M, Tomita Y, Mukai S, Ito K, Eto M. Predictive Model of Objective Response to Nivolumab Monotherapy for Advanced Renal Cell Carcinoma by Machine Learning Using Genetic and Clinical Data: The SNiP-RCC Study. JCO Clin Cancer Inform 2025; 9:e2400167. [PMID: 40279530 DOI: 10.1200/cci-24-00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/10/2024] [Accepted: 02/24/2025] [Indexed: 04/27/2025] Open
Abstract
PURPOSE Anti-PD-1 antibodies are widely used for cancer treatment, including in advanced renal cell carcinoma (RCC). However, the therapeutic response varies among patients. This study aimed to predict tumor response to nivolumab anti-PD-1 antibody treatment for advanced RCC by integrating genetic and clinical data using machine learning (ML). METHODS Clinical and single-nucleotide polymorphism (SNP) data obtained in the SNPs in nivolumab PD-1 inhibitor for RCC study, which enrolled Japanese patients treated with nivolumab monotherapy for advanced clear cell RCC, were used. A point-wise linear (PWL) algorithm, logistic regression with elastic-net regularization, and eXtreme Gradient Boosting were used in this study. AUC values for objective response and C-indices for progression-free survival (PFS) were calculated to evaluate the utility of the models. RESULTS Among the three ML algorithms, the AUC values to predict objective response were highest for the PWL algorithm among all the data sets. Three predictive models (clinical model, small SNP model, and large SNP model) were created by the PWL algorithm using the clinical data alone and using eight and 49 SNPs in addition to the clinical data. C-indices for PFS by the clinical model, small SNP model, and large SNP model were 0.522, 0.600, and 0.635, respectively. CONCLUSION The results demonstrated that the SNP models created by ML produced excellent predictions of tumor response to nivolumab monotherapy for advanced clear cell RCC and will be helpful in treatment decisions.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shota Nemoto
- Industrial & Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Ryo Ikegami
- Industrial & Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Leandro Blas
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideaki Miyake
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masayuki Takahashi
- Department of Urology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiko Tsuchiya
- Department of Urology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Naoya Masumori
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keita Kobayashi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Masahiro Nozawa
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kojiro Ohba
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Katsuyoshi Hashine
- Department of Urology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takanobu Motoshima
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Mita
- Department of Urology, Hiroshima City Asa Citizens Hospital, Hiroshima, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shuichi Tatarano
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiko Tomita
- Department of Urology and Molecular Oncology, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Shoichiro Mukai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Keiichi Ito
- Department of Urology, National Defense Medical College, Saitama, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
48
|
Lu Z, Xu J, Li J. The Transcription Factor ATF2 Accelerates Clear Cell Renal Cell Carcinoma Progression Through Activating the PLEKHO1/NUS1 Pathway. Mol Carcinog 2025; 64:617-628. [PMID: 39777695 DOI: 10.1002/mc.23868] [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: 09/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignant cancer with high mortality rate. Activating transcription factor 2 (ATF2) and pleckstrin homology domain containing O1 (PLEKHO1) were reported to participate in numerous cancers. However, their roles and the detailed mechanisms in ccRCC development remain largely unknown. RT-qPCR and western blot were used to measure the levels of PLEKHO1, ATF2, and nuclear undecaprenyl pyrophosphate synthase 1 (NUS1). Cell proliferation, apoptosis, invasion, migration and stemness were evaluated using CCK-8 assay, flow cytometry, transwell invasion assay, wound-healing assay and sphere formation assay, respectively. Dual-luciferase reporter assay was conducted to verify the relationship between ATF2 and PLEKHO1. The interaction between PLEKHO1 and NUS1 was proved by Co-IP assay. Xenograft models were utilized to evaluate the tumorigenic capability of ccRCC cells upon PLEKHO1 knockdown. PLEKHO1, ATF2 and NUS1 expression were significantly elevated in ccRCC, and PLEKHO1 might be a prognosis biomarker for ccRCC. PLEKHO1 depletion significantly inhibited cell proliferation, invasion, migration, stemness, and induced cell apoptosis in ccRCC cells. ATF2 activated PLEKHO1 expression via transcription regulation, and PLEKHO1 overexpression could reverse the suppressive effects of ATF2 knockdown on the malignant behaviors of ccRCC cells. Moreover, PLEKHO1 directly bound to NUS1, and PLEKHO1 depletion markedly restrained ccRCC progression through targeting NUS1 in vitro and in vivo. Our findings suggested that ATF2 transcriptionally activated PLEKHO1 to promote the development of ccRCC via regulating NUS1 expression.
Collapse
Affiliation(s)
- Zheng Lu
- Gravel Center, Nanyang First People's Hospital, Nanyang, China
| | - Jinge Xu
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Junyu Li
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
49
|
Alves Â, Ferreira M, Eiras M, Lima L, Medeiros R, Teixeira AL, Dias F. Exosome-derived hsa-miR-200c-3p, hsa-miR-25-3p and hsa-miR-301a-3p as potential biomarkers and therapeutic targets for restoration of PTEN expression in clear cell renal cell carcinoma. Int J Biol Macromol 2025; 302:140607. [PMID: 39900161 DOI: 10.1016/j.ijbiomac.2025.140607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is an aggressive kidney cancer subtype with limited biomarkers and therapeutic options. Thus, the present study aimed to evaluate the biomarker and therapeutic potential of Phosphatase and Tensin Homologue (PTEN)-regulating microRNAs (miRNAs) in 2D and 3D ccRCC models. Extracellular vesicles (EVs) from four renal cell lines were characterized, and selected miRNAs (hsa-miR-200c-3p, hsa-miR-25-3p, and hsa-miR-301a-3p) were quantified in cells and EVs. PTEN mRNA levels were measured intracellularly. 786-O cells were transfected with miRNA inhibitors in both models and effects on miRNA and PTEN expression were assessed alongside phenotypic alterations. The expression of target miRNAs increased with ccRCC cell aggressiveness, both intracellularly and in EVs, while PTEN mRNA expression decreased. Combined inhibition of these miRNAs significantly increased PTEN expression, reducing tumor cell proliferation and migration in 2D models and decreasing spheroid size and metabolic capacity in 3D models. These miRNAs show potential as biomarkers for monitoring disease aggressiveness and as therapeutic targets in ccRCC, potentially leading to more effective and personalized treatment approaches for ccRCC patients.
Collapse
Affiliation(s)
- Ângela Alves
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Ferreira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Eiras
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Luís Lima
- Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Faculty of Medicine (FMUP), University of Porto, 4200-319 Porto, Portugal; Laboratory Medicine, Clinical Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; Biomedicine Research Center (CEBIMED), Research Innovation and Development Institute (FP-I3ID), Faculty of Health Sciences, Fernando Pessoa University (UFP), 4249-004 Porto, Portugal; Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| |
Collapse
|
50
|
Wei Z, Ye Y, Liu C, Wang Q, Zhang Y, Chen K, Cheng G, Zhang X. MIER2/PGC1A elicits sunitinib resistance via lipid metabolism in renal cell carcinoma. J Adv Res 2025; 70:287-305. [PMID: 38702028 PMCID: PMC11976417 DOI: 10.1016/j.jare.2024.04.032] [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: 02/04/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is one of the most common malignant tumors of the urinary system and accounts for more than 90 % of all renal tumors. Resistance to targeted therapy has emerged as a pivotal factor that contributes to the progressive deterioration of patients with advanced RCC. Metabolic reprogramming is a hallmark of tumorigenesis and progression, with an increasing body of evidence indicating that abnormal lipid metabolism plays a crucial role in the advancement of renal clear cell carcinoma. OBJECTIVES Clarify the precise mechanisms underlying abnormal lipid metabolism and drug resistance. METHODS Bioinformatics screening and analyses were performed to identify hub gene. qRT-PCR, western blot, chromatin immunoprecipitation (ChIP) assays, and other biological methods were used to explore and verify related pathways. Various cell line models and animal models were used to perform biological functional experiments. RESULTS In this study, we identified Mesoderm induction early response 2 (MIER2) as a novel biomarker for RCC, demonstrating its role in promoting malignancy and sunitinib resistance by influencing lipid metabolism in RCC. Mechanistically, MIER2 facilitated P53 deacetylation by binding to HDAC1. Acetylation modification augmented the DNA-binding stability and transcriptional function of P53, while deacetylation of P53 hindered the transcriptional process of PGC1A, leading to intracellular lipid accumulation in RCC. Furthermore, Trichostatin A (TSA), an inhibitor of HDAC1, was found to impede the MIER2/HDAC1/P53/PGC1A pathway, offering potential benefits for patients with sunitinib-resistant renal cell cancer. CONCLUSION Our findings highlight MIER2 as a key player in anchoring HDAC1 and inhibiting PGC1A expression through the deacetylation of P53, thereby inducing lipid accumulation in RCC and promoting drug resistance. Lipid-rich RCC cells compensate for energy production and sustain their own growth in a glycolysis-independent manner, evading the cytotoxic effects of targeted drugs and ultimately culminating in the development of drug resistance.
Collapse
Affiliation(s)
- Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunxuan Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Shenzhen Huazhong University of Science and Technology Research Institute, China.
| |
Collapse
|