1
|
Meng K, Chen Z, Zhang Y, Chen X, Chen T, Song Y, Jia X, Liu M. Unraveling the mechanisms of tricetin in renal cell carcinoma treatment through network pharmacology and experimental validation. Med Oncol 2025; 42:192. [PMID: 40319129 DOI: 10.1007/s12032-025-02744-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: 02/13/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Renal cell carcinoma (RCC), one of the most common types of kidney cancer, represents a major global health concern due to its increasing incidence and high death rates. While conventional treatment modalities have improved patient outcomes, they often face limitations such as drug resistance, severe adverse effects, and limited efficacy in advanced or metastatic cases. These challenges highlight the urgent need for novel therapeutic approaches to enhance RCC management. Tricetin, a natural flavonoid abundant in cereal plants, has shown promising anticancer activity in various malignancies by inhibiting cancer cell proliferation, migration, and invasion. However, the molecular mechanisms underlying Tricetin's effects on RCC remain poorly understood. In this research, we employed network pharmacology to identify key molecular targets of Tricetin in RCC and evaluated its binding affinity to these targets using molecular docking techniques. Bioinformatics analyses were conducted to predict the potential biological pathways involved. Furthermore, in vitro experiments using RCC cell lines (786-O and ACHN) demonstrated that Tricetin suppresses cell proliferation and migration, likely through modulation of the EGFR/PI3K/Akt signaling pathway. Overall, our findings offer new insights into the therapeutic potential of Tricetin and provide a foundation for developing targeted treatment strategies to improve RCC outcomes.
Collapse
Affiliation(s)
- Kai Meng
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Zixuan Chen
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yu Zhang
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xingyu Chen
- School of Health Policy and Management, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Taoying Chen
- School of Food Science and Engineering, Guizhou Province, Guiyang University, Nanming St, Guiyang, 550005, People's Republic of China
| | - Ya Song
- School of Life Sciences, Bengbu Medical University, Bengbu, 233000, China
| | - Xing Jia
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Min Liu
- Department of Urology, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
2
|
Younis OM, Dhaydel AS, Alghwyeen WF, Abu Hantash NR, Allan LM, Qasem IM, Saeed A. The role of ANGPTL4 in cancer: A meta-analysis of observational studies and multi-omics investigation. PLoS One 2025; 20:e0320343. [PMID: 40233044 PMCID: PMC11999138 DOI: 10.1371/journal.pone.0320343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/16/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Angiopoietin-like protein 4 (ANGPTL4) plays a crucial role in processes such as angiogenesis, inflammation, and metabolism. Despite numerous studies suggesting its involvement in cancer, a definitive role remains unclear. We introduce the first comprehensive meta-analysis and pan-cancer bioinformatics study on ANGPTL4, aiming to unravel its implications across various cancer types. METHODS Moderate-to high-quality observational studies were retrieved from PubMed, Scopus, and Embase. A meta-analysis was conducted using the R package "meta." Survival analysis was performed using GEPIA2 and TIMER2.0. Immune infiltration, mutational burden, and drug resistance analyses was done via GSCAlite. Co-expression and gene set enrichment analyses (GSEA) were carried out using cBioportal and enrichr, respectively. RESULTS Increased ANGPTL4 expression was linked to worse tumor grade (OR = 1.51, P = 0.023), stage (OR = 2.42, P < 0.001), lymph node metastasis (OR = 1.76, P = 0.012), vascular invasion (OR = 2.16, P = 0.01), and lymphatic invasion (OR = 2.20, P < 0.001). Furthermore, ANGPTL4 expression was linked to worse OS (HR = 1.40, 95% CI: 1.29,1.50, P = 0.0001). Single gene level analysis revealed that ANGPTL4 upregulated epithelial-to-mesenchymal transition (EMT) in 23 different cancers. Immune infiltration varied between cancer types, but increased infiltration of cancer-associated fibroblasts was observed in most cancers. Mutation analysis revealed increased alterations in TP53 and CDKN2A in cohorts with ANGPTL4 alterations. GSEA of co-expressed genes revealed involvement in hypoxia, EMT, VEGF-A complex, TGF-B pathways, and extracellular matrix organization. CONCLUSIONS ANGPTL4 plays a significant role in tumor progression via its positive regulation of EMT and angiogenesis, while possibly harboring a TGF-B dependent role in systemic metastasis. Therefore, ANGPTL4 is a suitable target for future drug development.
Collapse
Affiliation(s)
- Osama M. Younis
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, United States of America
| | | | | | | | - Leen M. Allan
- School of Medicine, The Hashemite University, Al Zarqaa, Jordan
| | | | - Anwaar Saeed
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
3
|
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
|
4
|
Grigoraș A, Amalinei C. The Role of Perirenal Adipose Tissue in Carcinogenesis-From Molecular Mechanism to Therapeutic Perspectives. Cancers (Basel) 2025; 17:1077. [PMID: 40227577 PMCID: PMC11987925 DOI: 10.3390/cancers17071077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Perirenal adipose tissue (PRAT) exhibits particular morphological features, with its activity being mainly related to thermogenesis. However, an expanded PRAT area seems to play a significant role in cardiovascular diseases, diabetes mellitus, and chronic kidney disease pathogenesis. Numerous studies have demonstrated that PRAT may support cancer progression and invasion, mainly in obese patients. The mechanism underlying these processes is of dysregulation of PRAT's secretion of adipokines and pro-inflammatory cytokines, such as leptin, adiponectin, chemerin, apelin, omentin-1, vistatin, nesfatin-1, and other pro-inflammatory cytokines, modulated by tumor cells. Cancer cells may also induce a metabolic reprogramming of perirenal adipocytes, leading to increased lipids and lactate transfer to the tumor microenvironment, contributing to cancer growth in a hypoxic milieu. In addition, the PRAT browning process has been specifically detected in renal cell carcinoma (RCC), being characterized by upregulated expression of brown/beige adipocytes markers (UCP1, PPAR-ɣ, c/EBPα, and PGC1α) and downregulated white fat cells markers, such as LEPTIN, SHOX2, HOXC8, and HOXC9. Considering its multifaceted role in cancer, modulation of PRAT's role in tumor progression may open new directions for oncologic therapy improvement. Considering the increasing evidence of the relationship between PRAT and tumor cells, our review aims to provide a comprehensive analysis of the perirenal adipocytes' impact on tumor progression and metastasis.
Collapse
Affiliation(s)
- Adriana Grigoraș
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| | - Cornelia Amalinei
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| |
Collapse
|
5
|
Zhang S, Miao H, Han T, Wu X, Liang C, Qian J, Shao P. MCM8 promotes NR4A1-mediated E2F1 transcription and facilitates renal cell carcinoma through enhancing aerobic glycolysis. Cell Biol Toxicol 2025; 41:51. [PMID: 39992472 PMCID: PMC11850455 DOI: 10.1007/s10565-025-10002-0] [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: 08/04/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
Renal cell carcinoma (RCC) is a type of renal malignancy originated from the urinary tubular epithelial system. Despite its high incidence, the molecular mechanisms driving its pathogenesis remain poorly understood, limiting therapeutic advancements. This study explored the link between MCM8 and RCC progression. MCM8 displays significantly high expression in RCC tissues and was closely associated with RCC pathological staging. Knocking down endogenous MCM8 in RCC cells significantly suppressed malignant phenotypes, while simultaneously inducing apoptosis. Similarly, in vivo experiments confirmed these findings, showing a pronounced reduction in tumor growth upon MCM8 silencing. Mechanistic investigations revealed that MCM8 regulates E2F1 expression by interacting with the transcription factor NR4A1, thereby affecting E2F1 transcriptional activity. Additionally, MCM8 and E2F1 collaboratively influence aerobic glycolysis and the cellular behavior of RCC cells. In conclusion, this study identifies MCM8 as a tumor-promoting factor in RCC, with its oncogenic role potentially mediated by its regulation of E2F1 expression.
Collapse
MESH Headings
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/genetics
- E2F1 Transcription Factor/metabolism
- E2F1 Transcription Factor/genetics
- Humans
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Cell Line, Tumor
- Glycolysis
- Animals
- Gene Expression Regulation, Neoplastic
- Mice
- Mice, Nude
- Male
- Apoptosis
- Female
- Transcription, Genetic
- Cell Proliferation
Collapse
Affiliation(s)
- Shaobo Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haoqi Miao
- Nanjing Medical University, Nanjing, 210029, China
| | - Tian Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiangzhen Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chao Liang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jian Qian
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Pengfei Shao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
6
|
Mahadevan A, Yazdanpanah O, Patel V, Benjamin DJ, Kalebasty AR. Ophthalmologic toxicities of antineoplastic agents in genitourinary cancers: Mechanisms, management, and clinical implications. Curr Probl Cancer 2025; 54:101171. [PMID: 39708456 DOI: 10.1016/j.currproblcancer.2024.101171] [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: 08/06/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
Genitourinary cancers affect over 480,000 patients in the United States annually. While promising therapeutic modalities continue to emerge, notably immune checkpoint inhibitors, molecular targeted therapies, antibody-drug conjugates, and radioligand therapies, these treatments are associated with a spectrum of adverse side-effects, including ophthalmologic toxicities. In this review, we cover the most commonly used antineoplastic agents for the kidneys, bladder, urinary tracts, prostate, testis, and penis, detailing mechanism, indication, and recent trials supporting their use. For each category of antineoplastic therapy, we describe the epidemiology, management, and clinical presentation, of common ophthalmologic toxicities stemming from these agents. This review serves to augment awareness and recognition of possible ophthalmologic manifestations resulting from the use of antineoplastic agents in genitourinary malignancy. Early identification of these side effects can hasten ophthalmology referral and ultimately improve visual outcomes in patients experiencing medication-induced ocular toxicities.
Collapse
Affiliation(s)
- Aditya Mahadevan
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Omid Yazdanpanah
- Division of Hematology/Oncology, University of California Irvine Health, Orange, CA, USA.
| | - Vivek Patel
- Department of Ophthalmology, University of California Irvine Health, Orange, CA, USA.
| | | | | |
Collapse
|
7
|
Feng X, Wang Z, Cen M, Zheng Z, Wang B, Zhao Z, Zhong Z, Zou Y, Lv Q, Li S, Huang L, Huang H, Qiu X. Deciphering potential molecular mechanisms in clear cell renal cell carcinoma based on the ubiquitin-conjugating enzyme E2 related genes: Identifying UBE2C correlates to infiltration of regulatory T cells. Biofactors 2025; 51:e2143. [PMID: 39614426 DOI: 10.1002/biof.2143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Renal clear cell carcinoma (ccRCC) is a highly aggressive and common form of kidney cancer, with limited treatment options for advanced stages. Recent studies have highlighted the importance of the ubiquitin-proteasome system in tumor progression, particularly the role of ubiquitin-conjugating enzyme E2 (UBE2) family members. However, the prognostic significance of UBE2-related genes (UBE2RGs) in ccRCC remains unclear. In this study, bulk RNA-sequencing and single-cell RNA-sequencing data from ccRCC patients were retrieved from the Cancer Genome Atlas and Gene Expression Omnibus databases. Differential expression analysis was performed to identify UBE2RGs associated with ccRCC. A combination of 10 machine learning methods was applied to develop an optimal prognostic model, and its predictive performance was evaluated using area under the curve (AUC) values for 1-, 3-, and 5-year overall survival (OS) in both training and validation cohorts. Functional enrichment analyses of gene ontology and Kyoto Encyclopedia of Genes and Genomes were conducted to explore the biological pathways involved. Correlation analysis was conducted to investigate the association between the risk score and tumor mutational burden (TMB) and immune cell infiltration. Immunotherapy and chemotherapy sensitivity were assessed by immunophenoscore and tumor immune, dysfunction, and exclusion scores to identify potential predictive significance. In vitro, knockdown of the key gene UBE2C in 786-O cells by specific small interfering RNA to validate its impact on apoptosis, migration, cell cycle, migration, invasion of tumor cells, and induction of regulatory T cells (Tregs). Analysis of sc-RNA revealed that UBE2 activity was significantly upregulated in malignant cells, suggesting its role in tumor progression. A three-gene prognostic model comprising UBE2C, UBE2D3, and UBE2T was constructed by Lasoo Cox regression and demonstrated robust predictive accuracy, with AUC values of 0.745, 0.766, and 0.771 for 1-, 3-, and 5-year survival, respectively. The model was validated as an independent prognostic factor in ccRCC. Patients in the high-risk group had a worse prognosis, higher TMB scores, and low responsiveness to immunotherapy. Additionally, immune infiltration and chemotherapy sensitivity analyses revealed that UBE2RGs are associated with various immune cells and drugs, suggesting that UBE2RGs could be a potential therapeutic target for ccRCC. In vitro experiments confirmed that the reduction of UBE2C led to an increase in apoptosis rate, as well as a decrease in tumor cell invasion and metastasis abilities. Additionally, si-UBE2C cells reduced the release of the cytokine Transforming Growth Factor-beta 1 (TGF-β1), leading to a decreased ratio of Tregs in the co-culture system. This study presents a novel three-gene prognostic model based on UBE2RGs that demonstrates significant predictive value for OS, immunotherapy, and chemotherapy in ccRCC patients. The findings underscore the potential of UBE2 family members as biomarkers and therapeutic targets in ccRCC, warranting further investigation in prospective clinical trials.
Collapse
Affiliation(s)
- Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Zhenwei Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zongtai Zheng
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Bangqi Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhihui Zhong
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Yesong Zou
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Qian Lv
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Shiyu Li
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Hai Huang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Ruijin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Valdés A, Pizarro G, González-Montero J, Rojas C, Burotto M. Targeting HIF-2α: the role of belzutifan in clear cell renal carcinoma management. Expert Rev Clin Pharmacol 2025; 18:17-27. [PMID: 39670660 DOI: 10.1080/17512433.2024.2436433] [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: 08/30/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Belzutifan is a first-in-class hypoxia-inducible factor-2 alpha (HIF-2α) inhibitor. It targets the von Hippel-Lindau protein (pVHL)-HIF-vascular endothelial growth factor (VEGF) pathway, which is crucial in cellular responses to hypoxia. By inhibiting HIF-2α, belzutifan disrupts the transcription of genes involved in tumor growth and angiogenesis. AREAS COVERED In this review, we describe the pVHL-HIF-VEGF pathway and how it led to the development of HIF inhibitors, including belzutifan. A search was conducted for trials involving Belzutifan, including phase I-III trials. We describe the relevant toxicity, with emphasis on hypoxia and anemia. EXPERT OPINION Belzutifan is a relatively safe drug, with manageable adverse events, including anemia and hypoxia as on-target toxicity. Ongoing trials are studying its benefit in overall survival for RCC in first-line treatment and its potential in other malignancies. The LITESPARK-005 trial reported the benefit of belzutifan in progression-free survival (PFS) compared to everolimus in later lines of treatment, with improvement in quality-of-life outcomes. Given its different mechanism of action to currently available treatments, belzutifan is expected to play a prominent role in the treatment of clear cell renal carcinoma and other cancers.
Collapse
Affiliation(s)
- Alejandro Valdés
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
- Department of Medical Oncology, Instituto Nacional del Cáncer, Santiago, Chile
| | - Gonzalo Pizarro
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
- Department of Medical Oncology, Hospital Sótero del Río, Santiago, Chile
| | | | - Carlos Rojas
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| | - Mauricio Burotto
- Department of Medical Oncology, Bradford Hill Clinical Research Center, Santiago, Chile
| |
Collapse
|
9
|
Ran L, Hu G, Zhao R, Zhang C. The increased ANXA5 expression indicates poor prognosis in renal clear cell carcinoma. Asian J Surg 2024:S1015-9584(24)01796-2. [PMID: 39191573 DOI: 10.1016/j.asjsur.2024.08.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Affiliation(s)
- Lingxiang Ran
- Department of Urology, the first people's hospital of Hefei, Hefei, Anhui, 230041, China; Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China.
| | - Guangmo Hu
- Department of Urology, the first people's hospital of Hefei, Hefei, Anhui, 230041, China
| | - Rui Zhao
- Acupuncture Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Chao Zhang
- Department of Urology, the first people's hospital of Hefei, Hefei, Anhui, 230041, China
| |
Collapse
|
10
|
Ye Z, Xu J, Zhang X, Zhang Y, Ivanova D, Lu W, Zhang J, Li F, Chen X, Wang Y, Wang M, Xie B. Identification and Validation of Tumor Microenvironment-Associated Signature in Clear-Cell Renal Cell Carcinoma through Integration of DNA Methylation and Gene Expression. Int J Mol Sci 2024; 25:6792. [PMID: 38928496 PMCID: PMC11203551 DOI: 10.3390/ijms25126792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The tumor microenvironment (TME) is crucial in tumor development, metastasis, and response to immunotherapy. DNA methylation can regulate the TME without altering the DNA sequence. However, research on the methylation-driven TME in clear-cell renal cell carcinoma (ccRCC) is still lacking. In this study, integrated DNA methylation and RNA-seq data were used to explore methylation-driven genes (MDGs). Immune scores were calculated using the ESTIMATE, which was employed to identify TME-related genes. A new signature connected with methylation-regulated TME using univariate, multivariate Cox regression and LASSO regression analyses was developed. This signature consists of four TME-MDGs, including AJAP1, HOXB9, MYH14, and SLC6A19, which exhibit high methylation and low expression in tumors. Validation was performed using qRT-PCR which confirmed their downregulation in ccRCC clinical samples. Additionally, the signature demonstrated stable predictive performance in different subtypes of ccRCC. Risk scores are positively correlated with TMN stages, immune cell infiltration, tumor mutation burden, and adverse outcomes of immunotherapy. Interestingly, the expression of four TME-MDGs are highly correlated with the sensitivity of first-line drugs in ccRCC treatment, especially pazopanib. Molecular docking indicates a high affinity binding between the proteins and pazopanib. In summary, our study elucidates the comprehensive role of methylation-driven TME in ccRCC, aiding in identifying patients sensitive to immunotherapy and targeted therapy, and providing new therapeutic targets for ccRCC treatment.
Collapse
Affiliation(s)
- Zijian Ye
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jialiang Xu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Xin Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
| | - Yifan Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Weiyu Lu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
| | - Jianning Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Meijiao Wang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Yang S, Yang X, Hou Z, Zhu L, Yao Z, Zhang Y, Chen Y, Teng J, Fang C, Chen S, Jia M, Liu Z, Kang S, Chen Y, Li G, Niu Y, Cai Q. Rationale for immune checkpoint inhibitors plus targeted therapy for advanced renal cell carcinoma. Heliyon 2024; 10:e29215. [PMID: 38623200 PMCID: PMC11016731 DOI: 10.1016/j.heliyon.2024.e29215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Renal cell carcinoma (RCC) is a frequent urological malignancy characterized by a high rate of metastasis and lethality. The treatment strategy for advanced RCC has moved through multiple iterations over the past three decades. Initially, cytokine treatment was the only systemic treatment option for patients with RCC. With the development of medicine, antiangiogenic agents targeting vascular endothelial growth factor and mammalian target of rapamycin and immunotherapy, immune checkpoint inhibitors (ICIs) have emerged and received several achievements in the therapeutics of advanced RCC. However, ICIs have still not brought completely satisfactory results due to drug resistance and undesirable side effects. For the past years, the interests form researchers have been attracted by the combination of ICIs and targeted therapy for advanced RCC and the angiogenesis and immunogenic tumor microenvironmental variations in RCC. Therefore, we emphasize the potential principle and the clinical progress of ICIs combined with targeted treatment of advanced RCC, and summarize the future direction.
Collapse
Affiliation(s)
- Siwei Yang
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xianrui Yang
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zekai Hou
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Liang Zhu
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhili Yao
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | | | - Yanzhuo Chen
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jie Teng
- Affiliated Hospital of Hebei University, Baoding, China
| | - Cheng Fang
- Taihe County People's Hospital, Anhui, China
| | - Songmao Chen
- Department of Urology, Fujian Provincial Hospital, Fujian, China
- Provincial Clinical Medical College of Fujian Medical University, Fujian, China
| | - Mingfei Jia
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Hebei, China
| | - Zhifei Liu
- Department of Urology, Tangshan People's Hospital, Hebei, China
| | - Shaosan Kang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Hebei, China
| | - Yegang Chen
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gang Li
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qiliang Cai
- Department of Urology, Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|