1
|
Dong L, Zhang X, Yu X, Liu G, Yang L. Proteoglycan-degrading enzymes engineered for enhanced tumor microenvironment interaction in renal cell carcinoma. Int J Biol Macromol 2025; 307:140440. [PMID: 39884611 DOI: 10.1016/j.ijbiomac.2025.140440] [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: 10/15/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
This work optimized proteoglycan-degrading enzymes through targeted mutagenesis to enhance their interaction with the tumor microenvironment in Renal Cell Carcinoma (RCC). A comprehensive mutagenesis approach identified 60 key mutations significantly improving enzymatic activity, stability, and structural integrity. When compared to Wild Type (WT) enzyme, a remarkable increase in specific activity by 35 % (p < 0.001) and a considerable decrease in the Km values for hyaluronidases from 2.5 mM to 1.5 mM (p < 0.05), as a result of these modifications. Computational methods are then employed to analyze the active site of the enzymes to detect potential residues that may alter. These computational techniques include molecular docking and protein structure prediction. The structural models of the enzymes are created by utilizing homology modeling and crystallography. These models demonstrate the spatial arrangement of the amino acid enzymes. It also illustrated the specific mutations to improve the potential of enzymes to relate to the Extracellular Matrix (ECM) of tumors. The computational screening methods effectively predicted how the modifications impact enzyme catalytic efficiency and stability. The modified enzymes retained 85 % of the enzyme activity, while the WT retained 60 %. Thus, the modified enzymes demonstrated better thermal stability than WT. Vitro test analyses show that the proteoglycan breakdown was significantly reduced by 70 % (p < 0.001), and for effective proteoglycan breakdown, hyaluronidase concentration is needed. This work proposed a novel therapeutic approach called proteoglycan-degrading enzymes for the treatment of RCC. These proteoglycan-degrading enzymes are more stable and effective for treating RCC, as demonstrated in the outcomes. Customized proteoglycan-degrading enzymes make the therapy more effective. The effective breakdown of the tumor's ECM in RCC models establishes this customized proteoglycan-degrading enzyme. These enzymes are effective for this customized cancer treatment as they improve stability, activity, and interaction with the TME.
Collapse
Affiliation(s)
- Lingling Dong
- Second Department of Cardiovascular Medicine, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning Province, China
| | - Xiaoli Zhang
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110004, PR China
| | - Xiaopeng Yu
- Oncology Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Gang Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lina Yang
- Second Department of Cardiovascular Medicine, Shengjing Hospital Affiliated to China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110004, Liaoning Province, China.
| |
Collapse
|
2
|
Huang CY, Chen MC, Wu CY, Lin YC, Huang YL, Shiue HS, Pu YS, Hsueh YM. Interaction of tissue inhibitor of metalloproteinase 3 gene polymorphism, blood cadmium and total urinary arsenic levels on clear cell renal cell carcinoma. Sci Rep 2025; 15:10267. [PMID: 40133536 PMCID: PMC11937534 DOI: 10.1038/s41598-025-94807-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
In renal cell carcinoma (RCC), tissue inhibitor of metalloproteinase (TIMP) 3 expression is lost, suggesting that the TIMP3 gene may function as a tumor suppressor gene. Cadmium (Cd) and arsenic (As) exposure may affect the expression of TIMP3. Here we investigate the association of clear cell RCC with TIMP3 polymorphisms, and explore whether TIMP3 polymorphisms modify the relationship between blood Cd or total urinary As levels and clear cell RCC respectively. We recruited 281 clear cell RCC patients and 689 sex- and age-matched controls. The clear cell RCC was diagnosed by pathological evaluation after image-guided biopsy or surgical resection of the renal tumor. Concentrations of blood Cd and lead, and also total urinary As, were measured. We determined TIMP3 polymorphisms using the Agena Bioscience MassARRAY system. Odds ratio (OR) of clear cell RCC was significantly inversely correlated with TIMP3 rs9609643 GA/AA genotype, with OR = 0.63 (95% confidence interval, CI, 0.44-0.91). For TIMP3 rs715572 AA compared to the GG/GA genotype, the OR of clear cell RCC was 1.60 with 95% CI of 1.01-2.56. Individuals with high blood Cd concentrations and the TIMP3 rs9609643 GG genotype exhibited a higher OR of clear cell RCC than reference groups (OR = 4.48, 95% CI 2.09-9.60). This study presents a novel finding that the GA/AA genotype of TIMP3 rs9609643 significantly decreased the clear cell RCC risk, and AA genotype of TIMP3 rs715572 significantly increased the clear cell RCC risk. Furthermore, this study first identified that the TIMP3 rs9609643 risk genotypes appear to interact with high blood Cd levels to increase the OR of clear cell RCC.
Collapse
Affiliation(s)
- Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Chieh Chen
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Yin Wu
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chin Lin
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Geriatric Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ya-Li Huang
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 110, Taiwan
| | - Horng-Sheng Shiue
- Department of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yeong-Shiau Pu
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Mei Hsueh
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei, 110, Taiwan.
| |
Collapse
|
3
|
Santagata S, Trotta AM, D’Alterio C, Napolitano M, Rea G, Di Napoli M, Portella L, Ieranò C, Guardascione G, Coppola E, Caux C, Dubois B, Boyle HJ, Carles J, Rossetti S, Azzaro R, Feroce F, Perdonà S, Fordellone M, Bello AM, Califano D, Chiodini P, Pignata S, Scala S. KIR2DL2/DL3+NKs and Helios+Tregs in Peripheral Blood Predict Nivolumab Response in Patients with Metastatic Renal Cell Cancer. Clin Cancer Res 2024; 30:4755-4767. [PMID: 39167621 PMCID: PMC11474171 DOI: 10.1158/1078-0432.ccr-24-0729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/31/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE To identify predictive factors of nivolumab sensitivity, peripheral blood NKs and regulatory T-cell (Treg) were evaluated in patients with metastatic renal cell carcinoma (mRCC) enrolled in the REVOLUTION trial. EXPERIMENTAL DESIGN Fifty-seven mRCCs being treated with nivolumab, as at least second-line of therapy, and 62 healthy donors were longitudinally evaluated (0-1-3-6-12 months) for peripheral NKs and Tregs, phenotype, and function. Multivariable logistic regression was conducted to identify the independent predictors. The 0.632+ internal cross-validation was used to avoid overfitting. The best cutoff value based on a 3-month clinical response was applied to progression-free survival (PFS) and overall survival (OS). Kaplan-Meier curves for PFS and OS were produced. RESULTS At pretreatment, mRCCs displayed high frequency of NKp46+NKs, NKp30+NKs, KIR2DL1+NKs, KIR2DL2/DL3+NKs, and PD1+NKs with reduced NK degranulation as well as high frequency of Tregs, PD1+Tregs, Helios+Tregs, and ENTPD1+Tregs. Responder patients, identified as a clinical response after 3 months of treatment, presented at pretreatment significantly low CD3+, high KIR2DL2/DL3+NKs, high PD1+Tregs, and high Helios+Tregs. Upon multivariate analysis, only KIR2DL2/DL3NKs and Helios+Tregs held as independent predictors of nivolumab responsiveness. The KIR2DL2/DL3+NKs >35.3% identified patients with longer OS, whereas the Helios+Tregs >34.3% displayed significantly longer PFS. After 1-month of nivolumab, responder patients showed low CD3+, high NKs, KIR2DL2/DL3+NKs, and ICOS+Tregs. Among these subpopulations, CD3+ and KIR2DL2/DL3+NKs held as independent predictors of nivolumab efficacy. Low CD3+ (≤71%) was significantly associated with longer PFS, whereas high KIR2DL2/DL3+NKs (>23.3%) were associated with both PFS and OS. CONCLUSIONS Pretreatment evaluation of Helios+Tregs/KIR2DL2/DL3+NKs and 1-month posttreatment CD3+/ KIR2DL2/DL3+NKs will predict nivolumab response in mRCCs.
Collapse
Affiliation(s)
- Sara Santagata
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Anna Maria Trotta
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Crescenzo D’Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Maria Napolitano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Giuseppina Rea
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Marilena Di Napoli
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Caterina Ieranò
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Giuseppe Guardascione
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Elisabetta Coppola
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Christophe Caux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France.
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France.
| | - Bertrand Dubois
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France.
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France.
| | - Helen J. Boyle
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.
| | - Joan Carles
- Oncology Department, Val d’Hebron University, Barcelona, Spain.
| | - Sabrina Rossetti
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Rosa Azzaro
- Transfusion Medicine Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Florinda Feroce
- Department of Pathology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Mario Fordellone
- Unità di Statistica Medica Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Daniela Califano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Paolo Chiodini
- Unità di Statistica Medica Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.
| | - Sandro Pignata
- Uro-Gynecological Oncology, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
4
|
Liu X, Li W, Yi L, Wang J, Liu W, Cheng H, Ren S. CDK4/6 inhibitors dephosphorylate RNF26 to stabilize TSC1 and increase the sensitivity of ccRCC to mTOR inhibitors. Br J Cancer 2024; 131:444-456. [PMID: 38890443 PMCID: PMC11300639 DOI: 10.1038/s41416-024-02750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The combined use of CDK4/6 inhibitors and mTOR inhibitors has achieved some clinical success in ccRCC. Exploring the underlying mechanism of the CDK4/6 pathway in cancer cells and the drug interactions of CDK4/6 inhibitors in combination therapy could help identify new therapeutic strategies for ccRCC. Notably, CDK4/6 inhibitors inactivate the mTOR pathway by increasing the protein levels of TSC1, but the mechanism by which CDK4/6 inhibitors regulate TSC1 is still unclear. METHODS Mass spectrometry analysis, coimmunoprecipitation analysis, GST pull-down assays, immunofluorescence assays, Western blot analysis and RT‒qPCR analysis were applied to explore the relationships among CDK4, RNF26 and TSC1. Transwell assays, tube formation assays, CCK-8 assays, colony formation assays and xenograft assays were performed to examine the biological role of RNF26 in renal cancer cells.TCGA-KIRC dataset analysis and RT‒qPCR analysis were used to examine the pathways affected by RNF26 silencing. RESULTS CDK4/6 inhibitors stabilized TSC1 in cancer cells. We showed that CDK4 enhances the interaction between TSC1 and RNF26 and that RNF26 activates the mTOR signaling pathway in ccRCC, contributes to ccRCC progression and angiogenesis, and promotes tumorigenesis. We then found that RNF26 functions as an E3 ligase of TSC1 to regulate CDK4-induced TSC1. This finding suggested that RNF26 promotes ccRCC progression and angiogenesis to some extent by negatively regulating TSC1. CONCLUSION Our results revealed a novel CDK4/RNF26/TSC1 axis that regulates the anticancer efficacy of CDK4/6 inhibitors and mTOR inhibitors in ccRCC.
Collapse
Affiliation(s)
- Xinlin Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China
- Hunan Engineering Research Center of Smart and Precise Medicine, Changsha, Hunan, 410011, China
| | - Wei Li
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China
- Hunan Engineering Research Center of Smart and Precise Medicine, Changsha, Hunan, 410011, China
| | - Lu Yi
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China
- Hunan Engineering Research Center of Smart and Precise Medicine, Changsha, Hunan, 410011, China
| | - Jianxi Wang
- Department of Urology, The Third Hospital of Changsha, Changsha, Hunan, 410011, China
| | - Wentao Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Uro-Oncology Institute of Central South University, Changsha, Hunan, 410011, China.
- Hunan Engineering Research Center of Smart and Precise Medicine, Changsha, Hunan, 410011, China.
| | - Hongtao Cheng
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer. No.116 Zhuo Daoquan South Road, Wuhan, Hubei, 430079, China.
| | - Shangqing Ren
- Robotic Minimally Invasive Surgery Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
5
|
Wang X, Qian L, Qian Z, Wu Q, Cheng D, Wei J, Song L, Huang S, Chen X, Wang P, Weng G. Therapeutic options for different metastatic sites arising from renal cell carcinoma: A review. Medicine (Baltimore) 2024; 103:e38268. [PMID: 38788027 PMCID: PMC11124732 DOI: 10.1097/md.0000000000038268] [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: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Renal cell carcinoma (RCC) stands among the top 10 malignant neoplasms with the highest fatality rates. It exhibits pronounced heterogeneity and robust metastatic behavior. Patients with RCC may present with solitary or multiple metastatic lesions at various anatomical sites, and their prognoses are contingent upon the site of metastasis. When deliberating the optimal therapeutic approach for a patient, thorough evaluation of significant risk factors such as the feasibility of complete resection, the presence of oligometastases, and the patient's functional and physical condition is imperative. Recognizing the nuanced differences in RCC metastasis to distinct organs proves advantageous in contemplating potential treatment modalities aimed at optimizing survival outcomes. Moreover, discerning the metastatic site holds promise for enhancing risk stratification in individuals with metastatic RCC. This review summarizes the recent data pertaining to the current status of different RCC metastatic sites and elucidates their role in informing clinical management strategies across diverse metastatic locales of RCC.
Collapse
Affiliation(s)
- Xue Wang
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Lin Qian
- Department of Urologic Surgery, Ningbo Yinzhou No. 2 Hospital, Ningbo, China
| | - Zengxing Qian
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Qihang Wu
- Department of Clinical Laboratory, Ningbo Yinzhou No. 2 Hospital, Ningbo, China
| | - Dongying Cheng
- Department of community, Ningbo Yinzhou No. 3 Hospital, Ningbo, China
| | - Junjun Wei
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Lingmin Song
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Shuaihuai Huang
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Xiaodong Chen
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| | - Ping Wang
- Department of Clinical Laboratory, Ningbo Yinzhou No. 2 Hospital, Ningbo, China
| | - Guobin Weng
- Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, China
| |
Collapse
|
6
|
Baston C, Parosanu AI, Stanciu IM, Nitipir C. Metastatic Kidney Cancer: Does the Location of the Metastases Matter? Moving towards Personalized Therapy for Metastatic Renal Cell Carcinoma. Biomedicines 2024; 12:1111. [PMID: 38791072 PMCID: PMC11117570 DOI: 10.3390/biomedicines12051111] [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: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The management of renal cell carcinoma (RCC) has been revolutionized over the past two decades with several practice-changing treatments. Treatment for RCC often requires a multimodal approach: Local treatment, such as surgery or ablation, is typically recommended for patients with localized tumors, while stage IV cancers often require both local and systemic therapy. The treatment of advanced RCC heavily relies on immunotherapy and targeted therapy, which are highly contingent upon histological subtypes. Despite years of research on biomarkers for RCC, the standard of care is to choose systemic therapy based on the risk profile according to the International Metastatic RCC Database Consortium and Memorial Sloan Kettering Cancer Centre models. However, many questions still need to be answered. Should we consider metastatic sites when deciding on treatment options for metastatic RCC? How do we choose between dual immunotherapy and combinations of immunotherapy and tyrosine kinase inhibitors? This review article aims to answer these unresolved questions surrounding the concept of personalized medicine.
Collapse
Affiliation(s)
- Catalin Baston
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (C.B.); (I.-M.S.); (C.N.)
- Department of Urology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Andreea Ioana Parosanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (C.B.); (I.-M.S.); (C.N.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Ioana-Miruna Stanciu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (C.B.); (I.-M.S.); (C.N.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Cornelia Nitipir
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Sanitary Heroes Boulevard, 050474 Bucharest, Romania; (C.B.); (I.-M.S.); (C.N.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| |
Collapse
|
7
|
Pathmanathan S, Tariq A, Pearce A, Rhee H, Kyle S, Raveenthiran S, Wong D, McBean R, Marsh P, Goodman S, Dhiantravan N, Esler R, Dunglison N, Navaratnam A, Yaxley J, Thomas P, Pattison DA, Goh JC, Gan CL, Roberts MJ. Clinical impact of Prostate-Specific Membrane Antigen Positron Emission Tomography (PET) on intensification or deintensification of advanced renal cell carcinoma management. Eur J Nucl Med Mol Imaging 2023; 51:295-303. [PMID: 37592084 PMCID: PMC10684606 DOI: 10.1007/s00259-023-06380-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
PURPOSE There is an emerging role of the use of Prostate-Specific Membrane Antigen (PSMA) Positron Emission Tomography (PET) in renal cell carcinoma. Herein, we report our experience in use of PSMA PET in recurrent or metastatic renal cell carcinoma (RCC). METHODS A retrospective analysis of all patients who underwent PSMA PET for suspected recurrent or de-novo metastatic RCC between 2015 and 2020 at three institutions was performed. The primary outcome was change in management (intensification or de-intensification) following PSMA PET scan. Secondary outcomes included histopathological correlation of PSMA avid sites, comparison of sites of disease on PSMA PET to diagnostic CT and time to systemic treatment. RESULTS
Collapse
Affiliation(s)
- Shivanshan Pathmanathan
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Arsalan Tariq
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Adam Pearce
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Handoo Rhee
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Samuel Kyle
- Department of, Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Sheliyan Raveenthiran
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Rhiannon McBean
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Phillip Marsh
- Department of Diagnostic Radiology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Steven Goodman
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Nattakorn Dhiantravan
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Rachel Esler
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Nigel Dunglison
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Anojan Navaratnam
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - John Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Paul Thomas
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - David A Pattison
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Jeffrey C Goh
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Chun Loo Gan
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
8
|
Koç I, Kiratli H, Kapucu Y. Protein kinase inhibitors in the management of clear cell renal cell carcinoma metastatic to the iris. J Fr Ophtalmol 2023; 46:e365-e367. [PMID: 37612190 DOI: 10.1016/j.jfo.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 08/25/2023]
Affiliation(s)
- I Koç
- Ocular Oncology Service, Department of Ophthalmology, Hacettepe University School of Medicine, Sihhiye, 06100 Ankara, Turkey.
| | - H Kiratli
- Ocular Oncology Service, Department of Ophthalmology, Hacettepe University School of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Y Kapucu
- Ocular Oncology Service, Department of Ophthalmology, Hacettepe University School of Medicine, Sihhiye, 06100 Ankara, Turkey
| |
Collapse
|
9
|
Zhang Z, Cao C, Zhou CL, Li X, Miao C, Shen L, Singla RK, Lu X. Identification of a novel 5-methylcytosine-related signature for prognostic prediction of kidney renal papillary cell carcinoma and a Putative target for drug repurposing. Transl Oncol 2023; 36:101741. [PMID: 37523897 PMCID: PMC10400932 DOI: 10.1016/j.tranon.2023.101741] [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: 07/26/2022] [Revised: 03/14/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Many studies have demonstrated the crucial roles of 5-methylcytosine (m5C) RNA methylation in cancer pathogenesis. METHODS Two datasets, including TCGA-KIRP and ICGC, and related clinical information were downloaded, where the expression of 13 m5C regulators was examined. We applied LASSO regression to construct a multi-m5C-regulator-based signature in the TCGA cohort, which was further validated using the ICGC cohort. Univariate and multivariate Cox regressions were applied to evaluate the independent prognostic value of our model. The differences in biological functions and immune characterizations between high and low-risk groups divided based on the risk scores were also investigated via multiple approaches, such as enrichment analyses, mutation mining, and immune scoring. Finally, the sensitivities of commonly used targeted drugs were tested, and the connectivity MAP (cMAP) was utilized to screen potentially effective molecules for patients in the high-risk group. Experimental validation was done following qPCR tests in Caki-2 and HK-2 cell lines. RESULTS 3 m5C regulators, including ALYREF, DNMT3B and YBX1, were involved in our model. Survival analysis revealed a worse prognosis for patients in the high-risk group. Cox regression results indicated our model's superior predictive performance compared to single-factor prognostic evaluation. Functional enrichment analyses indicated a higher mutation frequency and poorer tumor microenvironment of patients in the high-risk group. qPCR-based results revealed that ALYREF, DNMT3B, and YBX1 were significantly up-regulated in Caki-2 cell lines compared with HK-2 cell lines. Molecules like BRD-K72451865, Levosimendan, and BRD-K03515135 were advised by cMAP for patients in the high-risk group. CONCLUSION Our study presented a novel predictive model for KIRP prognosis. Furthermore, the results of our analysis provide new insights for investigating m5C events in KIRP pathogenesis.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Chunhua Cao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China; Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, China
| | - Chun-Li Zhou
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, China
| | - Xilong Li
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Changhong Miao
- Department of Anesthesiology, Cancer Hospital of Fudan University, Shanghai 200032, China
| | - Li Shen
- Department of Biomedical Engineering, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India.
| | - Xihua Lu
- Department of Anesthesiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
10
|
Séraudie I, Pillet C, Cesana B, Bazelle P, Jeanneret F, Evrard B, Chalmel F, Bouzit A, Battail C, Long JA, Descotes JL, Cochet C, Filhol O. A new scaffold-free tumoroid model provides a robust preclinical tool to investigate invasion and drug response in Renal Cell Carcinoma. Cell Death Dis 2023; 14:622. [PMID: 37736770 PMCID: PMC10517165 DOI: 10.1038/s41419-023-06133-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Clear cell Renal Cell Carcinoma (ccRCC) is one of the most prevalent kidney cancers, which is often asymptomatic and thus discovered at a metastatic state (mRCC). mRCC are highly heterogeneous tumors composed of subclonal populations that lead to poor treatment response rate. Several recent works explored the potential of ccRCC tumoroids culture derived from patients. However, these models were produced following a scaffold-based method using collagen I or Matrigel that exhibit lot variability and whose complexity could induce treatment response modifications and phenotypic alterations. Following the observation that ccRCC tumoroids can create their own niche by secreting extracellular matrix components, we developed the first scaffold-free tumoroid model of ccRCC tumors. Tumoroids from mice as well as from human tumors were generated with high success rate (≥90%) using a magnetic suspension method and standard culture media. Immunofluorescence analysis revealed their self-organization capacities to maintain multiple tumor-resident cell types, including endothelial progenitor cells. Transcriptomic analysis showed the reproducibility of the method highlighting that the majority of gene expression patterns was conserved in tumoroids compared to their matching tumor tissue. Moreover, this model enables to evaluate drug effects and invasiveness of renal cancer cells in a 3D context, providing a robust preclinical tool for drug screening and biomarker assessment in line with alternative ex vivo methods like tumor tissue slice culture or in vivo xenograft models.
Collapse
Grants
- EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- INSERM, CEA, Ligue Comité de l’Isère, University Grenoble Alpes, Centre Hospitalier Universitaire de Grenoble-Alpes (CHUGA), Groupement des Entreprises Françaises dans la Lutte contre le Cancer (GEFLUC)
- CEA, UGA
- CEA, Inserm
- UGA, Inserm, CEA
- CHU, Ligue Comité de l’Isère
- Inserm, Ligue Comité de l’Isère
Collapse
Affiliation(s)
- Irinka Séraudie
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Catherine Pillet
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Beatrice Cesana
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Pauline Bazelle
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Florian Jeanneret
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UA 13, F-38000, Grenoble, France
| | - Bertrand Evrard
- University Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Frédéric Chalmel
- University Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Assilah Bouzit
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Christophe Battail
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UA 13, F-38000, Grenoble, France
| | - Jean-Alexandre Long
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Jean Luc Descotes
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Claude Cochet
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Odile Filhol
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France.
| |
Collapse
|
11
|
Krawczyk K, Śladowska K, Holko P, Kawalec P. Comparative safety of tyrosine kinase inhibitors in the treatment of metastatic renal cell carcinoma: a systematic review and network meta-analysis. Front Pharmacol 2023; 14:1223929. [PMID: 37745049 PMCID: PMC10512702 DOI: 10.3389/fphar.2023.1223929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/11/2023] [Indexed: 09/26/2023] Open
Abstract
Objective: This study aimed to compare the safety profile of tyrosine kinase inhibitors (TKIs) approved for use as monotherapy or combination therapy for the first-line treatment of adult patients with metastatic clear cell renal cell carcinoma (RCC). Methods: A systematic review with frequentist network meta-analysis (NMA) was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We included randomized controlled trials (RCTs) investigating the use of: cabozantinib, pazopanib, sorafenib, sunitinib, tivozanib, cabozantinib + nivolumab, lenvatinib + pembrolizumab, axitinib + avelumab, and axitinib + pembrolizumab in previously untreated adult patients with metastatic clear cell RCC. Eligible studies were identified by two reviewers in MEDLINE (via PubMed), EMBASE, and Cochrane Library. The risk of bias for RCTs was assessed using the Cochrane Collaboration tool. The P score was used to determine the treatment ranking. The mean probability of an event along with the relative measures of the NMA was considered with the treatment rankings. Results: A total of 13 RCTs were included in the systematic review and NMA. Sorafenib and tivozanib used as monotherapy were the best treatment options. Sorafenib achieved the highest P score for treatment discontinuation due to adverse events (AEs), fatigue, nausea, vomiting of any grade, and hypertension of any grade or grade ≥3. Tivozanib achieved the highest P score for AEs, grade ≥3 AEs, dose modifications due to AEs, and grade ≥3 diarrhea. Sunitinib was the best treatment option in terms of diarrhea and dysphonia of any grade, while cabozantinib, pazopanib, and axitinib + pembrolizumab-in terms of grade ≥3 fatigue, nausea, and vomiting. TKIs used in combination were shown to have a poorer safety profile than those used as monotherapy. Lenvatinib + pembrolizumab was considered the worst option in terms of any AEs, grade ≥3 AEs, treatment discontinuation due to AEs, dose modifications due to AEs, fatigue of any grade, nausea, vomiting, and grade ≥3 nausea. Axitinib + avelumab was the worst treatment option in terms of dysphonia, grade ≥3 diarrhea, and hypertension, while cabozantinib + nivolumab was the worst option in terms of grade ≥3 vomiting. Interestingly, among the other safety endpoints, cabozantinib monotherapy had the lowest P score for diarrhea and hypertension of any grade. Conclusion: The general safety profile, including common AEs, is better when TKIs are used as monotherapy vs. in combination with immunological agents. To confirm these findings, further research is needed, including large RCTs.
Collapse
Affiliation(s)
- Kinga Krawczyk
- Faculty of Health Sciences, Institute of Public Health, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna Śladowska
- Department of Nutrition and Drug Research, Faculty of Health Sciences, Institute of Public Health, Jagiellonian University Medical College, Krakow, Poland
| | - Przemysław Holko
- Department of Nutrition and Drug Research, Faculty of Health Sciences, Institute of Public Health, Jagiellonian University Medical College, Krakow, Poland
| | - Paweł Kawalec
- Department of Nutrition and Drug Research, Faculty of Health Sciences, Institute of Public Health, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
12
|
Piao XM, Byun YJ, Zheng CM, Song SJ, Kang HW, Kim WT, Yun SJ. A New Treatment Landscape for RCC: Association of the Human Microbiome with Improved Outcomes in RCC. Cancers (Basel) 2023; 15:cancers15030935. [PMID: 36765892 PMCID: PMC9913391 DOI: 10.3390/cancers15030935] [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: 01/09/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Microbes play different roles in metabolism, local or systemic inflammation, and immunity, and the human microbiome in tumor microenvironment (TME) is important for modulating the response to immunotherapy in cancer patients. Renal cell carcinoma (RCC) is an immunogenic tumor, and immunotherapy is the backbone of its treatment. Correlations between the microbiome and responsiveness to immune checkpoint inhibitors have been reported. This review summarizes the recent therapeutic strategies for RCC and the effects of TME on the systemic therapy of RCC. The current understanding and advances in microbiome research and the relationship between the microbiome and the response to immunotherapy for RCC are also discussed. Improving our understanding of the role of the microbiome in RCC treatment will facilitate the development of microbiome targeting therapies to modify the tumor microbiome and improve treatment outcomes.
Collapse
Affiliation(s)
- Xuan-Mei Piao
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Young Joon Byun
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Chuang-Ming Zheng
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sun Jin Song
- Department of Emergency, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Ho Won Kang
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Won Tae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
| | - Seok Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju 28644, Republic of Korea
- Correspondence: ; Tel.: +82-43-269-6142
| |
Collapse
|
13
|
Zhang Z, Lin S, Yin J, Yu W, Xu C. CircRNF220 plays a pathogenic role to facilitate cell progression of AML in vitro via sponging miR-330-5p to induce upregulation of SOX4. Histol Histopathol 2022; 37:1019-1030. [PMID: 35611720 DOI: 10.14670/hh-18-472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) are a specific family of non-coding RNAs (ncRNAs) with important function in disease progression. This research is performed to study circRNA Ring Finger Protein 220 (circRNF220) in acute myeloid leukemia (AML). METHODS CircRNF220, microRNA-330-5p (miR-330-5p) and sex-determining region Y-related high-mobility group box 4 (SOX4) were measured via quantitative real-time polymerase chain reaction (qRT-PCR). 3-(4, 5-dimethylthiazol-2-y1)-2, 5- diphenyl tetrazolium bromide (MTT) and EdU assays were used to assess cell proliferation. Cell cycle and apoptosis were detected using flow cytometry. Cell invasion was determined by transwell assay. Glycolytic metabolism was assessed by glucose consumption and lactate production. The target interaction was implemented via dual-luciferase reporter and RNA pull-down assays. SOX4 protein detection was conducted by western blot. RESULTS Expression detection identified that circRNF220 was overexpressed in AML. In vitro experiments showed that silence of circRNF220 promoted cell apoptosis but impeded proliferation, cell cycle progression, invasion and glycolytic metabolism in AML cells. Target analysis indicated that circRNF220 directly targeted miR-330-5p, and the effects of si-circRNF220 were abrogated by miR-330-5p inhibitor. Moreover, circRNF220 targeted miR-330-5p to increase the expression of SOX4 and SOX4 promoted cell progression of AML. CONCLUSION All these findings revealed that circRNF220 contributed to AML cell development in vitro via upregulating SOX4 expression by targeting miR-330-5p.
Collapse
Affiliation(s)
- Zewen Zhang
- Division of Hematology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Shujun Lin
- Division of Nephrology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Jun Yin
- Department of Clinical Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Wenjun Yu
- Division of Hematology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chengwei Xu
- Blood Purification Room, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
14
|
Pei L, Dong C, Wang Y, Lv X, Jia G, Zhang A. Circular RNA circSDHC (hsa_circ_0015004) regulates tumor growth and angiogenesis via regulating centrosomal protein 55 expression in renal cell carcinoma. Histol Histopathol 2022; 37:971-983. [PMID: 35506422 DOI: 10.14670/hh-18-467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is the main aggressive subtype of kidney cancer. Circular RNAs have been shown to exert critical roles in RCC. However, little is known about the regulatory mechanism of hsa_circ_0015004 (circSDHC) in RCC. METHODS 35 patients with RCC were recruited in the research. Expression changes of circSDHC were determined by real-time quantitative polymerase chain reaction (RT-qPCR). The effects of circSDHC inhibition on cell proliferation, apoptosis, angiogenesis, migration, and invasion were analyzed. The regulation mechanism of circSDHC was surveyed by bioinformatics analysis. The effect of circSDHC on tumorigenesis was validated by xenograft assay. RESULTS We observed an observable elevation in circSDHC expression in RCC tissues and cell lines. Functionally, circSDHC silencing decreased xenograft tumor growth and induced RCC cell apoptosis, repressed RCC cell proliferation, angiogenesis, migration, and invasion in vitro. Mechanically, circSDHC modulated centrosomal protein 55 (CEP55) expression by functioning as a miR-130a-3p sponge. Also, miR-130a-3p silencing offset circSDHC knockdown-mediated impacts on malignant phenotypes and angiogenesis of RCC cells. Furthermore, exogenetic expression of CEP55 counteracted miR-130a-3p overexpression-mediated effects on malignant phenotypes and angiogenesis of RCC cells. CONCLUSION Silencing of circSDHC restrained cell malignant phenotypes and angiogenesis via reducing CEP55 expression by releasing miR-130a-3p in RCC, providing a new mechanism for understanding the progression of RCC.
Collapse
Affiliation(s)
- Long Pei
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Chunhui Dong
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Yanchao Wang
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Xianqiang Lv
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Gaopei Jia
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China
| | - Aili Zhang
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
15
|
Mao W, Wang K, Zhang W, Chen S, Xie J, Zheng Z, Li X, Zhang N, Zhang Y, Zhang H, Peng B, Yao X, Che J, Zheng J, Chen M, Li W. Transfection with Plasmid-Encoding lncRNA-SLERCC nanoparticle-mediated delivery suppressed tumor progression in renal cell carcinoma. J Exp Clin Cancer Res 2022; 41:252. [PMID: 35986402 PMCID: PMC9389749 DOI: 10.1186/s13046-022-02467-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background The accumulating evidence confirms that long non-coding RNAs (lncRNAs) play a critical regulatory role in the progression of renal cell carcinoma (RCC). But, the application of lncRNAs in gene therapy remains scarce. Here, we investigated the efficacy of a delivery system by introducing the plasmid-encoding tumor suppressor lncRNA-SLERCC (SLERCC) in RCC cells. Methods We performed lncRNAs expression profiling in paired cancer and normal tissues through microarray and validated in our clinical data and TCGA dataset. The Plasmid-SLERCC@PDA@MUC12 nanoparticles (PSPM-NPs) were tested in vivo and in vitro, including cellular uptake, entry, CCK-8 assay, tumor growth inhibition, histological assessment, and safety evaluations. Furthermore, experiments with nude mice xenografts model were performed to evaluate the therapeutic effect of PSPM-NPs nanotherapeutic system specific to the SLERCC. Results We found that the expression of SLERCC was downregulated in RCC tissues, and exogenous upregulation of SLERCC could suppress metastasis of RCC cells. Furthermore, high expression DNMT3A was recruited at the SLERCC promoter, which induced aberrant hypermethylation, eventually leading to downregulation of SLERCC expression in RCC. Mechanistically, SLERCC could directly bind to UPF1 and exert tumor-suppressive effects through the Wnt/β-catenin signaling pathway, thereby inhibiting progression and metastasis in RCC. Subsequently, the PSPM-NPs nanotherapeutic system can effectively inhibit the growth of RCC metastases in vivo. Conclusions Our findings suggested that SLERCC is a promising therapeutic target and that plasmid-encapsulated nanomaterials targeting transmembrane metastasis markers may open a new avenue for the treatment in RCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02467-2.
Collapse
|
16
|
Harada KI, Miyake H, Furukawa J, Fujimoto N, Fujisawa M. Comprehensive assessments of immuno-oncology drug-based combination therapies as first-line treatment for advanced renal cell carcinoma. Int J Urol 2022; 29:816-822. [PMID: 35636920 DOI: 10.1111/iju.14922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022]
Abstract
Over the last decade, there have been substantial progress in the field of systemic therapy for advanced renal cell carcinoma. Through the transition from treatment with cytokines to molecular-targeted agents, and currently to immuno-oncology drugs, the prognostic outcomes of patients with advanced renal cell carcinoma have been markedly improved. In particular, based on the promising outcomes of recently conducted pivotal randomized clinical trials, immuno-oncology drug-based combination therapy by either dual immune checkpoint inhibition or combined inhibition of an immune checkpoint and tyrosine kinase, is currently regarded as a standard of care for treatment-naïve advanced renal cell carcinoma patients. However, insufficient data are available with respect to the selection of optimal systemic therapies for advanced renal cell carcinoma in the first-line setting due to the lack of a head-to-head comparison between approved immuno-oncology drug-based combination therapies. In this review, therefore, we summarize interesting findings associated with first-line combination therapies for advanced renal cell carcinoma obtained from both randomized clinical trials and real-world clinical practices, in order to present useful guidance to help make treatment decisions for patients with treatment-naïve advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Ken-Ichi Harada
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hideaki Miyake
- Department of Urology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Junya Furukawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
17
|
The RBPJ/DAPK3/UBE3A signaling axis induces PBRM1 degradation to modulate the sensitivity of renal cell carcinoma to CDK4/6 inhibitors. Cell Death Dis 2022; 13:295. [PMID: 35368029 PMCID: PMC8976838 DOI: 10.1038/s41419-022-04760-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 11/09/2022]
Abstract
AbstractRenal cell carcinoma (RCC) is a kind of malignant tumor originating from the renal tubular epithelium. Approximately 30% of patients with renal cancer are found to have metastasis when first diagnosed. Exploring other effective treatment methods in addition to surgery is an urgent need in the research field of renal cell carcinoma. Polybromo 1 (PBRM1) is the second most mutated gene in RCC, with a mutation rate of ~40%. Notably, the posttranscriptional modification of PBRM1 in RCC is unclear. In this study, we performed unbiased mass spectrometry of PBRM1 and identified ubiquitin-protein ligase E3A (UBE3A), an extensively studied E3 ligase that can bind with PBRM1 and regulate the stability of PBRM1 in renal cancer cells. We further found that RBPJ/DAPK3 modulated the E3 ligase activity of UBE3A by interfering with the PKA phosphorylation of UBE3A. Finally, we demonstrated that the RBPJ/DAPK3/UBE3A/PBRM1/p21 axis contributed to the sensitivity of renal cancer cells to CDK4/6 inhibitors. In addition, in combination with RBPJ inhibitors, CDK4/6 inhibitors showed synergistically enhanced effects on renal cancer cells. In summary, we not only revealed a novel RBPJ/DAPK3/UBE3A/PBRM1/p21 signaling axis but also identified a combination strategy for overcoming the resistance of renal cancer cells to CDK4/6 inhibitors.
Collapse
|
18
|
Hu J, Chen Y, Gao L, Ge C, Xie X, Lei P, Zhang Y, Liang P. A Novel Pyroptosis-Related Gene Signature for Predicting Prognosis in Kidney Renal Papillary Cell Carcinoma. Front Genet 2022; 13:851384. [PMID: 35401700 PMCID: PMC8984942 DOI: 10.3389/fgene.2022.851384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis is defined as an inflammatory form of programmed cell death. Increasing studies have demonstrated that pyroptosis is closely related to tumor development and antitumor process. However, the role of pyroptosis in kidney renal papillary cell carcinoma (KIRP) remains obscure. In this study, we analyzed the expression of 52 pyroptosis-related genes (PRGs) in KIRP, of which 20 differentially expressed PRGs were identified between tumor and normal tissues. Consensus clustering analysis based on these PRGs was used to divided patients into two clusters, from which a significant difference in survival was found (p = 0.0041). The prognostic risk model based on six PRGs (CASP8, CASP9, CHMP2A, GPX4, IL6, and IRF1) was built using univariate Cox regression and LASSO–Cox regression analysis, with good performance in predicting one-, three-, and five-year overall survival. Kaplan–Meier survival analysis showed that the high-risk group had a poor survival outcome (p < 0.001) and risk score was an independent prognostic factor (HR: 2.655, 95% CI 1.192–5.911, p = 0.016). Immune profiling revealed differences in immune cell infiltration between the two groups, and the infiltration of M2 macrophages was significantly upregulated in the tumor immune microenvironment, implying that tumor immunity participated in the KIRP progression. Finally, we identified two hub genes in tumor tissues (IL6 and CASP9), which were validated in vitro. In conclusion, we conducted a comprehensive analysis of PRGs in KIRP and tried to provide a pyroptosis-related signature for predicting the prognosis.
Collapse
Affiliation(s)
- Jian Hu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajun Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Gao
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengguo Ge
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodu Xie
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pan Lei
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanfeng Zhang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peihe Liang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Peihe Liang,
| |
Collapse
|
19
|
Fontes-Sousa M, Magalhães H, Oliveira A, Carneiro F, dos Reis FP, Madeira PS, Meireles S. Reviewing Treatment Options for Advanced Renal Cell Carcinoma: Is There Still a Place for Tyrosine Kinase Inhibitor (TKI) Monotherapy? Adv Ther 2022; 39:1107-1125. [PMID: 35025061 PMCID: PMC8756748 DOI: 10.1007/s12325-021-02007-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023]
Abstract
Renal cell carcinoma (RCC) comprises a highly heterogeneous group of kidney tumours built upon distinct genetic- and epigenetic-driven mechanisms and molecular pathways. Therefore, responsiveness to treatment is considerably variable across patients, adding an extra layer of complexity to the already challenging therapeutic decision process. The last decade brought an unprecedented shift in the medical approach to advanced or metastatic RCC; in fact, immunotherapy-based combinations have significantly transformed the therapeutic arsenal and clinical outcomes of these patients. These strategies were quickly adopted by international guidelines committees as the new standards of care. However, this enhanced efficacy comes at the expense of tolerability, with a predictable negative impact on patients' quality of life. Moreover, subgroup and post hoc analyses of the major clinical trials have shown that not all patients benefit equally from these innovative approaches. In this context, a group of experts on kidney cancer met and discussed the state of the art in the field, with a special emphasis on the appropriateness of using monotherapy with an anti-angiogenesis tyrosine kinase inhibitor (TKI) to treat specific subgroups of patients with RCC. This article reviews the main topics that were considered to be pertinent for that discussion and establishes the profile of patients for whom TKI monotherapy remains a sensible frontline option by avoiding overtreatment and an unnecessary exposure to treatment-related toxicity.
Collapse
Affiliation(s)
| | - Helena Magalhães
- Hospital Pedro Hispano (Unidade Local de Saúde de Matosinhos), Rua Dr. Eduardo Torres, 4464-513 Senhora da Hora, Portugal
| | - Alicia Oliveira
- Hospital do Espírito Santo de Évora, Largo do Sr. da Pobreza, 7000-811 Évora, Portugal
| | - Filipa Carneiro
- Medical oncology department, Instituto Português de Oncologia Do Porto, Rua Dr. António Bernardino de Almeida 865, 4200-072 Porto, Portugal
| | - Filipa Palma dos Reis
- Hospital de Santo António Dos Capuchos (Centro Hospitalar Universitário de Lisboa Central), Alameda Santo António Dos Capuchos, 1169-050 Lisbon, Portugal
| | - Pedro Silvestre Madeira
- Instituto Português de Oncologia de Coimbra, Av. Prof. Dr. Bissaya Barreto No. 98, 3000-075 Coimbra, Portugal
| | - Sara Meireles
- Hospital de São João (Centro Hospitalar Universitário de São João), Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
20
|
Albiges L, Tannir NM, Burotto M, McDermott D, Plimack ER, Barthélémy P, Porta C, Powles T, Donskov F, George S, Kollmannsberger CK, Gurney H, Grimm MO, Tomita Y, Castellano D, Rini BI, Choueiri TK, Leung D, Saggi SS, Lee CW, McHenry MB, Motzer RJ. First-line Nivolumab plus Ipilimumab Versus Sunitinib in Patients Without Nephrectomy and With an Evaluable Primary Renal Tumor in the CheckMate 214 Trial. Eur Urol 2022; 81:266-271. [PMID: 34750035 DOI: 10.1016/j.eururo.2021.10.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/06/2021] [Indexed: 01/11/2023]
Abstract
We present an exploratory post hoc analysis from the phase 3 CheckMate 214 trial of first-line nivolumab plus ipilimumab (NIVO+IPI) versus sunitinib in a subgroup of 108 patients with advanced renal cell carcinoma (aRCC) without prior nephrectomy and with an evaluable primary tumor, a population under-represented in clinical trials. Patients with clear cell aRCC were randomized to NIVO+IPI every 3 wk for four doses followed by NIVO monotherapy, or sunitinib every day for 4 wk (6-wk cycle). Overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and primary tumor shrinkage were assessed. PFS and ORR were assessed per independent radiology review committee using RECIST version 1.1. With minimum study follow-up of 4 yr for intent-to-treat patients, OS favored NIVO+IPI (n = 53) over sunitinib (n = 55; hazard ratio 0.63, 95% confidence interval 0.40-1.0) among patients without prior nephrectomy. ORR was higher (34% vs 15%; p = 0.0041) and median duration of response was longer with NIVO+IPI versus sunitinib (20.5 vs 14.1 mo); the best overall response was partial response in either arm. A ≥30% reduction in the diameter of intact target renal tumors was achieved in 35% of patients with NIVO+IPI versus 20% with sunitinib. Safety was consistent with the global study population. In conclusion, in patients with aRCC without prior nephrectomy and with an evaluable primary tumor, NIVO+IPI showed survival benefits and renal tumor reduction versus sunitinib. This trial is registered at ClinicalTrials.gov as NCT02231749. PATIENT SUMMARY: In an exploratory analysis of a large global trial (CheckMate 214), we observed positive outcomes (both survival and tumor response to treatment) with nivolumab plus ipilimumab over sunitinib in a subgroup of patients with advanced kidney cancer who did not undergo removal of their primary kidney tumor. This subset of patients represents a population that has not been studied in clinical trials and for whom outcomes with new immunotherapy combination regimens are not yet known. We conclude that treatment with nivolumab plus ipilimumab offers these patients a survival benefit versus sunitinib, consistent with that observed in the overall study, as well as a notable kidney tumor reduction.
Collapse
Affiliation(s)
- Laurence Albiges
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - David McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Elizabeth R Plimack
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Philippe Barthélémy
- Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | | | - Thomas Powles
- Department of Urology, Barts Cancer Institute, Queen Mary University of London, Royal Free NHS Trust, London, UK
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Saby George
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Howard Gurney
- Department of Medical Oncology, Westmead Hospital and Macquarie University, Sydney, Australia
| | | | - Yoshihiko Tomita
- Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Daniel Castellano
- Medical Oncology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Brian I Rini
- Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Leung
- Department of Imaging, Bristol Myers Squibb, Princeton, NJ, USA
| | | | - Chung-Wei Lee
- Department of Clinical Trials, Bristol Myers Squibb, Princeton, NJ, USA
| | - M Brent McHenry
- Department of Biostatistics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
21
|
Maestroni U, Gasparro D, Ziglioli F, Guarino G, Campobasso D. Metastatic Clear Cell Renal Cell Carcinoma: The Great Pretender and the Great Dilemma. World J Oncol 2021; 12:178-182. [PMID: 34804281 PMCID: PMC8577602 DOI: 10.14740/wjon1406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/30/2021] [Indexed: 01/13/2023] Open
Abstract
Metastatic renal cell carcinoma (mRCC) may present with a wide range of clinical pictures. Reportedly, paraneoplastic syndromes are the first sign in 20% of cases and only 15% of cases show the classic triad (flank pain, gross hematuria, and palpable abdominal mass) at presentation. The remaining cases present with signs and symptoms related to the site of distant metastases. These data may explain the reason why about 20-30% of patients are metastatic at presentation. We report the case of a 63-year-old woman who came to our attention for lower back pain. After imaging studies, we detected a left kidney mass of 86 × 61 × 79 mm, multiple right pulmonary nodules and six bone lesions. She underwent left radical nephrectomy. After 1 month, she developed signs of spinal cord compression with neurological deficits and she underwent emergency spinal decompression. In order to allow complete motor recovery, the subsequent stereotactic body radiation therapy was not performed, and she is currently taking combination immunotherapy regimens. Management of mRCC is in a continuous evolution due to availability of new target therapies and the possibility of a multimodal approach with surgical, focal and radiotherapy treatments. However, the ideal treatment algorithm is yet to come. This is why mRCC diagnosis and management are still challenging for the clinicians.
Collapse
Affiliation(s)
- Umberto Maestroni
- Department of Urology, University Hospital of Parma/Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, Parma 43126, Italy
| | - Donatello Gasparro
- Medical Oncology Unit, University Hospital of Parma/Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, Parma 43126, Italy
| | - Francesco Ziglioli
- Department of Urology, University Hospital of Parma/Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, Parma 43126, Italy
| | - Giulio Guarino
- Department of Urology, University Hospital of Parma/Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, Parma 43126, Italy
| | - Davide Campobasso
- Department of Urology, University Hospital of Parma/Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, Parma 43126, Italy
| |
Collapse
|
22
|
Huang J, Cai W, Cai B, Kong W, Zhai W, Zhang J, Chen Y, Chen S, Bai Y, Huang Y, Xue W. Comprehensive Genomic Landscape in Chinese Clear Cell Renal Cell Carcinoma Patients. Front Oncol 2021; 11:697219. [PMID: 34568025 PMCID: PMC8459629 DOI: 10.3389/fonc.2021.697219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/18/2021] [Indexed: 02/03/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC). The genomic landscape in Chinese ccRCC needs to be elucidated. Herein, we investigated the molecular features of Chinese ccRCC patients. Genomic profiling of DNA was performed through next-generation sequencing (NGS) in Chinese patients with ccRCC between January 2017 and March 2020. Clinical information including age, gender, and tumor histology was collected. Immunohistochemistry (IHC) staining for PD-L1 expression was performed using PD-L1 IHC 22C3 pharmDx assay or Ventana PD-L1 SP263 assay. Data analyses were performed using R 3.6.1. A total of 880 Chinese ccRCC patients who have undergone NGS were included in this study. The most common somatic alterations were detected in VHL (59.7%), PBRM1 (18.0%), SETD2 (12.2%), BAP1 (10.2%), and TP53 (9.4%). Compared with The Cancer Genome Atlas (TCGA) database, a higher mutation frequency of VHL (59.7% vs. 50.0%, p < 0.001) and TP53 (9.4% vs. 3.5%, p < 0.001) and a lower mutation frequency of PBRM1 (18.0% vs. 31.0%, p < 0.001) were found in the Chinese cohort. Of the 460 patients who were evaluated for PD-L1 expression, 139 (30.2%) had positive PD-L1 expression. The median tumor mutational burden (TMB) value was 4.5 muts/Mb (range, 0–46.0). Five (0.7%) patients were identified as microsatellite instability-high (MSI-H). Furthermore, 52 (5.9%) patients were identified to carry pathogenic or likely pathogenic germline mutations in 22 cancer predisposition genes. This is the first large-scale comprehensive genomic analysis for Chinese ccRCC patients, and these results might provide a better understanding of molecular features in Chinese ccRCC patients, which can lead to an improvement in the personalized treatment for these patients.
Collapse
Affiliation(s)
- Jiwei Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Cai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Urology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Biao Cai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Kong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiqing Chen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yuezong Bai
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|