1
|
Aljabali AAA, Tambuwala MM, El-Tanani M, Hassan SS, Lundstrom K, Mishra V, Mishra Y, Hromić-Jahjefendić A, Redwan EM, Uversky VN. A comprehensive review of PRAME and BAP1 in melanoma: Genomic instability and immunotherapy targets. Cell Signal 2024; 124:111434. [PMID: 39326690 DOI: 10.1016/j.cellsig.2024.111434] [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: 08/24/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
In a thorough review of the literature, the complex roles of PRAME (preferentially expressed Antigen of Melanoma) and BAP1 (BRCA1-associated protein 1) have been investigated in uveal melanoma (UM) and cutaneous melanoma. High PRAME expression in UM is associated with poor outcomes and correlated with extraocular extension and chromosome 8q alterations. BAP1 mutations in the UM indicate genomic instability and a poor prognosis. Combining PRAME and BAP1 immunohistochemical staining facilitates effective risk stratification. Mechanistically, both genes are associated with genomic instability, making them promising targets for cancer immunotherapy. Hypomethylation of PRAME, specifically in its promoter regions, is critical for UM progression and contributes to epigenetic reprogramming. Additionally, miR-211 regulation is crucial in melanoma and has therapeutic potential. The way PRAME changes signaling pathways provides clues about the cause of cancer due to genomic instability related to modifications in DNA repair. Inhibition of poly(ADP-ribose) polymerase-1 (PARP-1) and PARP-2 in cells expressing PRAME could lead to potential therapeutic applications. Pathway enrichment analysis underscores the significance of PRAME and BAP1 in melanoma pathogenesis.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan.
| | - Murtaza M Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates.
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates.
| | - Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur, 721140, West Bengal, India.
| | | | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, 21934 Alexandria, Egypt.
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
2
|
Guberovic I, Frezza C. Functional implications of fumarate-induced cysteine succination. Trends Biochem Sci 2024; 49:775-790. [PMID: 38876954 DOI: 10.1016/j.tibs.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
Mutations in metabolic enzymes are associated with hereditary and sporadic forms of cancer. For example, loss-of-function mutations affecting fumarate hydratase (FH), the tricarboxylic acid (TCA) cycle enzyme, result in the accumulation of millimolar levels of fumarate that cause an aggressive form of kidney cancer. A distinct feature of fumarate is its ability to spontaneously react with thiol groups of cysteines in a chemical reaction termed succination. Although succination of a few proteins has been causally implicated in the molecular features of FH-deficient cancers, the stoichiometry, wider functional consequences, and contribution of succination to disease development remain largely unexplored. We discuss the functional implications of fumarate-induced succination in FH-deficient cells, the available methodologies, and the current challenges in studying this post-translational modification.
Collapse
Affiliation(s)
- Iva Guberovic
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christian Frezza
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
3
|
Li L, Hossain SM, Eccles MR. The Role of the PAX Genes in Renal Cell Carcinoma. Int J Mol Sci 2024; 25:6730. [PMID: 38928435 PMCID: PMC11203709 DOI: 10.3390/ijms25126730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Renal cell carcinoma (RCC) is a significant oncological challenge due to its heterogeneous nature and limited treatment options. The PAX developmental gene family encodes nine highly conserved transcription factors that play crucial roles in embryonic development and organogenesis, which have been implicated in the occurrence and development of RCC. This review explores the molecular landscape of RCC, with a specific focus on the role of the PAX gene family in RCC tumorigenesis and disease progression. Of the various RCC subtypes, clear cell renal cell carcinoma (ccRCC) is the most prevalent, characterized by the loss of the von Hippel-Lindau (VHL) tumor suppressor gene. Here, we review the published literature on the expression patterns and functional implications of PAX genes, particularly PAX2 and PAX8, in the three most common RCC subtypes, including ccRCC, papillary RCC (PRCC), and chromophobe RCC (ChRCC). Further, we review the interactions and potential biological mechanisms involving PAX genes and VHL loss in driving the pathogenesis of RCC, including the key signaling pathways mediated by VHL in ccRCC and associated mechanisms implicating PAX. Lastly, concurrent with our update regarding PAX gene research in RCC, we review and comment on the targeting of PAX towards the development of novel RCC therapies.
Collapse
Affiliation(s)
- Lei Li
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
| | - Sultana Mehbuba Hossain
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand; (L.L.); (S.M.H.)
- Maurice Wilkins Centre for Molecular Biodiscovery, Level 2, 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
4
|
Seema Mustafa, Jansen CS, Jani Y, Evans S, Zhuang TZ, Brown J, Nazha B, Master V, Bilen MA. The Evolving Landscape of Biomarkers for Immune Checkpoint Blockade in Genitourinary Cancers. Biomark Insights 2024; 19:11772719241254179. [PMID: 38827239 PMCID: PMC11143877 DOI: 10.1177/11772719241254179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 04/24/2024] [Indexed: 06/04/2024] Open
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have been approved for treatment of genitourinary malignancies and have revolutionized the treatment landscape of these tumors. However, despite the remarkable success of these therapies in some GU malignancies, many patients' tumors do not respond to these therapies, and others may experience significant side effects, such as immune-related adverse events (iRAEs). Accordingly, biomarkers and improved prognostic tools are critically needed to help predict which patients will respond to ICI, predict and mitigate risk of developing immune-related adverse events, and inform personalized choice of therapy for each patient. Ongoing clinical and preclinical studies continue to provide an increasingly robust understanding of the mechanisms of the response to immunotherapy, which continue to inform biomarker development and validation. Herein, we provide a comprehensive review of biomarkers of the response to immunotherapy in GU tumors and their role in selection of therapy and disease monitoring.
Collapse
Affiliation(s)
- Seema Mustafa
- Emory University School of Medicine, Atlanta, GA, USA
| | - Caroline S Jansen
- Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Sean Evans
- Emory University School of Medicine, Atlanta, GA, USA
| | - Tony Z Zhuang
- Emory University School of Medicine, Atlanta, GA, USA
| | - Jacqueline Brown
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bassel Nazha
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Viraj Master
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehmet Asim Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
5
|
Hwang J, Bang S, Choi MH, Hong SH, Kim SW, Lee HE, Yang JH, Park US, Choi YJ. Discovery and Validation of Survival-Specific Genes in Papillary Renal Cell Carcinoma Using a Customized Next-Generation Sequencing Gene Panel. Cancers (Basel) 2024; 16:2006. [PMID: 38893126 PMCID: PMC11171119 DOI: 10.3390/cancers16112006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
PURPOSE Papillary renal cell carcinoma (PRCC), the second most common kidney cancer, is morphologically, genetically, and molecularly heterogeneous with diverse clinical manifestations. Genetic variations of PRCC and their association with survival are not yet well-understood. This study aimed to identify and validate survival-specific genes in PRCC and explore their clinical utility. MATERIALS AND METHODS Using machine learning, 293 patients from the Cancer Genome Atlas-Kidney Renal Papillary Cell Carcinoma (TCGA-KIRP) database were analyzed to derive genes associated with survival. To validate these genes, DNAs were extracted from the tissues of 60 Korean PRCC patients. Next generation sequencing was conducted using a customized PRCC gene panel of 202 genes, including 171 survival-specific genes. Kaplan-Meier and Log-rank tests were used for survival analysis. Fisher's exact test was performed to assess the clinical utility of variant genes. RESULTS A total of 40 survival-specific genes were identified in the TCGA-KIRP database through machine learning and statistical analysis. Of them, 10 (BAP1, BRAF, CFDP1, EGFR, ITM2B, JAK1, NODAL, PCSK2, SPATA13, and SYT5) were validated in the Korean-KIRP database. Among these survival gene signatures, three genes (BAP1, PCSK2, and SPATA13) showed survival specificity in both overall survival (OS) (p = 0.00004, p = 1.38 × 10-7, and p = 0.026, respectively) and disease-free survival (DFS) (p = 0.00002, p = 1.21 × 10-7, and p = 0.036, respectively). Notably, the PCSK2 mutation demonstrated survival specificity uniquely in both the TCGA-KIRP (OS: p = 0.010 and DFS: p = 0.301) and Korean-KIRP (OS: p = 1.38 × 10-7 and DFS: p = 1.21 × 10-7) databases. CONCLUSIONS We discovered and verified genes specific for the survival of PRCC patients in the TCGA-KIRP and Korean-KIRP databases. The survival gene signature, including PCSK2 commonly obtained from the 40 gene signature of TCGA and the 10 gene signature of the Korean database, is expected to provide insight into predicting the survival of PRCC patients and developing new treatment.
Collapse
Affiliation(s)
- Jia Hwang
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (J.H.); (H.E.L.)
| | - Seokhwan Bang
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.B.); (S.-H.H.); (S.W.K.)
| | - Moon Hyung Choi
- Department of Radiology, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03312, Republic of Korea;
| | - Sung-Hoo Hong
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.B.); (S.-H.H.); (S.W.K.)
| | - Sae Woong Kim
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.B.); (S.-H.H.); (S.W.K.)
| | - Hye Eun Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (J.H.); (H.E.L.)
| | - Ji Hoon Yang
- Department of Computer Science and Engineering, Sogang University, Seoul 04107, Republic of Korea; (J.H.Y.); (U.S.P.)
| | - Un Sang Park
- Department of Computer Science and Engineering, Sogang University, Seoul 04107, Republic of Korea; (J.H.Y.); (U.S.P.)
| | - Yeong Jin Choi
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (J.H.); (H.E.L.)
| |
Collapse
|
6
|
Kiatprungvech N, Sangkum P, Malinee R, Sommaluan S, Korkiatsakul V, Worawichawong S, Rerkamnuaychoke B, Kongruang A, Aeesoa S, Lertsithichai P, Kijvikai K, Kongchareonsombat W, Siriboonpiputtana T. Genetic study of the CDKN2A and CDKN2B genes in renal cell carcinoma patients. Pract Lab Med 2024; 40:e00410. [PMID: 38867760 PMCID: PMC11167386 DOI: 10.1016/j.plabm.2024.e00410] [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: 03/28/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024] Open
Abstract
Objectives While recent studies have demonstrated several genetic alterations are associated with pathogenesis of RCC, the significance of cyclin-dependent kinase inhibitor 2A (CDKN2A) and cyclin-dependent kinase inhibitor 2B (CDKN2B) in tumorigenesis of RCC is less clear. We investigate the distribution of CDKN2A and CDKN2B mutations in patients with RCC and analyze the impact of CDKN2A and CDKN2B mutations on RCC. Methods A pathological examination was conducted using thirty fresh renal tissue samples with renal masses that had undergone partial or radical nephrectomy. Multiplex ligation-dependent probe amplification (MLPA) was used to detect genetic aberrations of CDKN2A and CDKN2B in genomic DNA isolated from samples. Subsequently, CDKN2A and CDKN2B mutations were confirmed using chromosomal microarray technique. Results Twenty-one patients were diagnosed with RCC, eight with benign diseases, including angiomyolipoma (AML) and oncocytoma, and one with mucinous adenocarcinoma of renal pelvis. Two of twenty-one patients (9.5 %) with clear-cell RCC were positive for CDKN2A and CDKN2B gene deletions. Interestingly, patients with CDKN2A and CDKN2B mutations were associated with sarcomatoid patterns of RCC (2 out of 4, 50 %). In contrast, no CDKN2A or CDKN2B deletions were detected in samples from benign renal tumors, papillary RCC, or other kidney cancers. Conclusions This study demonstrated the potential use of CDKN2A and CDKN2B as biomarkers for the prognostic and molecular classification of renal cancer. CDKN2A and CDKN2B mutations may be associated with RCC development and sarcomatoid changes. Further research is needed to understand the underlying molecular mechanisms of CDKN2A and CDKN2B in the pathogenesis of RCC.
Collapse
Affiliation(s)
- Nattaradee Kiatprungvech
- Division of Urology, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Premsant Sangkum
- Division of Urology, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rozita Malinee
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suchada Sommaluan
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Veerawat Korkiatsakul
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suchin Worawichawong
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Budsaba Rerkamnuaychoke
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adcharee Kongruang
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suraida Aeesoa
- Division of Urology, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Panuwat Lertsithichai
- Division of Breast and Endocrine, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kittinut Kijvikai
- Division of Urology, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wisoot Kongchareonsombat
- Division of Urology, Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
7
|
Yoshida T, Sakai K, Kaibori M, Ishida M, Tanaka S, Kubo S, Nakai T, De Velasco MA, Matsushima H, Tsuta K, Sekimoto M, Nishio K. Downregulated expression of PBRM1 in sarcomatoid hepatocellular carcinoma. Oncol Lett 2024; 27:124. [PMID: 38348389 PMCID: PMC10859838 DOI: 10.3892/ol.2024.14257] [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: 06/08/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
Sarcomatoid hepatocellular carcinoma (SHCC) is a rare and highly lethal subtype of HCC. The present study aimed to explore the unique markers of SHCC using whole gene expression analysis. Subsequently, gene expression analysis was performed using five sarcomatoid and seven carcinomatoid components of seven tissues from patients with SHCC. The results demonstrated a significant downregulation of polybromo 1 (PBRM1) gene expression in the sarcomatoid components. Immunohistochemical staining also indicated a decreased expression of PBRM1 in the sarcomatoid components. Moreover, gene ontology enrichment analysis revealed that most of the 336 differentially expressed genes between the sarcomatoid and carcinomatoid components were involved in functions associated with DNA replication and histone methylation, which was consistent with the loss of function of PBRM1 which encodes Switch/sucrose-non-fermentable chromatin remodeling complex protein. Therefore, the results of the present study suggested that PBRM1 may be a candidate biomarker for the evaluation of SHCC.
Collapse
Affiliation(s)
- Terufumi Yoshida
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-8686, Japan
| | - Shogo Tanaka
- Department of Hepato-Biliary-Pancreatic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Shoji Kubo
- Department of Hepato-Biliary-Pancreatic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Takuya Nakai
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan
| | - Marco A. De Velasco
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan
| | - Hideyuki Matsushima
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Koji Tsuta
- Department of Pathology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan
| |
Collapse
|
8
|
Jia L, Cowell LG, Kapur P. Understanding Factors that Influence Prognosis and Response to Therapy in Clear Cell Renal Cell Carcinoma. Adv Anat Pathol 2024; 31:96-104. [PMID: 38179997 DOI: 10.1097/pap.0000000000000428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
In this review, we highlight and contextualize emerging morphologic prognostic and predictive factors in renal cell carcinoma. We focus on clear cell renal cell carcinoma (ccRCC), the most common histologic subtype. Our understanding of the molecular characterization of ccRCC has dramatically improved in the last decade. Herein, we highlight how these discoveries have laid the foundation for new approaches to prognosis and therapeutic decision-making for patients with ccRCC. We explore the clinical relevance of common mutations, established gene expression signatures, intratumoral heterogeneity, sarcomatoid/rhabdoid morphology and PD-L1 expression, and discuss their impact on predicting response to therapy.
Collapse
Affiliation(s)
| | - Lindsay G Cowell
- Peter O'Donnell School of Public Health
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| | - Payal Kapur
- Department of Pathology
- Department of Urology, University of Texas Southwestern Medical Center
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| |
Collapse
|
9
|
Alchoueiry M, Cornejo K, Henske EP. Kidney cancer: Links between hereditary syndromes and sporadic tumorigenesis. Semin Diagn Pathol 2024; 41:1-7. [PMID: 38008653 DOI: 10.1053/j.semdp.2023.11.002] [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: 09/14/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023]
Abstract
Multiple hereditary syndromes predispose to kidney cancer, including Von Hippel-Lindau syndrome, BAP1-Tumor Predisposition Syndrome, Hereditary Papillary Renal Cell Carcinoma, Tuberous Sclerosis Complex, Birt-Hogg-Dubé syndrome, Hereditary Paraganglioma-Pheochromocytoma Syndrome, Fumarate Hydratase Tumor Predisposition Syndrome, and Cowden syndrome. In some cases, mutations in the genes that cause hereditary kidney cancer are tightly linked to similar histologic features in sporadic RCC. For example, clear cell RCC occurs in the hereditary syndrome VHL, and sporadic ccRCC usually has inactivation of the VHL gene. In contrast, mutations in FLCN, the causative gene for Birt-Hogg-Dube syndrome, are rarely found in sporadic RCC. Here, we focus on the genes and pathways that link hereditary and sporadic RCC.
Collapse
Affiliation(s)
- Michel Alchoueiry
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristine Cornejo
- Pathology Department, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Yan M, Wang J, Wang H, Zhou J, Qi H, Naji Y, Zhao L, Tang Y, Dai Y. Knockdown of NR3C1 inhibits the proliferation and migration of clear cell renal cell carcinoma through activating endoplasmic reticulum stress-mitophagy. J Transl Med 2023; 21:701. [PMID: 37807060 PMCID: PMC10560440 DOI: 10.1186/s12967-023-04560-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/22/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is closely associated with steroid hormones and their receptors affected by lipid metabolism. Recently, there has been growing interest in the carcinogenic role of NR3C1, the sole gene responsible for encoding glucocorticoid receptor. However, the specific role of NR3C1 in ccRCC remains unclear. The present study was thus developed to explore the underlying mechanism of NR3C1's carcinogenic effects in ccRCC. METHODS Expression of NR3C1 was verified by various tumor databases and assessed using RT-qPCR and western blot. Stable transfected cell lines of ccRCC with NR3C1 knockdown were constructed, and a range of in vitro and in vivo experiments were performed to examine the effects of NR3C1 on ccRCC proliferation and migration. Transcriptomics and lipidomics sequencing were then conducted on ACHN cells, which were divided into control and sh-NR3C1 group. Finally, the sequencing results were validated using transmission electron microscopy, mitochondrial membrane potential assay, immunofluorescence co-localization, cell immunofluorescent staining, and Western blot. The rescue experiments were designed to investigate the relationship between endoplasmic reticulum stress (ER stress) and mitophagy in ccRCC cells after NR3C1 knockdown, as well as the regulation of their intrinsic signaling pathways. RESULTS The expression of NR3C1 in ccRCC cells and tissues was significantly elevated. The sh-NR3C1 group, which had lower levels of NR3C1, exhibited a lower proliferation and migration capacity of ccRCC than that of the control group (P < 0.05). Then, lipidomic and transcriptomic sequencing showed that lipid metabolism disorders, ER stress, and mitophagy genes were enriched in the sh-NR3C1 group. Finally, compared to the control group, ER stress and mitophagy were observed in the sh-NR3C1 group, while the expression of ATF6, CHOP, PINK1, and BNIP3 was also up-regulated (P < 0.05). Furthermore, Ceapin-A7, an inhibitor of ATF6, significantly down-regulated the expression of PINK1 and BNIP3 (P < 0.05), and significantly increased the proliferation and migration of ccRCC cells (P < 0.05). CONCLUSIONS This study confirms that knockdown of NR3C1 activates ER stress and induces mitophagy through the ATF6-PINK1/BNIP3 pathway, resulting in reduced proliferation and migration of ccRCC. These findings indicate potential novel targets for clinical treatment of ccRCC.
Collapse
Affiliation(s)
- Minbo Yan
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Jinhua Wang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Haojie Wang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Jun Zhou
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Hao Qi
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Yaser Naji
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Liangyu Zhao
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China.
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China.
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
11
|
Kapur P, Rajaram S, Brugarolas J. The expanding role of BAP1 in clear cell renal cell carcinoma. Hum Pathol 2023; 133:22-31. [PMID: 35932824 PMCID: PMC9898467 DOI: 10.1016/j.humpath.2022.07.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Mutations drive renal cell carcinoma biology and tumor growth. The BRCA1-associated protein-1 (BAP1) gene is frequently mutated in clear cell renal cell carcinoma (ccRCC) and has emerged as a prognostic and putative predictive biomarker. In this review, we discuss the role of BAP1 as a signature event of a subtype of ccRCC marked by aggressiveness, inflammation, and possibly a heightened response to immunotherapy.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA.
| | - Satwik Rajaram
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA; Department of Internal Medicine (Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
12
|
Druggable Biomarkers Altered in Clear Cell Renal Cell Carcinoma: Strategy for the Development of Mechanism-Based Combination Therapy. Int J Mol Sci 2023; 24:ijms24020902. [PMID: 36674417 PMCID: PMC9864911 DOI: 10.3390/ijms24020902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Targeted therapeutics made significant advances in the treatment of patients with advanced clear cell renal cell carcinoma (ccRCC). Resistance and serious adverse events associated with standard therapy of patients with advanced ccRCC highlight the need to identify alternative 'druggable' targets to those currently under clinical development. Although the Von Hippel-Lindau (VHL) and Polybromo1 (PBRM1) tumor-suppressor genes are the two most frequently mutated genes and represent the hallmark of the ccRCC phenotype, stable expression of hypoxia-inducible factor-1α/2α (HIFs), microRNAs-210 and -155 (miRS), transforming growth factor-beta (TGF-ß), nuclear factor erythroid 2-related factor 2 (Nrf2), and thymidine phosphorylase (TP) are targets overexpressed in the majority of ccRCC tumors. Collectively, these altered biomarkers are highly interactive and are considered master regulators of processes implicated in increased tumor angiogenesis, metastasis, drug resistance, and immune evasion. In recognition of the therapeutic potential of the indicated biomarkers, considerable efforts are underway to develop therapeutically effective and selective inhibitors of individual targets. It was demonstrated that HIFS, miRS, Nrf2, and TGF-ß are targeted by a defined dose and schedule of a specific type of selenium-containing molecules, seleno-L-methionine (SLM) and methylselenocystein (MSC). Collectively, the demonstrated pleiotropic effects of selenium were associated with the normalization of tumor vasculature, and enhanced drug delivery and distribution to tumor tissue, resulting in enhanced efficacy of multiple chemotherapeutic drugs and biologically targeted molecules. Higher selenium doses than those used in clinical prevention trials inhibit multiple targets altered in ccRCC tumors, which could offer the potential for the development of a new and novel therapeutic modality for cancer patients with similar selenium target expression. Better understanding of the underlying mechanisms of selenium modulation of specific targets altered in ccRCC could potentially have a significant impact on the development of a more efficacious and selective mechanism-based combination for the treatment of patients with cancer.
Collapse
|
13
|
GRAMMATIKAKI STAMATIKI, KATIFELIS HECTOR, FAROOQI AMMADAHMAD, STRAVODIMOS KONSTANTINOS, KARAMOUZIS MICHALISV, SOULIOTIS KYRIAKOS, VARVARAS DIMITRIOS, GAZOULI MARIA. An Overview of Epigenetics in Clear Cell Renal Cell Carcinoma. In Vivo 2023; 37:1-10. [PMID: 36593023 PMCID: PMC9843790 DOI: 10.21873/invivo.13049] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023]
Abstract
Renal cell carcinoma (RCC) represents a heterogenous group of cancers with complex genetic background and histological varieties, which require various clinical therapies. Clear cell RCC represents the most common form of RCC that accounts for 3 out of 4 RCC cases. Screening methods for RCC lack sensitivity and specificity, and thus biomarkers that will allow early diagnosis are crucial. The impact of epigenetics in the development and progression of cancer, including RCC, is significant. Noncoding RNAs, histone modifications and DNA methylation represent fundamental epigenetic mechanisms and have been proved to be promising biomarkers. MicroRNAs have advantageous properties that facilitate early diagnosis of RCC, while their expression profiles have been assessed in renal cancer samples (tissue, blood, and urine). Current literature reports the up-regulation of mir122, mir1271 and mir15b in RCC specimens, which induces cell proliferation via FOXP-1 and PTEN genes. However, it should be noted that conflicting results are found in urine and serum patient samples. Moreover, promoters of at least 200 genes are methylated in renal cancers leading to epigenetic dysregulation. In this review, we analyze the vast plethora of studies that have evaluated the role of epigenetic mechanisms in RCC patients and their clinical importance.
Collapse
Affiliation(s)
- STAMATIKI GRAMMATIKAKI
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - HECTOR KATIFELIS
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - KONSTANTINOS STRAVODIMOS
- 1st Department of Urology, National & Kapodistrian University of Athens, Laiko Hospital, Athens, Greece
| | - MICHALIS V. KARAMOUZIS
- Molecular Oncology Unit, Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - KYRIAKOS SOULIOTIS
- School of Social and Education Policy, University of Peloponnese, Corinth, Greece,Health Policy Institute, Athens, Greece
| | - DIMITRIOS VARVARAS
- Health Policy Institute, Athens, Greece,Tiberia Hospital-GMV Care & Research, Rome, Italy
| | - MARIA GAZOULI
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Yang J, Wang K, Yang Z. Treatment strategies for clear cell renal cell carcinoma: Past, present and future. Front Oncol 2023; 13:1133832. [PMID: 37025584 PMCID: PMC10070676 DOI: 10.3389/fonc.2023.1133832] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent histological subtype of kidney cancer, which is prone to metastasis, recurrence, and resistance to radiotherapy and chemotherapy. The burden it places on human health due to its refractory nature and rising incidence rate is substantial. Researchers have recently determined the ccRCC risk factors and optimized the clinical therapy based on the disease's underlying molecular mechanisms. In this paper, we review the established clinical therapies and novel potential therapeutic approaches for ccRCC, and we support the importance of investigating novel therapeutic options in the context of combining established therapies as a research hotspot, with the goal of providing diversified therapeutic options that promise to address the issue of drug resistance, with a view to the early realization of precision medicine and individualized treatment.
Collapse
Affiliation(s)
- Junwei Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhichun Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
- *Correspondence: Zhichun Yang,
| |
Collapse
|
15
|
PBRM1 and BAP1: novel genetic mutations in malignant transformation of craniopharyngioma-a case report. Brain Tumor Pathol 2023; 40:40-44. [PMID: 36369599 DOI: 10.1007/s10014-022-00444-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022]
Abstract
Malignant craniopharyngioma is especially rare, so the causes and genetic mutations associated with the malignant transformation have not been explained in detail. We investigated the molecular genetic characteristics of malignant transformation in craniopharyngioma. A 53-year-old man with a history of adamantinomatous craniopharyngioma presented with complaints of subcutaneous swelling. Magnetic resonance imaging showed a less enhanced intradural supra-sellar lesion and a heterogeneously well-enhanced extradural invasive lesion infiltrating the dura mater, brain, frontal bone, and subcutaneous tissue. Histopathological examination of the recurrent tumor revealed typical findings of both craniopharyngioma (intradural supra-sellar lesion) and malignant transformation, such as marked nuclear atypia with mitosis (invasive extradural lesion), which were not present in the primary tumor. A genetic panel test with the Oncopanel system was performed to investigate the genetic mutations responsible for the malignant transformation. Four genetic mutations were identified: CTNNB1 c.C98T, TP53 p.C135fs*35(PLS = 3 UPD/LOH), PBRM1 p.R1000*(PLS = 3 UPD/LOH), and BAP1 p.L650fs*5(PLS = 3 UPD/LOH). Sanger sequencing showed CTNNB1 in both the intradural supra-sellar and extradural invasive lesions, but TP53, PBRM1, and BAP1 only in the extradural invasive lesion. The genetic mutations of PBRM1 and BAP1 may be genetic factors in the malignant transformation of adamantinomatous craniopharyngioma.
Collapse
|
16
|
Wang Q, Tang H, Luo X, Chen J, Zhang X, Li X, Li Y, Chen Y, Xu Y, Han S. Immune-Associated Gene Signatures Serve as a Promising Biomarker of Immunotherapeutic Prognosis for Renal Clear Cell Carcinoma. Front Immunol 2022; 13:890150. [PMID: 35686121 PMCID: PMC9171405 DOI: 10.3389/fimmu.2022.890150] [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: 03/05/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
As the most common type of renal cell carcinoma (RCC), the renal clear cell carcinoma (ccRCC) is highly malignant and insensitive to chemotherapy or radiotherapy. Although systemic immunotherapies have been successfully applied to ccRCC in recent years, screening for patients who can benefit most from these therapies is still essential and challenging due to immunological heterogeneity of ccRCC patients. To this end, we implemented a series of deep investigation on the expression and clinic data of ccRCC from The Cancer Genome Atlas (TCGA) International Consortium for Cancer Genomics (ICGC). We identified a total of 946 immune-related genes that were differentially expressed. Among them, five independent genes, including SHC1, WNT5A, NRP1, TGFA, and IL4R, were significantly associated with survival and used to construct the immune-related prognostic differential gene signature (IRPDGs). Then the ccRCC patients were categorized into high-risk and low-risk subgroups based on the median risk score of the IRPDGs. IRPDGs subgroups displays distinct genomic and immunological characteristics. Known immunotherapy-related genes show different mutation burden, wherein the mutation rate of VHL was higher than 40% in the two IRPDGs subgroups, and SETD2 and BAP1 mutations differed most between two groups with higher frequency in the high-risk subgroup. Moreover, IRPDGs subgroups had different abundance in tumor-infiltrating immune cells (TIICs) with distinct immunotherapy efficacy. Plasma cells, regulatory cells (Tregs), follicular helper T cells (Tfh), and M0 macrophages were enriched in the high-risk group with a higher tumor immune dysfunction and rejection (TIDE) score. In contrast, the low-risk group had abundant M1 macrophages, mast cell resting and dendritic cell resting infiltrates with lower TIDE score and benefited more from immune checkpoint inhibitors (ICI) treatment. Compared with other biomarkers, such as TIDE and tumor inflammatory signatures (TIS), IRPDGs demonstrated to be a better biomarker for assessing the prognosis of ccRCC and the efficacy of ICI treatment with the promise in screening precise patients for specific immunotherapies.
Collapse
Affiliation(s)
- Qi Wang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanmin Tang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuehui Luo
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Chen
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Li
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuesen Li
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuetong Chen
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yungang Xu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Suxia Han
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Ördög N, Borsos BN, Majoros H, Ujfaludi Z, Pankotai-Bodó G, Bankó S, Sükösd F, Kuthi L, Pankotai T. The clinical significance of epigenetic and RNAPII variabilities occurring in clear cell renal cell carcinoma as a potential prognostic marker. Transl Oncol 2022; 20:101420. [PMID: 35417813 PMCID: PMC9018449 DOI: 10.1016/j.tranon.2022.101420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/02/2022] [Accepted: 04/05/2022] [Indexed: 12/14/2022] Open
Abstract
30 ccRCC patients were used, to follow the epigenetic changes (γH2A.X, H3K4me3 and H3K9me3) and the alterations in the level of RNA polymerase II (RNAPII). The variabilities between the tumorous and non-tumorous parts of the tissue were measured by image analysis in which we monitored 30 cells from different positions of either the tumorous or the non-tumorous part of the tissue sections. These markers were classified to predict patient outcomes based on their individual cellular background. These results also support that detection of any alteration in the level of H3K4me3, H3K9me3, and γH2AX can account valuable information for presuming the progression of ccRCC and the clinical benefits to select the most efficient personalized therapy.
Patients diagnosed with clear cell renal cell carcinoma (ccRCC) have poor prognosis for recurrence and approximately 30–40% of them will later develop metastases. For this reason, the appropriate diagnosis and the more detailed molecular characterisation of the primary tumour, including its susceptibility to metastasis, are crucial to select the proper adjuvant therapy by which the most prosperous outcome can be achieved. Nowadays, clinicopathological variables are used for classification of the tumours. Apart from these, molecular biomarkers are also necessary to improve risk classification, which would be the most beneficial amongst modern adjuvant therapies. As a potential molecular biomarker, to follow the transcriptional kinetics in ccRCC patients (n=30), we analysed epigenetic changes (γH2A.X, H3K4me3, and H3K9me3) and the alterations in the level of RNA polymerase II (RNAPII) by immunohistochemical staining on dissected tissue sections. The variabilities between the tumorous and non-tumorous parts of the tissue were detected using quantitative image analysis by monitoring 30 cells from different positions of either the tumorous or the non-tumorous part of the tissue sections. Data obtained from the analyses were used to identify potential prognostic features and to associate them with the progression. These markers might have a value to predict patient outcomes based on their individual cellular background. These results also support that detection of any alteration in the level of H3K4me3, H3K9me3, and γH2A.X can account for valuable information for presuming the progression of ccRCC and the clinical benefits to select the most efficient personalised therapy.
Collapse
|
18
|
Antonios JP, Yalcin K, Darbinyan A, Koo A, Hong CS, DiLuna M, Erson-Omay Z. Biallelic inactivation of PBRM1 as a molecular driver in a rare pineoblastoma case: illustrative case. JOURNAL OF NEUROSURGERY: CASE LESSONS 2022; 3:CASE2213. [PMID: 36303510 PMCID: PMC9379698 DOI: 10.3171/case2213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pineoblastomas are a rare and aggressive pediatric neuroectodermal tumor subtype. Because of their rarity, pineoblastomas are still poorly understood, and there is little research delineating their molecular development and underlying genetic phenotype. Recent multiomic studies in pineoblastomas and pineal parenchymal tumors identified four clinically and biologically relevant consensus groups driven by signaling/processing pathways; however, molecular level alterations leading to these pathway changes are yet to be discovered, hence the importance of individually profiling every case of this rare tumor type. OBSERVATIONS The authors present the comprehensive somatic genomic profiling of a patient with pineoblastoma presenting with the loss of protein polybromo-1 (PBRM1) as a candidate genomic driver. Loss of PBRM1, a tumor suppressor, has been reported as a driver event in various cancer types, including renal cell carcinoma, bladder carcinoma, and meningiomas with papillary features. LESSONS This is the first report presenting biallelic loss of PBRM1 as a candidate molecular driver in relation to pineoblastoma.
Collapse
|
19
|
Bell HN, Kumar-Sinha C, Mannan R, Zakalik D, Zhang Y, Mehra R, Jagtap D, Dhanasekaran SM, Vaishampayan U. Pathogenic ATM and BAP1 germline mutations in a case of early-onset, familial sarcomatoid renal cancer. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006203. [PMID: 35483881 PMCID: PMC9059789 DOI: 10.1101/mcs.a006203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022] Open
Abstract
Metastatic renal cell carcinoma (RCC) remains an incurable malignancy, despite recent advances in systemic therapies. Genetic syndromes associated with kidney cancer account for only 5%-8% of all diagnosed kidney malignancies, and genetic predispositions to kidney cancer predisposition are still being studied. Genomic testing for kidney cancer is useful for disease molecular subtyping but provides minimal therapeutic information. Understanding how aberrations drive RCC development and how their contextual influences, such as chromosome loss, genome instability, and DNA methylation changes, may alter therapeutic response is of importance. We report the case of a 36-yr-old female with aggressive, metastatic RCC and a significant family history of cancer, including RCC. This patient harbors a novel, pathogenic, germline ATM mutation along with a rare germline variant of unknown significance in the BAP1 gene. In addition, somatic loss of heterozygosity (LOH) in BAP1 and ATM genes, somatic mutation and LOH in the VHL gene, copy losses in Chromosomes 9p and 14, and genome instability are also noted in the tumor, potentially dictating this patient's aggressive clinical course. Further investigation is warranted to evaluate the association of ATM and BAP1 germline mutations with increased risk of RCC and if these mutations should lead to enhanced and early screening.
Collapse
Affiliation(s)
- Hannah N Bell
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Chandan Kumar-Sinha
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Rahul Mannan
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Dana Zakalik
- Oakland University/Beaumont Hospital, Rochester, Michigan 48309, USA
| | - Yuping Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Rohit Mehra
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Deepa Jagtap
- Oakland University/Beaumont Hospital, Rochester, Michigan 48309, USA
| | - Saravana M Dhanasekaran
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ulka Vaishampayan
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
20
|
LZTS2, a Novel and Independent Prognostic Biomarker for Clear Cell Renal Cell Carcinoma. Pathol Res Pract 2022; 232:153831. [DOI: 10.1016/j.prp.2022.153831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 11/18/2022]
|
21
|
Gulati S, Previtera M, Lara PN. BRCA1-Associated Protein 1 (BAP-1) as a Prognostic and Predictive Biomarker in Clear Cell Renal Cell Carcinoma: A Systematic Review. KIDNEY CANCER 2021. [DOI: 10.3233/kca-210006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The gene that encodes BRCA1-associated protein 1 (BAP1) has been reported to be dysregulated in several human cancers such as uveal melanoma, malignant pleural mesothelioma, hepatocellular carcinoma, thymic epithelial tumors, and clear-cell renal cell carcinoma (ccRCC). The gene is located on the human chromosome 3p21.3, encoding a deubiquitinase and acts as a classic two-hit tumor suppressor gene. BAP1 predominantly resides in the nucleus, where it interacts with several chromatin-associated factors, as well as regulates calcium signaling in the cytoplasm. As newer therapies continue to evolve for the management of RCC, it is important to understand the role of BAP1 mutation as a prognostic and predictive biomarker. OBJECTIVE: We aimed to systematically evaluate the role of BAP1 mutations in patients with RCC in terms of its impact on prognosis and its role as a predictive biomarker. METHODS: Following PRISMA guidelines, we performed a systematic literature search using PubMed and Embase through March 2021. Titles and abstracts were screened to identify articles for full-text and then a descriptive review was performed. RESULTS: A total of 490 articles were initially identified. Ultimately 71 articles that met our inclusion criteria published between 2012–2021 were included in the analysis. Data were extracted and organized to reflect the role of BAP1 alterations as a marker of prognosis as well as a marker of response to treatments, such as mTOR inhibitors, VEGF tyrosine kinase inhibitors, and immune checkpoint inhibitors. CONCLUSIONS: Alterations in BAP1 appear to be uniformly associated with poor prognosis in patients with RCC. Knowledge gaps remain with regard to the predictive relevance of BAP1 alterations, especially in the context of immunotherapy. Prospective studies are required to more precisely ascertain the predictive value of BAP1 alterations in RCC.
Collapse
Affiliation(s)
- Shuchi Gulati
- Department of Medicine, Division of Hematology and Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Melissa Previtera
- Academic & Research Services Specialist, Donald C. Harrison Health Sciences Library, University of Cincinnati Libraries, Cincinnati, OH, USA
| | - Primo N. Lara
- Department of Internal Medicine, Division of Hematology and Oncology, UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
22
|
Ahluwalia P, Mondal AK, Sahajpal NS, Rojiani MV, Kolhe R. Gene signatures with therapeutic value: emerging perspective for personalized immunotherapy in renal cancer. Immunotherapy 2021; 13:1535-1547. [PMID: 34753298 DOI: 10.2217/imt-2021-0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Renal cancer is one of the deadliest urogenital diseases. In recent years, the advent of immunotherapy has led to significant improvement in the management of patients with renal cancer. Although cancer immunotherapy and its combinations had benefited numerous patients, several challenges need to be addressed. Apart from the high costs of treatment, the lack of predictive biomarkers and toxic side-effects have impeded its wider applicability. To address these issues, new biomarkers are required to predict responsiveness and design personalized treatment strategies. Recent advances in the field of single-cell sequencing and multi-dimensional spatial transcriptomics have identified clinically relevant subtypes of renal cancer. Furthermore, there is emerging potential for gene signatures based on immune cells, non-coding RNAs, and pathways such as metabolism and RNA modification. In this review article, we have discussed recent progress in the identification of gene signatures with predictive and prognostic potential in renal cancer.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Ashis K Mondal
- Department of Pathology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Nikhil S Sahajpal
- Department of Pathology, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Mumtaz V Rojiani
- Department of Pharmacology, Penn State University College of Medicine, PA 17033, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, GA 30912, USA
| |
Collapse
|
23
|
Racaniello GF, Laquintana V, Vergnaud J, Lopedota A, Cutrignelli A, Lopalco A, Leonetti F, Franco M, Fiume M, Pontrelli P, Gesualdo L, Fattal E, Denora N. Development of purified glycogen derivatives as siRNA nanovectors. Int J Pharm 2021; 608:121128. [PMID: 34560204 DOI: 10.1016/j.ijpharm.2021.121128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 10/20/2022]
Abstract
Purified Glycogen (PG) is a highly hyper branched carbohydrate, characterized by high water solubility and very moderate increase in viscosity. The dendrimeric structure of PG, appropriately functionalized, makes it an alternative to current synthetic gene delivery agents. The present study explores the preparation of purified glycogen polycationic derivatives (PGPDs), developed and characterized starting from a single step reaction between PG and N,N-dialkylamino alkyl halides. Subsequently PGPDs were used for the complexation of a model siRNA nucleic acid, a transfection reagent siRNA and a fluorescein-labelled dsRNA oligomer. PGPDs-siRNA complexes were fully characterized by agarose gel electrophoresis and their efficacy was assessed by both confocal microscopy and transfection assays on breast and renal cancer cells. Results proved that PGPDs-siRNA complexes were efficient and not cytotoxic, maintaining their spherical and dendrimeric structure and, particularly, were able to effectively transfect the target cells by releasing the siRNA.
Collapse
Affiliation(s)
| | - Valentino Laquintana
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Juliette Vergnaud
- Institut Galien Paris-Saclay, UMR CNRS 8612, Université Paris Saclay, Châtenay-Malabry, Paris, France
| | - Angela Lopedota
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Annalisa Cutrignelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Antonio Lopalco
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Francesco Leonetti
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Massimo Franco
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Mauro Fiume
- Department of Emergency and Organ Transplantation, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Paola Pontrelli
- Department of Emergency and Organ Transplantation, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy
| | - Elias Fattal
- Institut Galien Paris-Saclay, UMR CNRS 8612, Université Paris Saclay, Châtenay-Malabry, Paris, France
| | - Nunzio Denora
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "A. Moro", Orabona, St. 4, 70125 Bari, Italy.
| |
Collapse
|
24
|
Huang H, Zhu L, Huang C, Dong Y, Fan L, Tao L, Peng Z, Xiang R. Identification of Hub Genes Associated With Clear Cell Renal Cell Carcinoma by Integrated Bioinformatics Analysis. Front Oncol 2021; 11:726655. [PMID: 34660292 PMCID: PMC8516333 DOI: 10.3389/fonc.2021.726655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/06/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a common genitourinary cancer type with a high mortality rate. Due to a diverse range of biochemical alterations and a high level of tumor heterogeneity, it is crucial to select highly validated prognostic biomarkers to be able to identify subtypes of ccRCC early and apply precision medicine approaches. METHODS Transcriptome data of ccRCC and clinical traits of patients were obtained from the GSE126964 dataset of Gene Expression Omnibus and The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) database. Weighted gene co-expression network analysis (WGCNA) and differentially expressed gene (DEG) screening were applied to detect common differentially co-expressed genes. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes analysis, survival analysis, prognostic model establishment, and gene set enrichment analysis were also performed. Immunohistochemical analysis results of the expression levels of prognostic genes were obtained from The Human Protein Atlas. Single-gene RNA sequencing data were obtained from the GSE131685 and GSE171306 datasets. RESULTS In the present study, a total of 2,492 DEGs identified between ccRCC and healthy controls were filtered, revealing 1,300 upregulated genes and 1,192 downregulated genes. Using WGCNA, the turquoise module was identified to be closely associated with ccRCC. Hub genes were identified using the maximal clique centrality algorithm. After having intersected the hub genes and the DEGs in GSE126964 and TCGA-KIRC dataset, and after performing univariate, least absolute shrinkage and selection operator, and multivariate Cox regression analyses, ALDOB, EFHD1, and ESRRG were identified as significant prognostic factors in patients diagnosed with ccRCC. Single-gene RNA sequencing analysis revealed the expression profile of ALDOB, EFHD1, and ESRRG in different cell types of ccRCC. CONCLUSIONS The present results demonstrated that ALDOB, EFHD1, and ESRRG may act as potential targets for medical therapy and could serve as diagnostic biomarkers for ccRCC.
Collapse
Affiliation(s)
- Hao Huang
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Ling Zhu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Chao Huang
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
- Department of Otolaryngology-Head and Neck Surgery, Second Xiangya Hospital Central South University, Changsha, China
| | - Yi Dong
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Liangliang Fan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| |
Collapse
|
25
|
Aili A, Wen J, Xue L, Wang J. Mutational Analysis of PBRM1 and Significance of PBRM1 Mutation in Anti-PD-1 Immunotherapy of Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:712765. [PMID: 34447697 PMCID: PMC8383204 DOI: 10.3389/fonc.2021.712765] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/23/2021] [Indexed: 01/22/2023] Open
Abstract
Renal cell carcinoma is a common solid tumor. PBRM1 is one of the most mutation-prone genes in clear cell renal cell carcinoma (ccRCC) with the occurrence of mutation in 40% of ccRCC patients. Mutations in PBRM1 have been correlated with the efficacy of immunotherapy. However, the mutation types of PBRM1 are not well characterized. The effects of PBRM1 expression levels in the tumor microenvironment are not well studied. In addition, the mechanism and effect of anti-PD-1 immunotherapy in ccRCC tumor microenvironments are not well clarified. In this study, using bioinformatics methods we analyzed the alternation frequency and expression levels of PBRM1 in various tumors. Next, we experimentally validated their expression levels in ccRCC tissues from human and mouse models. We attempted to clarify the mechanisms of anti-PD-1 immunotherapy in ccRCC with various PBRM1 expression levels. Our results showed that deficiency of PBRM1 protein is correlated with CD4 T cell reduction in human and mouse ccRCC tissues. We also showed that anti-PD-1 Immunotherapy can increase the infiltration of T cells in both PBRM1 high and PBRM1 low tumors but to different degrees. Our study indicates that the reduction of CD4 cells in tumor tissues with low expression of PBRM1 may explain the compromised efficacy of anti-PD-1 immunotherapy in patients with PBRM1 mutated ccRCC. Our study sheds light on the potential of PBRM1 as a therapeutic target in ccRCC.
Collapse
Affiliation(s)
| | - Jie Wen
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Lixiang Xue
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China.,Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
26
|
Chung C. From oxygen sensing to angiogenesis: Targeting the hypoxia signaling pathway in metastatic kidney cancer. Am J Health Syst Pharm 2021; 77:2064-2073. [PMID: 33016992 DOI: 10.1093/ajhp/zxaa308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE This article summarizes examples of current and emerging therapies that target the hypoxia and angiogenesis signaling pathways in the clear cell type of renal cell cancer (RCC), with an emphasis on the hypoxia signaling pathway. SUMMARY Mammalian cells transduce signals of decreased oxygen to hypoxia inducible factor (HIF), an intracellular heterodimer that mediates the adaptation of normal and tumor cells to oxygen deprivation. HIF is frequently overexpressed in cancer cells and is involved in the transcriptional activation of many genes essential for cell invasion, migration, survival, and angiogenesis (including vascular endothelial growth factor [VEGF]). Moreover, HIF confers resistance to cytotoxic chemotherapy and radiation therapy and is associated with poor prognosis in patients with cancer. Blocking the activity of HIF inhibits the expression of VEGF and oncogenic pathways, resulting in the inhibition of tumor growth. Interestingly, activation of oncogenes and/or inactivation of tumor suppressor genes (eg, the gene encoding von Hippel-Lindau [VHL] tumor suppressor protein) can activate tumorigenesis even with normal levels of oxygen, providing support for the notion that the HIF-VHL-VEGF axis is amenable to targeted therapies for the treatment of RCC. This article highlights the current understanding of the hypoxia signaling pathway and its relevance to RCC development. Pharmacologic agents targeting the hypoxia and angiogenesis signaling pathways are discussed. CONCLUSION Development of novel therapeutic agents that target the hypoxia and angiogenesis signaling pathways holds promise in the management of metastatic clear cell RCC.
Collapse
|
27
|
Panunzio A, Tafuri A, Princiotta A, Gentile I, Mazzucato G, Trabacchin N, Antonelli A, Cerruto MA. Omics in urology: An overview on concepts, current status and future perspectives. Urologia 2021; 88:270-279. [PMID: 34169788 DOI: 10.1177/03915603211022960] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent technological advances in molecular biology have led to great progress in the knowledge of structure and function of cells and their main constituents. In this setting, 'omics' is standing out in order to significantly improve the understanding of etiopathogenetic mechanisms of disease and contribute to the development of new biochemical diagnostics and therapeutic tools. 'Omics' indicates the scientific branches investigating every aspect of cell's biology, including structures, functions and dynamics pathways. The main 'omics' are genomics, epigenomics, proteomics, transcriptomics, metabolomics and radiomics. Their diffusion, success and proliferation, addressed to many research fields, has led to many important acquisitions, even in Urology. Aim of this narrative review is to define the state of art of 'omics' application in Urology, describing the most recent and relevant findings, in both oncological and non-oncological diseases, focusing the attention on urinary tract infectious, interstitial cystitis, urolithiasis, prostate cancer, bladder cancer and renal cell carcinoma. In Urology the majority of 'omics' applications regard the pathogenesis and diagnosis of the investigated diseases. In future, its role should be implemented in order to develop specific predictors and tailored treatments.
Collapse
Affiliation(s)
- Andrea Panunzio
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Alessandro Tafuri
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy.,Department of Neuroscience, Imaging and Clinical Science, Physiology and Physiopathology division, "G. D'Annunzio" University, Chieti, Italy
| | - Alessandro Princiotta
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Ilaria Gentile
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Giovanni Mazzucato
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Nicolò Trabacchin
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Alessandro Antonelli
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Maria Angela Cerruto
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| |
Collapse
|
28
|
Gallan AJ, Parilla M, Segal J, Ritterhouse L, Antic T. BAP1-Mutated Clear Cell Renal Cell Carcinoma. Am J Clin Pathol 2021; 155:718-728. [PMID: 33210135 DOI: 10.1093/ajcp/aqaa176] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES While aberrations in the VHL gene and chromosome 3p resulting in clear cell renal cell carcinoma (CCRCC) are well established, we know that additional mutations in chromatin remodeling genes PBRM1, SETD2, and BRCA1-associated protein 1 (BAP1) contribute to pathogenesis in some cases. Given the known aggressive clinical behavior of BAP1-mutated CCRCC, we sought to define the pathologic phenotype of BAP1-mutated CCRCC. METHODS We identified 14 cases of molecularly proven BAP1-mutated CCRCC and investigated their clinicopathologic features. RESULTS BAP1-mutated CCRCC frequently showed papillary, tubulopapillary, or expanded nested architecture; demonstrated granular to diffusely eosinophilic cytoplasm with prominent eosinophilic globules; and contained high-grade nuclei. This morphology demonstrates significant overlap with Xp11 translocation renal cell carcinoma (RCC). Immunohistochemistry notably demonstrates loss of BAP1 expression in almost all tumors, in addition to strong p504S expression. A conventional CCRCC component was frequently present adjacent to the characteristic BAP1 areas and showed retained BAP1 expression and only patchy p504S. Approximately two-thirds of BAP1-mutated CCRCCs were stage pT3, renal vein invasion was common, and 50% developed metastases. CONCLUSIONS Herein, we describe the histologic and immunohistochemical findings in BAP1-mutated CCRCC, which has important implications for utilization of molecular testing, prognosis, future therapeutics, and distinction from other RCC subtypes such as Xp11 translocation RCC.
Collapse
Affiliation(s)
| | - Megan Parilla
- Department of Pathology, University of Chicago, Chicago, IL
| | - Jeremy Segal
- Department of Pathology, University of Chicago, Chicago, IL
| | | | - Tatjana Antic
- Department of Pathology, University of Chicago, Chicago, IL
| |
Collapse
|
29
|
Poon DMC, Chan CK, Chan K, Chu WH, Kwong PWK, Lam W, Law KS, Lee EKC, Liu PL, Sze HCK, Wong JHM, Chan ESY. Consensus statements on the management of metastatic renal cell carcinoma from the Hong Kong Urological Association and the Hong Kong Society of Uro-Oncology 2019. Asia Pac J Clin Oncol 2021; 17 Suppl 3:27-38. [PMID: 33860644 DOI: 10.1111/ajco.13581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND To establish a set of consensus statements for the management of metastatic renal cell carcinoma, a total of 12 urologists and clinical oncologists from two professional associations in Hong Kong formed an expert consensus panel. METHODS Through a series of meetings and using the modified Delphi method, the panelists presented recent evidence, discussed clinical experiences, and drafted consensus statements on several areas of focus regarding the management of metastatic renal cell carcinoma. Each statement was eventually voted upon by every panelist based on the practicability of recommendation. RESULTS A total of 46 consensus statements were ultimately accepted and established by panel voting. CONCLUSIONS Derived from recent evidence and expert insights, these consensus statements were aimed at providing practical guidance to optimize metastatic renal cell carcinoma management and promote a higher standard of clinical care.
Collapse
Affiliation(s)
- Darren Ming-Chun Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong.,Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, New Territories, Hong Kong
| | - Chun-Ki Chan
- Division of Urology, Department of Surgery, Princess Margaret Hospital, New Territories, Hong Kong
| | - Kuen Chan
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong
| | - Wing-Hong Chu
- Suite 418, Central Building, 1 Pedder Street, Central, Hong Kong
| | | | - Wayne Lam
- Department of Surgery, Queen Mary Hospital, University of Hong Kong, Hong Kong Island, Hong Kong
| | - Ka-Suet Law
- Department of Oncology, Princess Margaret Hospital, New Territories, Hong Kong
| | - Eric Ka-Chai Lee
- Department of Clinical Oncology, Tuen Mun Hospital, New Territories, Hong Kong
| | - Pak-Ling Liu
- Department of Surgery, Caritas Medical Centre, Kowloon, Hong Kong
| | | | - Joseph Hon-Ming Wong
- Division of Urology, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, New Territories, Hong Kong
| | | |
Collapse
|
30
|
Zheng W, Zhang S, Guo H, Chen X, Huang Z, Jiang S, Li M. Multi-omics analysis of tumor angiogenesis characteristics and potential epigenetic regulation mechanisms in renal clear cell carcinoma. Cell Commun Signal 2021; 19:39. [PMID: 33761933 PMCID: PMC7992844 DOI: 10.1186/s12964-021-00728-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tumor angiogenesis, an essential process for cancer proliferation and metastasis, has a critical role in prognostic of kidney renal clear cell carcinoma (KIRC), as well as a target in guiding treatment with antiangiogenic agents. However, tumor angiogenesis subtypes and potential epigenetic regulation mechanisms in KIRC patient remains poorly characterized. System evaluation of angiogenesis subtypes in KIRC patient might help to reveal the mechanisms of KIRC and develop more target treatments for patients. METHOD Ten independent tumor angiogenesis signatures were obtained from molecular signatures database (MSigDB) and gene set variation analysis was performed to calculate the angiogenesis score in silico using the Cancer Genome Atlas (TCGA) KIRC dataset. Tumor angiogenesis subtypes in 539 TCGA-KIRC patients were identified using consensus clustering analysis. The potential regulation mechanisms was studied using gene mutation, copy number variation, and differential methylation analysis (DMA). The master transcription factors (MTF) that cause the difference in tumor angiogenesis signals were completed by transcription factor enrichment analysis. RESULTS The angiogenesis score of a prognosis related angiogenesis signature including 189 genes was significantly correlated with immune score, stroma score, hypoxia score, and vascular endothelial growth factor (VEGF) signal score in 539 TCGA KIRC patients. MMRN2, CLEC14A, ACVRL1, EFNB2, and TEK in candidate gene set showed highest correlation coefficient with angiogenesis score in TCGA-KIRC patients. In addition, all of them were associated with overall survival in both TCGA-KIRC and E-MTAB-1980 KIRC data. Clustering analysis based on 183 genes in angiogenesis signature identified two prognosis related angiogenesis subtypes in TCGA KIRC patients. Two clusters also showed different angiogenesis score, immune score, stroma score, hypoxia score, VEGF signal score, and microenvironment score. DMA identified 59,654 differential methylation sites between two clusters and part of these sites were correlated with tumor angiogenesis genes including CDH13, COL4A3, and RHOB. In addition, RFX2, SOX13, and THRA were identified as top three MTF in regulating angiogenesis signature in KIRC patients. CONCLUSION Our study indicate that evaluation the angiogenesis subtypes of KIRC based on angiogenesis signature with 183 genes and potential epigenetic mechanisms may help to develop more target treatments for KIRC patients. Video Abstract.
Collapse
Affiliation(s)
- Wenzhong Zheng
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Shiqiang Zhang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, Guangdong, People's Republic of China
| | - Xiaobao Chen
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Zhangcheng Huang
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Shaoqin Jiang
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China
| | - Mengqiang Li
- Department of Urology, Fujian Province, Fujian Medical University Union Hospital, Gulou District, 29 Xinquan Road, Fuzhou, 200001, People's Republic of China.
| |
Collapse
|
31
|
Pyziak K, Sroka-Porada A, Rzymski T, Dulak J, Łoboda A. Potential of enhancer of zeste homolog 2 inhibitors for the treatment of SWI/SNF mutant cancers and tumor microenvironment modulation. Drug Dev Res 2021; 82:730-753. [PMID: 33565092 DOI: 10.1002/ddr.21796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/17/2022]
Abstract
Enhancer of zeste homolog 2 (EZH2), a catalytic component of polycomb repressive complex 2 (PRC2), is commonly overexpressed or mutated in many cancer types, both of hematological and solid nature. Till now, plenty of EZH2 small molecule inhibitors have been developed and some of them have already been tested in clinical trials. Most of these inhibitors, however, are effective only in limited cases in the context of EZH2 gain-of-function mutated tumors such as lymphomas. Other cancer types with aberrant EZH2 expression and function require alternative approaches for successful treatment. One possibility is to exploit synthetic lethal strategy, which is based on the phenomenon that concurrent loss of two genes is detrimental but the deletion of either of them leaves cell viable. In the context of EZH2/PRC2, the most promising synthetic lethal target seems to be SWItch/Sucrose Non-Fermentable chromatin remodeling complex (SWI/SNF), which is known to counteract PRC2 functions. SWI/SNF is heavily involved in carcinogenesis and its subunits have been found mutated in approximately 20% of tumors of different kinds. In the current review, we summarize the existing knowledge of synthetic lethal relationships between EZH2/PRC2 and components of the SWI/SNF complex and discuss in detail the potential application of existing EZH2 inhibitors in cancer patients harboring mutations in SWI/SNF proteins. We also highlight recent discoveries of EZH2 involvement in tumor microenvironment regulation and consequences for future therapies. Although clinical studies are limited, the fundamental research might help to understand which patients are most likely to benefit from therapies using EZH2 inhibitors.
Collapse
Affiliation(s)
- Karolina Pyziak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.,Biology R&D, Ryvu Therapeutics S.A., Kraków, Poland
| | | | | | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
32
|
Singh D. Current updates and future perspectives on the management of renal cell carcinoma. Life Sci 2020; 264:118632. [PMID: 33115605 DOI: 10.1016/j.lfs.2020.118632] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) refers to renal-epithelial cancer, which represents over 90% of kidney cancer and is a cause for cancer related deaths in the world. Studies suggested somatic VHL mutations to be the cause for the occurrence of cancer, but with the time, more latest genomic and biological studies have detected variation in epigenetic regulatory genes and showed significant heterogeneity of the intratumor that may lead to strategies of diagnostic, predictive, and therapeutic importance. Immune dysfunction is responsible for almost all types of renal cancer, and angiogenesis and immunosuppression function together in the tumor microenvironment of renal cell carcinoma (RCC). Over the past few years, advancement in the management of the RCC has finally revolutionized with the arrival of the entrapped immune inhibitors which particularly concentrated on the receptor (programmed cell death-1) and focus on the new generation receptor i.e. TKRI (tyrosine-kinase receptor inhibitors). The present review deals with the comprehensive review of RCC and emphasizes on its types, pathogenesis and advancement in these diseases. This review also overviews the role of innate and adaptive immune response-related mechanism, the function of cancer stem cell in this diseases, therapeutic targeted drugs and hormonal signaling pathways as an emerging strategy in the management of the renal cancer.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad 211007, Uttar Pradesh, India.
| |
Collapse
|
33
|
Ohsugi H, Yoshida T, Ohe C, Ikeda J, Sugi M, Kinoshita H, Tsuta K, Matsuda T. The SSPN Score, a Novel Scoring System Incorporating PBRM1 Expression, Predicts Postoperative Recurrence for Patients with Non-metastatic Clear Cell Renal Cell Carcinoma. Ann Surg Oncol 2020; 28:2359-2366. [PMID: 32940805 DOI: 10.1245/s10434-020-09075-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND The loss of PBRM1 expression (as identified by immunohistochemistry) is associated with a high risk of postoperative recurrence for patients with clear cell renal cell carcinoma (ccRCC). The authors developed a scoring system to predict recurrence based on clinicopathologic factors incorporating PBRM1 expression. METHODS This study retrospectively reviewed 479 ccRCC patients who underwent radical surgery between 2006 and 2017. The study extracted a subset of 389 non-metastatic ccRCC patients for whom relevant clinicopathologic factors were available. The primary end point was recurrence-free survival (RFS). The Kaplan-Meier method and the Cox proportional hazards model were used for statistical analysis. Leibovich score, SSIGN score, and University of California, Los Angeles (UCLA) Integrated Staging System were included as conventional prediction models. RESULTS Of the 389 patients, 53 (13.6%) experienced recurrence during a median period of 61 months. Multivariable analyses showed that that the independent factors for RFS were ≥ pT3 (hazard ratio [HR] 3.64; P < 0.001), sarcomatoid or rhabdoid component (HR 3.29; P = 0.005), PBRM1 negativity (HR 3.39; P = 0.001), and necrosis (HR 3.60; P < 0.001). A scoring system calculated with these factors, named the SSPN (stage, sarcomatoid, PBRM1 expression, and necrosis) score, showed significant differences in RFS among the following four groups; low-risk group (0 factors), intermediate-risk group (1 factor), high-risk group (2 to 3 factors), and very high-risk group (4 factors) (P < 0.001). The authors' model also showed a greater predictive accuracy for 5-year RFS than the conventional models (0.841 vs 0.747-0.792). CONCLUSIONS The SSPN score, which integrates clinicopathologic findings and PBRM1 expression, can accurately predict postoperative recurrence for patients with non-metastatic ccRCC after radical surgery.
Collapse
Affiliation(s)
- Haruyuki Ohsugi
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Chisato Ohe
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Osaka, Japan
| | - Junichi Ikeda
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Motohiko Sugi
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Koji Tsuta
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan.
| |
Collapse
|
34
|
Song XD, Tian YN, Li H, Liu B, Zhang AL, Hong Y. Research progress on advanced renal cell carcinoma. J Int Med Res 2020; 48:300060520924265. [PMID: 32529862 PMCID: PMC7294379 DOI: 10.1177/0300060520924265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/15/2020] [Indexed: 01/29/2023] Open
Abstract
Renal cell carcinoma (RCC) is a malignant tumor and the third most common urinary disease. It was estimated that RCC affected over 350,000 individuals in 2013, and there are nearly 140,000 deaths annually due to this disease. The initial masses in RCC patients are mostly confined to a single organ. However, due to the metastatic spread of cancer cells through the circulatory system, more than 30% of RCC patients relapse after surgery. The appearance of distant metastases often means that patients enter the advanced stage of cancer with low quality of life and a short expected survival time. This review aims to describe the extant research on advanced RCC, including its pathophysiology, heterogeneity, diagnosis, treatment, and prospects. We try to highlight the most suitable means of treating advanced RCC patients, focusing on comprehensive personalized treatments.
Collapse
Affiliation(s)
- Xin-da Song
- Department of Urinary Surgery, Graduate School of Peking Union Medical College, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Yi-nong Tian
- Department of Urinary Surgery, Xinle City Hospital, Shijiazhuang, Hebei Province, China
| | - Hao Li
- Department of Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bin Liu
- Department of Urinary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Ai-li Zhang
- Department of Urinary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yang Hong
- Department of Clinical Pharmacology Research, Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
35
|
Ball MW, An JY, Gomella PT, Gautam R, Ricketts CJ, Vocke CD, Schmidt LS, Merino MJ, Srinivasan R, Malayeri AA, Metwalli AR, Linehan WM. Growth Rates of Genetically Defined Renal Tumors: Implications for Active Surveillance and Intervention. J Clin Oncol 2020; 38:1146-1153. [PMID: 32083993 PMCID: PMC7145590 DOI: 10.1200/jco.19.02263] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2020] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Published series of growth rates of renal tumors on active surveillance largely consist of tumors without pathologic or genetic data. Growth kinetics of genetically defined renal tumors are not well known. Here, we evaluate the growth of genetically defined renal tumors and their association with patient clinical and genetic characteristics. PATIENTS AND METHODS We evaluated patients with an inherited kidney cancer susceptibility syndrome as a result of a pathologic germline alteration of VHL, MET, FLCN, or BAP1 with at least 1 solid renal mass managed with active surveillance at our institution. Tumor growth rates (GR) were calculated and patients were stratified by genetic alteration and other clinical and genetic factors to analyze differences in growth rates using linear regression and comparative statistics. RESULTS A total of 292 patients with 435 genetically defined tumors were identified, including 286 VHL-deficient, 91 FLCN-deficient, 52 MET-activated, and 6 BAP1-deficient tumors. There were significant differences in GRs when stratified by genetic alteration. BAP1-deficient tumors had the fastest median GR (0.6 cm/y; interquartile range [IQR], 0.57-0.68 cm/y), followed by VHL-deficient tumors (GR, 0.37 cm/y; IQR, 0.25-0.57 cm/y), FLCN-deficient tumors (GR, 0.10 cm/y; IQR, 0.04-0.24 cm/y), and tumors with MET activation (GR, 0.15 cm/y; IQR, 0.053-0.32 cm/y; P < .001). Tumors from the same patient had similar GRs. Younger age was independently associated with higher GR (P = .005). CONCLUSION In a cohort of genetically defined tumors, tumor growth rates varied in a clinically and statistically different manner according to genetic subtype. Rapid growth of BAP1-deficient tumors indicates that these patients should be managed with caution. The faster growth of tumors in younger patients may support more frequent imaging, whereas the slower growth of other tumors may support extended surveillance beyond annual imaging in some instances.
Collapse
Affiliation(s)
- Mark W. Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Julie Y. An
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Patrick T. Gomella
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rabindra Gautam
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Cathy D. Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Laura S. Schmidt
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Maria J. Merino
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ashkan A. Malayeri
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Adam R. Metwalli
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
36
|
Tronik-Le Roux D, Sautreuil M, Bentriou M, Vérine J, Palma MB, Daouya M, Bouhidel F, Lemler S, LeMaoult J, Desgrandchamps F, Cournède PH, Carosella ED. Comprehensive landscape of immune-checkpoints uncovered in clear cell renal cell carcinoma reveals new and emerging therapeutic targets. Cancer Immunol Immunother 2020; 69:1237-1252. [PMID: 32166404 DOI: 10.1007/s00262-020-02530-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 02/18/2020] [Indexed: 12/18/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) constitutes the most common renal cell carcinoma subtype and has long been recognized as an immunogenic cancer. As such, significant attention has been directed toward optimizing immune-checkpoints (IC)-based therapies. Despite proven benefits, a substantial number of patients remain unresponsive to treatment, suggesting that yet unreported, immunosuppressive mechanisms coexist within tumors and their microenvironment. Here, we comprehensively analyzed and ranked forty-four immune-checkpoints expressed in ccRCC on the basis of in-depth analysis of RNAseq data collected from the TCGA database and advanced statistical methods designed to obtain the group of checkpoints that best discriminates tumor from healthy tissues. Immunohistochemistry and flow cytometry confirmed and enlarged the bioinformatics results. In particular, by using the recursive feature elimination method, we show that HLA-G, B7H3, PDL-1 and ILT2 are the most relevant genes that characterize ccRCC. Notably, ILT2 expression was detected for the first time on tumor cells. The levels of other ligand-receptor pairs such as CD70:CD27; 4-1BB:4-1BBL; CD40:CD40L; CD86:CTLA4; MHC-II:Lag3; CD200:CD200R; CD244:CD48 were also found highly expressed in tumors compared to adjacent non-tumor tissues. Collectively, our approach provides a comprehensible classification of forty-four IC expressed in ccRCC, some of which were never reported before to be co-expressed in ccRCC. In addition, the algorithms used allowed identifying the most relevant group that best discriminates tumor from healthy tissues. The data can potentially assist on the choice of valuable immune-therapy targets which hold potential for the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
- Diana Tronik-Le Roux
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France. .,Université de paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75010, Paris, France. .,CEA, Direction de La Recherche Fondamentale, Service de Recherche en Hémato-Immunologie, Hôpital Saint-Louis, IUH, 1, avenue Claude Vellefaux, 75010, Paris, France.
| | - Mathilde Sautreuil
- Laboratory of Mathematics and Informatics (MICS), CentraleSupélec, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Mahmoud Bentriou
- Laboratory of Mathematics and Informatics (MICS), CentraleSupélec, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Jérôme Vérine
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France.,Service D'Anatomo-Pathologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Maria Belén Palma
- Cátedra de Citología, Histología Y Embriología A, Facultad de Ciencias Médicas, UNLP, Buenos Aires, Argentina
| | - Marina Daouya
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France.,Université de paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - Fatiha Bouhidel
- Service D'Anatomo-Pathologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Sarah Lemler
- Laboratory of Mathematics and Informatics (MICS), CentraleSupélec, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Joel LeMaoult
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France.,Université de paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75010, Paris, France
| | - François Desgrandchamps
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France.,Service D'Urologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Paul-Henry Cournède
- Laboratory of Mathematics and Informatics (MICS), CentraleSupélec, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Edgardo D Carosella
- Commissariat à L'Energie Atomique Et Aux Energies Alternatives (CEA), Direction de La Recherche Fondamentale (DRF), Service de Recherche en Hémato-Immunologie (SRHI), Hôpital Saint-Louis, Paris, France.,Université de paris, U976 HIPI Unit, Institut de Recherche Saint-Louis, 75010, Paris, France
| |
Collapse
|
37
|
Feng Z, Zhang L, Qi Z, Shen Q, Hu Z, Chen F. Identifying BAP1 Mutations in Clear-Cell Renal Cell Carcinoma by CT Radiomics: Preliminary Findings. Front Oncol 2020; 10:279. [PMID: 32185138 PMCID: PMC7058626 DOI: 10.3389/fonc.2020.00279] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/18/2020] [Indexed: 12/01/2022] Open
Abstract
To evaluate the potential application of computed tomography (CT) radiomics in the prediction of BRCA1-associated protein 1 (BAP1) mutation status in patients with clear-cell renal cell carcinoma (ccRCC). In this retrospective study, clinical and CT imaging data of 54 patients were retrieved from The Cancer Genome Atlas–Kidney Renal Clear Cell Carcinoma database. Among these, 45 patients had wild-type BAP1 and nine patients had BAP1 mutation. The texture features of tumor images were extracted using the Matlab-based IBEX package. To produce class-balanced data and improve the stability of prediction, we performed data augmentation for the BAP1 mutation group during cross validation. A model to predict BAP1 mutation status was constructed using Random Forest Classification algorithms, and was evaluated using leave-one-out-cross-validation. Random Forest model of predict BAP1 mutation status had an accuracy of 0.83, sensitivity of 0.72, specificity of 0.87, precision of 0.65, AUC of 0.77, F-score of 0.68. CT radiomics is a potential and feasible method for predicting BAP1 mutation status in patients with ccRCC.
Collapse
Affiliation(s)
- Zhan Feng
- Department of Radiology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Lixia Zhang
- Department of Radiology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhong Qi
- Department of Radiology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Qijun Shen
- Department of Radiology, Hangzhou First People's Hospital, Hangzhou, China
| | - Zhengyu Hu
- Department of Radiology, Second People's Hospital of Yuhang District, Hangzhou, China
| | - Feng Chen
- Department of Radiology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
George G, Schmidt L, Tolat P, Riese M, Kilari D. Salvage ipilimumab associated with a significant response in sarcomatoid renal cell carcinoma. J Immunother Cancer 2020; 8:e000584. [PMID: 32114501 PMCID: PMC7057424 DOI: 10.1136/jitc-2020-000584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2020] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Metastatic sarcomatoid renal cell carcinoma (sRCC) is an aggressive variant of RCC with generally poor prognosis. Treatment with vascular endothelial growth factor inhibitors or chemotherapy generates only short-lived responses. Recent research has suggested a role for combination checkpoint inhibition as first line treatment for metastatic sRCC. This therapy consists of induction with cytotoxic T-lymphocyte-associated protein 4 inhibitor, ipilimumab, administered with programmed cell death protein 1 (PD-1) inhibitor, nivolumab. After completion of four cycles of combination therapy, single-agent maintenance nivolumab is recommended until progression. Patients who progress on maintenance nivolumab are switched to alternate therapy. Herein, we present a case of a patient with RCC who progressed on maintenance nivolumab who, on retreatment with ipilimumab, demonstrated a significant response In addition, we summarize important findings to support the role of salvage ipilimumab in patients with sRCC. CASE PRESENTATION A 46-year-old man presented with flank pain and hematuria, the work up of which noted a left kidney mass for which he underwent nephrectomy and was diagnosed with localized sRCC with 60% sarcomatoid differentiation. Within 3 months of nephrectomy, he presented with recurrent flank pain and was diagnosed with recurrence of disease. He was treated with ipilimumab 1 mg/kg and nivolumab 3 mg/kg for four doses and demonstrated a partial response. He was then transitioned to single agent nivolumab maintenance. After 3 months on maintenance therapy, he was noted to have progression of disease. Given prior response to immune check point combination, it was decided to rechallenge the patient with 1 mg/kg ipilimumab. After two doses of ipilimumab and nivolumab combination therapy, the patient was noted to have a partial response. He maintained a response for an additional 9 months and treatment was eventually discontinued due to grade 3 toxicity and progression. CONCLUSIONS This case report demonstrates the utility of retreatment with ipilimumab as a salvage option for patients progressing on maintenance PD-1 inhibitors in metastatic RCC. Further studies are needed to identify predictors of response and toxicity to this approach, as well as the optimal scheduling of ipilimumab with maintenance nivolumab.
Collapse
Affiliation(s)
- Gemlyn George
- Department of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Laura Schmidt
- Department of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Parag Tolat
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mathew Riese
- Department of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI 53213, USA
| | - Deepak Kilari
- Department of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
39
|
Abstract
Long non-coding RNAs (lncRNAs) are regulators of cellular machinery that are commonly dysregulated in genitourinary malignancies. Accordingly, the investigation of lncRNAs is improving our understanding of genitourinary cancers, from development to progression and dissemination. lncRNAs are involved in major oncogenic events in genitourinary malignancies, including androgen receptor (AR) signalling in prostate cancer, hypoxia-inducible factor (HIF) pathway activation in renal cell carcinoma and invasiveness in bladder cancer, as well as multiple other proliferation and survival mechanisms. In line with their putative oncogenic roles, new lncRNA-based classifications are emerging as potent predictors of prognosis. In clinical practice, detection of oncogenic lncRNAs in serum or urine might enable early cancer detection, and lncRNAs might also be promising therapeutic targets for patients with genitourinary cancer. Furthermore, as predictors of sensitivity to anticancer treatments, lncRNAs could be integrated into future precision medicine strategies. Overall, lncRNAs are promising new candidates for molecular studies and for discovery of innovative biomarkers and are putative therapeutic targets in genitourinary oncology.
Collapse
|
40
|
Li Y, Ding Q, Xiong Z, Wen H, Feng C. Overexpression of steroid sulfotransferase genes is associated with worsened prognosis and with immune exclusion in clear cell-renal cell carcinoma. Aging (Albany NY) 2019; 11:9209-9219. [PMID: 31655797 PMCID: PMC6834411 DOI: 10.18632/aging.102392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022]
Abstract
Aim: Steroid sulfotransferase (SULT) plays physiological roles but its role in clear cell-renal cell carcinoma (ccRCC) remains unclear. We therefore investigated genetic alteration of steroid SULT genes in ccRCC. Results: Overexpression of any of SULT genes occurred in ~8% of ccRCC patients. Overexpression of steroid SULT genes was associated with worsened prognosis. Steroid SULT gene-upregulated ccRCC cases showed mutual exclusivity with mutations of VHL, SETD2 and PBRM1, and with focal deletions of 3p and 9p, respectively. Expressions of SULT genes were negatively correlated with that of VHL, SETD2 and PBRM1, respectively. While no cancer-intrinsic pathway was enriched, immune signatures were significantly enriched in SULT gene-overexpressed cases, resulting in significantly fewer infiltration of lymphocytes. Targeting SULT1B1 significantly inhibited growth of ccRCC cells. Conclusion: Steroid SULT genes were associated with worsened prognosis and with immune exclusion in ccRCC. Methods: In silico reproduction of TGGA and GTEx datasets was performed. Data were processed comprehensively using the platforms of cBioPotal, GEPIA, Human Protein Atlas, TIMER, respectively. Functional annotation was analyzed using platforms of NET-GE and GSEA, respectively. In vitro assays were performed for validation.
Collapse
Affiliation(s)
- Yuqing Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Zuquan Xiong
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Hui Wen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| |
Collapse
|
41
|
Stühler V, Maas JM, Rausch S, Stenzl A, Bedke J. Immune checkpoint inhibition for the treatment of renal cell carcinoma. Expert Opin Biol Ther 2019; 20:83-94. [PMID: 31587590 DOI: 10.1080/14712598.2020.1677601] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: The systemic therapy in metastatic renal cell carcinoma (mRCC) is moving from tyrosine kinase inhibitors (TKIs) and mammalian target of rapamycin (mTOR) inhibitors to immune checkpoint inhibitors and its combination with TKIs.Areas covered: This review provides a general overview using immune checkpoint inhibition for the treatment of RCC. Clinical results from conducted and ongoing clinical trials are summarized and checkpoint inhibition is reviewed in the context to other available systemic therapies such as TKIs for mRCC based on the different International Metastastic RCC Database Consortium (IMCD) risk groups. Furthermore, prospects for the use of predictive biomarkers in the decision-making process of chosen therapy will be given.Expert opinion: Using checkpoint inhibition in mRCC has demonstrated a superior efficacy for patients with IMDC intermediate and poor risk for ipilimumab combined with nivolumab. Furthermore, therapeutic regimes with tyrosine kinase inhibition plus immune checkpoint-inhibition were recently presented and demonstrated superiority in all risk groups for axitinib plus pembrolizumab in overall survival and progression-free survival (PFS) and axitinib plus avelumab in PFS compared to sunitinib monotherapy. Novel biomarkers of response to further optimize therapeutic selection and patient outcomes are ongoing medical objectives.
Collapse
Affiliation(s)
- Viktoria Stühler
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Jan Moritz Maas
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Steffen Rausch
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen, Tübingen, Germany
| | - Jens Bedke
- Department of Urology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
42
|
Zeng J, Xiang W, Zhang Y, Huang C, Chen K, Chen Z. Ubiquitous expressed transcript promotes tumorigenesis by acting as a positive modulator of the polycomb repressive complex 2 in clear cell renal cell carcinoma. BMC Cancer 2019; 19:874. [PMID: 31481081 PMCID: PMC6724258 DOI: 10.1186/s12885-019-6069-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 08/20/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The ubiquitous expressed transcript (UXT) plays a key role in various tumors by regulating transcriptional activity of multiple transcription factors, including androgen receptor (AR). However, the role of UXT in clear cell renal cell carcinoma (ccRCC) is still unknown. METHODS Yeast two-hybrid screening, GST pull-down and co-immunoprecipitation assays were performed to detect the interacting protein of UXT. Chromatin immunoprecipitation (ChIP) was performed to investigate the levels of histone H3 lysine 27 trimethylation at the HOXA9 promoters. CCK-8 assays, colony formation assays and Transwell assays were performed to detect the proliferation, colony formation, migration and invasion of renal cancer cells. Quantitative PCR analysis was performed to detect the expressions of UXT in human ccRCC samples. RESULTS The enhancer of zeste homolog 2 (EZH2) is a novel UXT interacting protein and UXT interacts with EZH2 in the nucleus. In addition, UXT interacts with the polycomb repressive complex 2 (PRC2) through directly binding to EZH2 and suppressor of zeste 12 homolog (SUZ12), but not to embryonic ectoderm development (EED). Moreover, the UXT activates EZH2 histone methyltransferase activity by facilitating EZH2 binding with SUZ12. We further provided striking evidences that knockdown of UXT inhibits proliferation, colony formation, migration and invasion of renal cancer cells, in an EZH2-dependent manner. Importantly, the upregulation of UXT expression was observed in clinical ccRCC samples, and the high expression level of UXT was associated with advanced stage, distant metastasis and poor overall survival in patients with ccRCC. CONCLUSION The UXT is a novel regulator of the PRC2 and acts as a renal cancer oncogene that affects the progression and survival of ccRCC patients.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
- Department of Urology, the First Affiliated Hospital of Nanchang University, Nanchang, 330000 People’s Republic of China
| | - Wei Xiang
- College of Basic Medicine, Hubei University of Traditional Chinese Medicine, Wuhan, 430065 People’s Republic of China
| | - Yucong Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
- Department of Geriatric, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Chunhua Huang
- College of Basic Medicine, Hubei University of Traditional Chinese Medicine, Wuhan, 430065 People’s Republic of China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 People’s Republic of China
| |
Collapse
|
43
|
Christensen MB, Wadt K, Jensen UB, Lautrup CK, Bojesen A, Krogh LN, van Overeem Hansen T, Gerdes AM. Exploring the hereditary background of renal cancer in Denmark. PLoS One 2019; 14:e0215725. [PMID: 31034483 PMCID: PMC6488054 DOI: 10.1371/journal.pone.0215725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Every year more than 800 patients in Denmark are diagnosed with renal cell carcinoma (RCC) of which 3-5% are expected to be part of a hereditary renal cancer syndrome. We performed genetic screening of causative and putative RCC-genes (VHL, FH, FLCN, MET, SDHB, BAP1, MITF, CDKN2B) in RCC-patients suspected of a genetic predisposition. METHODS The cohort consisted of forty-eight Danish families or individuals with early onset RCC, a family history of RCC, a family history of RCC and melanoma or both RCC- and melanoma diagnosis in the same individual. DNA was extracted from peripheral blood samples or cancer-free formalin-fixed paraffin-embedded tissue. RESULTS One start codon variant of unknown clinical significance (VUS) (c.3G>A, p.Met1Ile) and one missense VUS (c.631A>C, p.Met211Leu) was found in VHL in a patient with RCC-onset at twenty-eight years of age but without other manifestations or family history of von Hippel-Lindau (VHL). Furthermore, in three families we found three different variants in BAP1, one of which was a novel non-segregating missense variant (c.1502G>A, p.Ser501Asn) in a family with two brothers affected with RCC. Finally, we found the known E318K-substitution in MITF in a RCC-affected member of a family with multiple melanomas. No variants were detected in CDKN2B. CONCLUSION Although we did find three VUS's in BAP1 in three families and a pathogenic variant in MITF in one family, pathogenic germline variants in BAP1, MITF or CDKN2B are not frequent causes of hereditary renal cancer in Denmark. It is possible that the high prevalence of risk factors such as male gender, smoking and obesity has influenced the development of cancer in the patients of the current study. Further investigations into putative predisposing genes and risk factors of RCC are necessary to enable better prediction of renal cancer risk or presymptomatic testing of relatives in hereditary renal cancer families.
Collapse
Affiliation(s)
| | - Karin Wadt
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anders Bojesen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Genetics, Sygehus Lillebaelt, Vejle, Denmark
| | | | | | - Anne-Marie Gerdes
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
44
|
Lameirinhas A, Miranda-Gonçalves V, Henrique R, Jerónimo C. The Complex Interplay between Metabolic Reprogramming and Epigenetic Alterations in Renal Cell Carcinoma. Genes (Basel) 2019; 10:E264. [PMID: 30986931 PMCID: PMC6523766 DOI: 10.3390/genes10040264] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/24/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common malignancy affecting the kidney. Current therapies are mostly curative for localized disease, but do not completely preclude recurrence and metastization. Thus, it is imperative to develop new therapeutic strategies based on RCC biological properties. Presently, metabolic reprograming and epigenetic alterations are recognized cancer hallmarks and their interactions are still in its infancy concerning RCC. In this review, we explore RCC biology, highlighting genetic and epigenetic alterations that contribute to metabolic deregulation of tumor cells, including high glycolytic phenotype (Warburg effect). Moreover, we critically discuss available data concerning epigenetic enzymes' regulation by aberrant metabolite accumulation and their consequences in RCC emergence and progression. Finally, we emphasize the clinical relevance of uncovering novel therapeutic targets based on epigenetic reprograming by metabolic features to improve treatment and survival of RCC patients.
Collapse
Affiliation(s)
- Ana Lameirinhas
- Cancer Biology & Epigenetics Group-Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), 4200-072 Porto, Portugal.
- Master in Oncology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal.
| | - Vera Miranda-Gonçalves
- Cancer Biology & Epigenetics Group-Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), 4200-072 Porto, Portugal.
| | - Rui Henrique
- Cancer Biology & Epigenetics Group-Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), 4200-072 Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar⁻ University of Porto (ICBAS-UP), 4050-313 Porto, Portugal.
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group-Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar⁻ University of Porto (ICBAS-UP), 4050-313 Porto, Portugal.
| |
Collapse
|
45
|
Stukalin I, Wells JC, Graham J, Yuasa T, Beuselinck B, Kollmansberger C, Ernst DS, Agarwal N, Le T, Donskov F, Hansen AR, Bjarnason GA, Srinivas S, Wood LA, Alva AS, Kanesvaran R, Fu SYF, Davis ID, Choueiri TK, Heng DYC. Real-world outcomes of nivolumab and cabozantinib in metastatic renal cell carcinoma: results from the International Metastatic Renal Cell Carcinoma Database Consortium. ACTA ACUST UNITED AC 2019; 26:e175-e179. [PMID: 31043824 DOI: 10.3747/co.26.4595] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objectives In the present study, we explored the real-world efficacy of the immuno-oncology checkpoint inhibitor nivolumab and the tyrosine kinase inhibitor cabozantinib in the second-line setting. Methods Using the International Metastatic Renal Cell Carcinoma Database Consortium (imdc) dataset, a retrospective analysis of patients with metastatic renal cell carcinoma (mrcc) treated with nivolumab or cabozantinib in the second line after prior therapy targeted to the vascular endothelial growth factor receptor (vegfr) was performed. Baseline characteristics and imdc risk factors were collected. Overall survival (os) and time to treatment failure (ttf) were calculated using Kaplan-Meier curves. Overall response rates (orrs) were determined for each therapy. Multivariable Cox regression analysis was performed to determine survival differences between cabozantinib and nivolumab treatment. Results The analysis included 225 patients treated with nivolumab and 53 treated with cabozantinib. No significant difference in median os was observed: 22.10 months [95% confidence interval (ci): 17.18 months to not reached] with nivolumab and 23.70 months (95% ci: 15.52 months to not reached) with cabozantinib (p = 0.61). The ttf was also similar at 6.90 months (95% ci: 4.60 months to 9.20 months) with nivolumab and 7.39 months (95% ci: 5.52 months to 12.85 months) with cabozantinib (p = 0.20). The adjusted hazard ratio (hr) for nivolumab compared with cabozantinib was 1.30 (95% ci: 0.73 to 2.3), p = 0.38. When adjusted by imdc criteria and age, the hr was 1.32 (95% ci: 0.74 to 2.38), p = 0.35. Conclusions Real-world imdc data indicate comparable os and ttf for nivolumab and cabozantinib. Both agents are reasonable therapeutic options for patients progressing after initial first-line vegfr-targeted therapy.
Collapse
Affiliation(s)
- I Stukalin
- Alberta: Tom Baker Cancer Center, University of Calgary, Calgary (Stukalin, Wells, Heng)
| | - J C Wells
- Alberta: Tom Baker Cancer Center, University of Calgary, Calgary (Stukalin, Wells, Heng).,Ontario: Queen's University, Kingston (Wells); London Health Sciences Centre, London (Ernst); Princess Margaret Cancer Centre, University Health Network, Toronto (Hansen); Sunnybrook Odette Cancer Centre, Toronto (Bjarnason)
| | - J Graham
- Alberta: Tom Baker Cancer Center, University of Calgary, Calgary (Stukalin, Wells, Heng)
| | - T Yuasa
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | - B Beuselinck
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | | | - D S Ernst
- Ontario: Queen's University, Kingston (Wells); London Health Sciences Centre, London (Ernst); Princess Margaret Cancer Centre, University Health Network, Toronto (Hansen); Sunnybrook Odette Cancer Centre, Toronto (Bjarnason)
| | - N Agarwal
- United States: University of Utah Huntsman Cancer Institute, Salt Lake City, UT (Agarwal); University of Texas Southwestern Medical Center, Dallas, TX (Le); Stanford Medical Center, Stanford, CA (Srinivas); University of Michigan, Ann Arbor, MI (Alva); Dana-Farber Cancer Institute, Boston, MA (Choueiri)
| | - T Le
- United States: University of Utah Huntsman Cancer Institute, Salt Lake City, UT (Agarwal); University of Texas Southwestern Medical Center, Dallas, TX (Le); Stanford Medical Center, Stanford, CA (Srinivas); University of Michigan, Ann Arbor, MI (Alva); Dana-Farber Cancer Institute, Boston, MA (Choueiri)
| | - F Donskov
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | - A R Hansen
- Ontario: Queen's University, Kingston (Wells); London Health Sciences Centre, London (Ernst); Princess Margaret Cancer Centre, University Health Network, Toronto (Hansen); Sunnybrook Odette Cancer Centre, Toronto (Bjarnason)
| | - G A Bjarnason
- Ontario: Queen's University, Kingston (Wells); London Health Sciences Centre, London (Ernst); Princess Margaret Cancer Centre, University Health Network, Toronto (Hansen); Sunnybrook Odette Cancer Centre, Toronto (Bjarnason)
| | - S Srinivas
- United States: University of Utah Huntsman Cancer Institute, Salt Lake City, UT (Agarwal); University of Texas Southwestern Medical Center, Dallas, TX (Le); Stanford Medical Center, Stanford, CA (Srinivas); University of Michigan, Ann Arbor, MI (Alva); Dana-Farber Cancer Institute, Boston, MA (Choueiri)
| | - L A Wood
- Nova Scotia: Queen Elizabeth II Health Sciences Centre, Halifax (Wood)
| | - A S Alva
- United States: University of Utah Huntsman Cancer Institute, Salt Lake City, UT (Agarwal); University of Texas Southwestern Medical Center, Dallas, TX (Le); Stanford Medical Center, Stanford, CA (Srinivas); University of Michigan, Ann Arbor, MI (Alva); Dana-Farber Cancer Institute, Boston, MA (Choueiri)
| | - R Kanesvaran
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | - S Y F Fu
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | - I D Davis
- non-United States international: Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan (Yuasa); University Hospitals Leuven, Leuven, Belgium (Beuselinck); Aarhus University Hospital, Aarhus, Denmark (Donskov); National Cancer Centre Singapore, Singapore (Kanesvaran); Auckland City Hospital, Auckland, New Zealand (Fu); Monash University Eastern Health Clinical School, Melbourne, Australia (Davis)
| | - T K Choueiri
- United States: University of Utah Huntsman Cancer Institute, Salt Lake City, UT (Agarwal); University of Texas Southwestern Medical Center, Dallas, TX (Le); Stanford Medical Center, Stanford, CA (Srinivas); University of Michigan, Ann Arbor, MI (Alva); Dana-Farber Cancer Institute, Boston, MA (Choueiri)
| | - D Y C Heng
- Alberta: Tom Baker Cancer Center, University of Calgary, Calgary (Stukalin, Wells, Heng)
| |
Collapse
|
46
|
Ding B, Yan L, Zhang Y, Wang Z, Zhang Y, Xia D, Ye Z, Xu H. Analysis of the role of mutations in the KMT2D histone lysine methyltransferase in bladder cancer. FEBS Open Bio 2019; 9:693-706. [PMID: 30984543 PMCID: PMC6443872 DOI: 10.1002/2211-5463.12600] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 12/19/2022] Open
Abstract
Histone lysine methyltransferases (HMT) comprise a subclass of epigenetic regulators; dysregulation of these enzymes affects gene expression, which may lead to tumorigenesis. Here, we performed an integrated analysis of 50 HMTs in bladder cancer and found intrinsic links between copy number alterations, mutations, gene expression levels, and clinical outcomes. Through integrative analysis, we identified six HMT genes (PRDM9,ASH1L,SETD3,SETD5,WHSC1L1, and KMT2D) that may play a key role in the development and progression of bladder cancer. Of these six HMTs, histone lysine N‐methyltransferase 2D (KMT2D) exhibited the highest mutation rate in bladder cancer. Our comparison of the mRNA and miRNA expression profiles of mutated and wild‐type KMT2D suggested that two signaling pathways (FOX1–miR‐1224‐5p–DLK1 and HIF/GATA5–miR‐133a‐3p–DRD5) may mediate the tumor suppressive effect of the KMT2D mutation. In summary, our findings indicate that mutations in HMT genes, especially KMT2D mutation, may play a role in the development of bladder cancer.
Collapse
Affiliation(s)
- Beichen Ding
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Libin Yan
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Yucong Zhang
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Zhize Wang
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Yangjun Zhang
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Ding Xia
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Zhangqun Ye
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| | - Hua Xu
- Department of Urology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,Institute of Urology of Hubei Province Wuhan China
| |
Collapse
|
47
|
Kulkarni RA, Bak DW, Wei D, Bergholtz SE, Briney CA, Shrimp JH, Alpsoy A, Thorpe AL, Bavari AE, Crooks DR, Levy M, Florens L, Washburn MP, Frizzell N, Dykhuizen EC, Weerapana E, Linehan WM, Meier JL. A chemoproteomic portrait of the oncometabolite fumarate. Nat Chem Biol 2019; 15:391-400. [PMID: 30718813 PMCID: PMC6430658 DOI: 10.1038/s41589-018-0217-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 11/29/2018] [Indexed: 01/02/2023]
Abstract
Hereditary cancer disorders often provide an important window into novel mechanisms supporting tumor growth. Understanding these mechanisms thus represents a vital goal. Toward this goal, here we report a chemoproteomic map of fumarate, a covalent oncometabolite whose accumulation marks the genetic cancer syndrome hereditary leiomyomatosis and renal cell carcinoma (HLRCC). We applied a fumarate-competitive chemoproteomic probe in concert with LC-MS/MS to discover new cysteines sensitive to fumarate hydratase (FH) mutation in HLRCC cell models. Analysis of this dataset revealed an unexpected influence of local environment and pH on fumarate reactivity, and enabled the characterization of a novel FH-regulated cysteine residue that lies at a key protein-protein interface in the SWI-SNF tumor-suppressor complex. Our studies provide a powerful resource for understanding the covalent imprint of fumarate on the proteome and lay the foundation for future efforts to exploit this distinct aspect of oncometabolism for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Rhushikesh A Kulkarni
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Daniel W Bak
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Sarah E Bergholtz
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Chloe A Briney
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Jonathan H Shrimp
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Aktan Alpsoy
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Abigail L Thorpe
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Arissa E Bavari
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Michaella Levy
- Stowers Institute for Medical Research, Kansas City, MI, USA
| | | | - Michael P Washburn
- Stowers Institute for Medical Research, Kansas City, MI, USA.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KA, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | | | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Jordan L Meier
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MA, USA.
| |
Collapse
|
48
|
Mio C, Bulotta S, Russo D, Damante G. Reading Cancer: Chromatin Readers as Druggable Targets for Cancer Treatment. Cancers (Basel) 2019; 11:cancers11010061. [PMID: 30634442 PMCID: PMC6356452 DOI: 10.3390/cancers11010061] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/30/2018] [Accepted: 01/03/2019] [Indexed: 02/07/2023] Open
Abstract
The epigenetic machinery deputed to control histone post-translational modifications is frequently dysregulated in cancer cells. With epigenetics being naturally reversible, it represents a good target for therapies directed to restore normal gene expression. Since the discovery of Bromodomain and Extra Terminal (BET) inhibitors, a great effort has been spent investigating the effects of chromatin readers’ inhibition, specifically the class of proteins assigned to bind acetylated and methylated residues. So far, focused studies have been produced on epigenetic regulation, dissecting a specific class of epigenetic-related proteins or investigating epigenetic therapy in a specific tumor type. In this review, recent steps toward drug discovery on the different classes of chromatin readers have been outlined, highlighting the pros and cons of current therapeutic approaches.
Collapse
Affiliation(s)
- Catia Mio
- Department of Medical Area, University of Udine, 33100 Udine, Italy.
| | - Stefania Bulotta
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy.
| | - Diego Russo
- Department of Health Sciences, "Magna Graecia" University of Catanzaro, 88100 Catanzaro, Italy.
| | - Giuseppe Damante
- Department of Medical Area, University of Udine, 33100 Udine, Italy.
- Medical Genetics Institute, University Hospital of Udine, 33100 Udine, Italy.
| |
Collapse
|
49
|
Prognostic and Predictive Markers, and Stratifications Tables, for the Detection and Treatment of Renal Cell Carcinoma. Urol Oncol 2019. [DOI: 10.1007/978-3-319-42623-5_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
50
|
Jin S, Wu J, Zhu Y, Gu W, Wan F, Xiao W, Dai B, Zhang H, Shi G, Shen Y, Zhu Y, Ye D. Comprehensive Analysis of BAP1 Somatic Mutation in Clear Cell Renal Cell Carcinoma to Explore Potential Mechanisms in Silico. J Cancer 2018; 9:4108-4116. [PMID: 30519310 PMCID: PMC6277624 DOI: 10.7150/jca.27281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/24/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose: Aim of this study was to comprehensively analyze BRCA1-associated protein-1 (BAP1) somatic mutation in clear cell renal cell carcinoma (ccRCC) and explore potential therapeutic pathways and molecules. Patients and methods: In this study, we analyzed 445 ccRCC cases from The Cancer Genome Atlas (TCGA). Comprehensive analysis including survival, transcriptome and methylation between BAP1 mutated and wild-type cases was performed using bioinformatics tools in silico. Pathways and molecules related to BAP1 mutation were analyzed using Database for Annotation, Visualization and Integrated Discovery (DAVID) and protein-protein interaction (PPI) network. Results: BAP1 mutated ccRCC patients had a worse overall survival (OS) and disease free survival (DFS) than BAP1 wild-type patients. We found 583 up-regulated and 1216 down-regulated different expressed genes (DEGs) in BAP1 mutated tumors. Up-regulated DEGs were enriched in molecular functions and biological processes like protein binding, protein transport and ubiquitin protein ligase binding. Down-regulated DEGs were enriched in pathways like Rap1 signaling pathway, Notch pathway and altered molecular functions like metal ion binding and ubiquitin-protein transferase activity. Furthermore, CAD, TSPO, CTNNB1 and MAPK3 were top hub genes selected using PPI network analysis. Finally, BAP1 mutation had a strong correlation with CpG island methylator phenotype (CIMP). Conclusion: Our study provides a comprehensive understanding of BAP1 functional somatic mutation in ccRCC patients. Several hub genes like CAD and TSPO may become potential therapeutic targets.
Collapse
Affiliation(s)
- Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weijie Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjun Xiao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yijun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|