51
|
Ha MJ, Banerjee S, Akbani R, Liang H, Mills GB, Do KA, Baladandayuthapani V. Personalized Integrated Network Modeling of the Cancer Proteome Atlas. Sci Rep 2018; 8:14924. [PMID: 30297783 PMCID: PMC6175854 DOI: 10.1038/s41598-018-32682-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Personalized (patient-specific) approaches have recently emerged with a precision medicine paradigm that acknowledges the fact that molecular pathway structures and activity might be considerably different within and across tumors. The functional cancer genome and proteome provide rich sources of information to identify patient-specific variations in signaling pathways and activities within and across tumors; however, current analytic methods lack the ability to exploit the diverse and multi-layered architecture of these complex biological networks. We assessed pan-cancer pathway activities for >7700 patients across 32 tumor types from The Cancer Proteome Atlas by developing a personalized cancer-specific integrated network estimation (PRECISE) model. PRECISE is a general Bayesian framework for integrating existing interaction databases, data-driven de novo causal structures, and upstream molecular profiling data to estimate cancer-specific integrated networks, infer patient-specific networks and elicit interpretable pathway-level signatures. PRECISE-based pathway signatures, can delineate pan-cancer commonalities and differences in proteomic network biology within and across tumors, demonstrates robust tumor stratification that is both biologically and clinically informative and superior prognostic power compared to existing approaches. Towards establishing the translational relevance of the functional proteome in research and clinical settings, we provide an online, publicly available, comprehensive database and visualization repository of our findings ( https://mjha.shinyapps.io/PRECISE/ ).
Collapse
Affiliation(s)
- Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sayantan Banerjee
- Operations Management and Quantitative, Techniques Area at the Indian Institute of Management, Indore, India
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Gordon B Mills
- Oregon Health and Science University, Portland, OR, 97239, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | |
Collapse
|
52
|
Richard C, Niogret J, Boidot R, Ghiringhelli F. EGFR amplification induces sensitivity to anti EGFR therapy in pancreatic acinar cell carcinoma. World J Gastrointest Oncol 2018; 10:103-107. [PMID: 29666669 PMCID: PMC5900454 DOI: 10.4251/wjgo.v10.i4.103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic acinar cell carcinoma (PACC) is a rare cancer. When the tumor is metastatic, few therapeutic options are available. Precision medicine using next-generation sequencing is defined by the administration of drugs based on the tumor genetic mutations. The usage of precision medicine for finding new therapeutic options for rare cancers is an emerging field. We have reported here the case of a patient bearing a multitreated metastatic PACC. This patient underwent somatic and constitutional exome analyses. The analyses revealed in the liver metastasis an amplification of the EGFR gene. Accordingly, the patient was treated with off-label usage of panitumumab. We observed rapid response with necrosis of the liver metastasis, while no efficacy was observed in the primary tumor. An exome analysis of the primary tumor revealed amplification of HER2 and MET with EGFR amplification. Such amplifications are known as a resistance mechanism to antiEGFR therapy. Our results suggest that exome analysis may be helpful to highlight targets in rare cancers, such as PACC. EGFR amplification in this pathology should be determined and could be used as a biomarker to propose antiEGFR therapy.
Collapse
Affiliation(s)
- Corentin Richard
- Platform of Transfer in Oncology, Center GF Leclerc, Dijon 21000, France
- INSERM U1231, Dijon 21000, France
- University of Bourgogne Franche Comte, Dijon 21000, France
| | - Julie Niogret
- Department of Medical Oncology, Georges Francois Leclerc Cancer Center, Dijon 21000, France
| | - Romain Boidot
- Platform of Transfer in Oncology, Center GF Leclerc, Dijon 21000, France
- INSERM U1231, Dijon 21000, France
| | - Francois Ghiringhelli
- Platform of Transfer in Oncology, Center GF Leclerc, Dijon 21000, France
- INSERM U1231, Dijon 21000, France
- University of Bourgogne Franche Comte, Dijon 21000, France
- Department of Medical Oncology, Georges Francois Leclerc Cancer Center, Dijon 21000, France
| |
Collapse
|
53
|
Pushkin AA, Burda YE, Sevast’yanov AA, Kulikovskiy VF, Burda SY, Golubinskaya PA, Zvyagina AK, Kulyushina NV. Renal cell carcinoma drug and cell therapy: today and tomorrow. RESEARCH RESULTS IN PHARMACOLOGY 2018. [DOI: 10.3897/rrpharmacology.4.25251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Today, considerable progress in the renal cell carcinoma (RCC) treatment has been made due to development of targeted and immunotherapeutic approaches to the RCC treatment, especially in metastasising carcinoma. In the early stages of RCC, it is possible to use partial or total surgical nephrectomy, but in metastases development, the range of efficient treatment methods is dramatically limited. Appearance of targeted drugs like PD-1 and CTLA-4 receptors and their ligands’ inhibitors in clinical practice has significantly increased the total survival rate of patients with renal cell carcinoma. Emergence of adoptive cell therapy has opened new possibilities and prospects in RCC treatment. Previously activated in vitro cells are used there, which provides antineoplastic activity. For example, it could be antigen-specific cytotoxic T-lymphocytes (CTL), lymphokine-activated natural killers (LAK-NK-cells) and tumour-infiltrating lymphocytes (TILs). In this review, the authors specified the main molecular markers, associated with RCC; and signalling pathways (VEGFR- and EGFR-signalling pathway), which directly take part in carcinogenesis. The paper also looks at clinically applicable targeted immune drugs and the principle of their effect on tumorous cells. Besides, modern clinical studies of cell drugs have been considered. At the moment, there are a number of variants of targeted and immune drugs for the metastatic RCC treatment. Patients have no opportunity to use all the available agents because of their cost and toxicity level. For the most efficient treatment of patients with diagnosed metastatic RCC, it is necessarily to carry out risk stratification and prognostic factors for the response to treatment.
Collapse
|
54
|
Nabi S, Kessler ER, Bernard B, Flaig TW, Lam ET. Renal cell carcinoma: a review of biology and pathophysiology. F1000Res 2018; 7:307. [PMID: 29568504 PMCID: PMC5850086 DOI: 10.12688/f1000research.13179.1] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2018] [Indexed: 01/13/2023] Open
Abstract
Over the past decade, our understanding of the biology and pathophysiology of renal cell carcinoma (RCC) has improved significantly. Insight into the disease process has helped us in developing newer therapeutic approaches toward RCC. In this article, we review the various genetic and immune-related mechanisms involved in the pathogenesis and development of this cancer and how that knowledge is being used to develop therapeutic targeted drugs for the treatment of RCC. The main emphasis of this review article is on the most common genetic alterations found in clear cell RCC and how various drugs are currently targeting such pathways. This article also looks at the role of the immune system in allowing the growth of RCC and how the immune system can be manipulated to reactivate cytotoxic immunity against RCC.
Collapse
Affiliation(s)
- Shahzaib Nabi
- Division of Medical Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO, USA
| | - Elizabeth R Kessler
- Division of Medical Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO, USA
| | - Brandon Bernard
- Division of Medical Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO, USA
| | - Thomas W Flaig
- Division of Medical Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO, USA
| | - Elaine T Lam
- Division of Medical Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
55
|
Högner A, Krause H, Jandrig B, Kasim M, Fuller TF, Schostak M, Erbersdobler A, Patzak A, Kilic E. PBRM1 and VHL expression correlate in human clear cell renal cell carcinoma with differential association with patient’s overall survival. Urol Oncol 2018; 36:94.e1-94.e14. [DOI: 10.1016/j.urolonc.2017.10.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 10/18/2022]
|
56
|
Yan F, Wang Y, Liu C, Zhao H, Zhang L, Lu X, Chen C, Wang Y, Lu T, Wang F. Identify clear cell renal cell carcinoma related genes by gene network. Oncotarget 2017; 8:110358-110366. [PMID: 29299153 PMCID: PMC5746388 DOI: 10.18632/oncotarget.22769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/03/2017] [Indexed: 12/26/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prominent type of kidney cancer in adults. The patients within metastatic ccRCC have a poor 5-year survival rate that is less than 10%. It is essential to identify ccRCC -related genes to help with the understanding of molecular mechanism of ccRCC. In this literature, we aim to identify genes related to ccRCC based on a gene network. We collected gene expression level data of ccRCC from the Cancer Genome Atlas (TCGA) for our analysis. We constructed a co-expression gene network as the first step of our study. Then, the network sparse boosting approach was performed to select the genes which are relevant to ccRCC. Results of our study show there are 15 genes selected from the all genes we collected. Among these genes, 7 of them have been demonstrated to play a key role in development and progression or in drug response of ccRCC. This finding offers clues of gene markers for the treatment of ccRCC.
Collapse
Affiliation(s)
- Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yue Wang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Chunhui Liu
- Zhongda Hospital Southeast University, Nanjing, P.R. China
| | - Huiling Zhao
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Liya Zhang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Xiaofan Lu
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Chen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yaoyan Wang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, P.R. China
| | - Fei Wang
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
57
|
Abstract
In 2011, Varela et al. reported that the PBRM1 gene is mutated in approximately 40% of clear cell renal cell carcinoma cases. Since then, the number of studies relating PBRM1 mutations to cancers has substantially increased. BAF180 has now been linked to more than 30 types of cancers, including ccRCC, cholangiocarcinomas, esophageal squamous cell carcinoma, bladder cancer, and breast cancer. The mutations associated with BAF180 are most often truncations, which result in a loss of protein expression. This loss has been shown to adversely affect the expression of genes, likely because BAF180 is the chromatin recognition subunit of PBAF. In addition, BAF180 functions in numerous DNA repair mechanisms. Its roles in mediating DNA repair are likely the mechanism by which BAF180 acts a tumor suppressor protein. As research on this protein gains more interest, scientists will begin to piece together the complicated puzzle of the BAF180 protein and why its loss often results in cancer.
Collapse
Affiliation(s)
- Sarah Hopson
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, Michigan 49931, United States
| | - Martin J. Thompson
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, Michigan 49931, United States
| |
Collapse
|
58
|
Kim BJ, Kim JH, Kim HS, Zang DY. Prognostic and predictive value of VHL gene alteration in renal cell carcinoma: a meta-analysis and review. Oncotarget 2017; 8:13979-13985. [PMID: 28103578 PMCID: PMC5355155 DOI: 10.18632/oncotarget.14704] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/28/2016] [Indexed: 11/25/2022] Open
Abstract
The von Hippel-Lindau (VHL) gene is often inactivated in sporadic renal cell carcinoma (RCC) by mutation or promoter hypermethylation. The prognostic or predictive value of VHL gene alteration is not well established. We conducted this meta-analysis to evaluate the association between the VHL alteration and clinical outcomes in patients with RCC. We searched PUBMED, MEDLINE and EMBASE for articles including following terms in their titles, abstracts, or keywords: 'kidney or renal', 'carcinoma or cancer or neoplasm or malignancy', 'von Hippel-Lindau or VHL', 'alteration or mutation or methylation', and 'prognostic or predictive'. There were six studies fulfilling inclusion criteria and a total of 633 patients with clear cell RCC were included in the study: 244 patients who received anti-vascular endothelial growth factor (VEGF) therapy in the predictive value analysis and 419 in the prognostic value analysis. Out of 663 patients, 410 (61.8%) had VHL alteration. The meta-analysis showed no association between the VHL gene alteration and overall response rate (relative risk = 1.47 [95% CI, 0.81-2.67], P = 0.20) or progression free survival (hazard ratio = 1.02 [95% CI, 0.72-1.44], P = 0.91) in patients with RCC who received VEGF-targeted therapy. There was also no correlation between the VHL alteration and overall survival (HR = 0.80 [95% CI, 0.56-1.14], P = 0.21). In conclusion, this meta-analysis indicates that VHL gene alteration has no prognostic or predictive value in patients with clear cell RCC.
Collapse
Affiliation(s)
- Bum Jun Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| | - Dae Young Zang
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Republic of Korea
| |
Collapse
|
59
|
Ge YZ, Xu LW, Zhou CC, Lu TZ, Yao WT, Wu R, Zhao YC, Xu X, Hu ZK, Wang M, Yang XB, Zhou LH, Zhong B, Xu Z, Li WC, Zhu JG, Jia RP. A BAP1 Mutation-specific MicroRNA Signature Predicts Clinical Outcomes in Clear Cell Renal Cell Carcinoma Patients with Wild-type BAP1. J Cancer 2017; 8:2643-2652. [PMID: 28900502 PMCID: PMC5595094 DOI: 10.7150/jca.20234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/25/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is the most prevalent histologic subtype of kidney cancers in adults, which could be divided into two distinct subgroups according to the BRCA1 associated protein-1 (BAP1) mutation status. In the current study, we comprehensively analyzed the genome-wide microRNA (miRNA) expression profiles in ccRCC, with the aim to identify the differentially expressed miRNAs between BAP1 mutant and wild-type tumors, and generate a BAP1 mutation-specific miRNA signature for ccRCC patients with wild-type BAP1. Methods: The BAP1 mutation status and miRNA profiles in BAP1 mutant and wild-type tumors were analyzed. Subsequently, the association of the differentially expressed miRNAs with patient survival was examined, and a BAP1 mutation-specific miRNA signature was generated and examined with Kaplan-Meier survival, univariate and multivariate Cox regression analyses. Finally, the bioinformatics methods were adopted for the target prediction of selected miRNAs and functional annotation analyses. Results: A total of 350 treatment-naïve primary ccRCC patients were selected from The Cancer Genome Atlas project, among which 35 (10.0%) subjects carried mutant BAP1 and had a shorter overall survival (OS) time. Furthermore, 33 miRNAs were found to be differentially expressed between BAP1 mutant and wild-type tumors, among which 11 (miR-149, miR-29b-2, miR-182, miR-183, miR-21, miR-365-2, miR-671, miR-365-1, miR-10b, miR-139, and miR-181a-2) were significantly associated with OS in ccRCC patients with wild-type BAP1. Finally, a BAP1 mutation-specific miRNA signature consisting of 11 miRNAs was generated and validated as an independent prognostic parameter. Conclusions: In summary, our study identified a total of 33 miRNAs differentially expressed between BAP1 mutant and wild-type tumors, and generated a BAP1 mutation-specific miRNA signature including eleven miRNAs, which could serve as a novel prognostic biomarker for ccRCC patients with wild-type BAP1.
Collapse
Affiliation(s)
- Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Lu-Wei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Chang-Cheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Tian-Ze Lu
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, 41 Jianshe Road, Nantong 226006, China
| | - Wen-Tao Yao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Ran Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - You-Cai Zhao
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Xiao Xu
- Department of Radiation Oncology, JiangSu Armed Police General Hospital, 8 Jiangdu South Road, Yangzhou 225003, China
| | - Zhi-Kai Hu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Min Wang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Xiao-Bing Yang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liu-Hua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Bing Zhong
- Department of Urology, Huaian First People's Hospital, Nanjing Medical University, 6 Beijing West Road, Huaian 223300, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Wen-Cheng Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jia-Geng Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Rui-Peng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| |
Collapse
|
60
|
Expression of long non-coding RNA MFI2-AS1 is a strong predictor of recurrence in sporadic localized clear-cell renal cell carcinoma. Sci Rep 2017; 7:8540. [PMID: 28819235 PMCID: PMC5561098 DOI: 10.1038/s41598-017-08363-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/06/2017] [Indexed: 01/09/2023] Open
Abstract
Prediction of recurrence is a challenge for the development of adjuvant treatments in clear-cell renal cell carcinoma (ccRCC). In these tumors, expression of long non-coding RNAs (lncRNAs) are deregulated and closely associated with prognosis. Thus, we aimed to predict ccRCC recurrence risk using lncRNA expression. We identified prognostic lncRNAs in a training set of 351 localized ccRCCs from The Cancer Genome Atlas and validated lncRNA-based recurrence classification in an independent cohort of 167 localized ccRCCs. We identified lncRNA MFI2-AS1 as best candidate in the training set. In the validation cohort, MFI2-AS1 expression was independently associated with shorter disease-free survival (Hazard Ratio (HR) for relapse 3.5, p = 0.0001). Combined with Leibovich classification, MFI2-AS1 status improved prediction of recurrence (C-index 0.70) compared to MFI2-AS1 alone (0.67) and Leibovich classification alone (0.66). In patients with aggressive tumors (Leibovich ≥5), MFI2-AS1 expression was associated with dramatically increased risk of relapse (HR 12.16, p < 0.0001) compared to patients with undetectable MFI2-AS1 who had favorable outcomes. Compared to normal samples, MFI2-AS1 was upregulated in tumor tissue, and higher expression was associated with metastatic dissemination. Overall, MFI2-AS1 status improves patient stratification in localized ccRCC, which supports further integration of lncRNAs in molecular cancer classifications.
Collapse
|
61
|
Epigenome Aberrations: Emerging Driving Factors of the Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2017; 18:ijms18081774. [PMID: 28812986 PMCID: PMC5578163 DOI: 10.3390/ijms18081774] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/29/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), the most common form of Kidney cancer, is characterized by frequent mutations of the von Hippel-Lindau (VHL) tumor suppressor gene in ~85% of sporadic cases. Loss of pVHL function affects multiple cellular processes, among which the activation of hypoxia inducible factor (HIF) pathway is the best-known function. Constitutive activation of HIF signaling in turn activates hundreds of genes involved in numerous oncogenic pathways, which contribute to the development or progression of ccRCC. Although VHL mutations are considered as drivers of ccRCC, they are not sufficient to cause the disease. Recent genome-wide sequencing studies of ccRCC have revealed that mutations of genes coding for epigenome modifiers and chromatin remodelers, including PBRM1, SETD2 and BAP1, are the most common somatic genetic abnormalities after VHL mutations in these tumors. Moreover, recent research has shed light on the extent of abnormal epigenome alterations in ccRCC tumors, including aberrant DNA methylation patterns, abnormal histone modifications and deregulated expression of non-coding RNAs. In this review, we discuss the epigenetic modifiers that are commonly mutated in ccRCC, and our growing knowledge of the cellular processes that are impacted by them. Furthermore, we explore new avenues for developing therapeutic approaches based on our knowledge of epigenome aberrations of ccRCC.
Collapse
|
62
|
Su X, Zhang J, Mouawad R, Compérat E, Rouprêt M, Allanic F, Parra J, Bitker MO, Thompson EJ, Gowrishankar B, Houldsworth J, Weinstein JN, Tost J, Broom BM, Khayat D, Spano JP, Tannir NM, Malouf GG. NSD1 Inactivation and SETD2 Mutation Drive a Convergence toward Loss of Function of H3K36 Writers in Clear Cell Renal Cell Carcinomas. Cancer Res 2017; 77:4835-4845. [PMID: 28754676 DOI: 10.1158/0008-5472.can-17-0143] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/21/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022]
Abstract
Extensive dysregulation of chromatin-modifying genes in clear cell renal cell carcinoma (ccRCC) has been uncovered through next-generation sequencing. However, a scientific understanding of the cross-talk between epigenetic and genomic aberrations remains limited. Here we identify three ccRCC epigenetic clusters, including a clear cell CpG island methylator phenotype (C-CIMP) subgroup associated with promoter methylation of VEGF genes (FLT4, FLT1, and KDR). C-CIMP was furthermore characterized by silencing of genes related to vasculature development. Through an integrative analysis, we discovered frequent silencing of the histone H3 K36 methyltransferase NSD1 as the sole chromatin-modifying gene silenced by DNA methylation in ccRCC. Notably, tumors harboring NSD1 methylation were of higher grade and stage in different ccRCC datasets. NSD1 promoter methylation correlated with SETD2 somatic mutations across and within spatially distinct regions of primary ccRCC tumors. ccRCC harboring epigenetic silencing of NSD1 displayed a specific genome-wide methylome signature consistent with the NSD1 mutation methylome signature observed in Sotos syndrome. Thus, we concluded that epigenetic silencing of genes involved in angiogenesis is a hallmark of the methylator phenotype in ccRCC, implying a convergence toward loss of function of epigenetic writers of the H3K36 histone mark as a root feature of aggressive ccRCC. Cancer Res; 77(18); 4835-45. ©2017 AACR.
Collapse
Affiliation(s)
- Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jianping Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roger Mouawad
- Department of Medical Oncology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France.,Fondation AVEC Laboratory, Paris, France
| | - Eva Compérat
- Department of Pathology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Morgan Rouprêt
- Department of Urology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Frederick Allanic
- Department of Medical Oncology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France.,Fondation AVEC Laboratory, Paris, France
| | - Jérôme Parra
- Department of Urology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Marc-Olivier Bitker
- Department of Urology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Erika J Thompson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorg Tost
- Laboratory for Epigenetics and Environment, Centre National de Recherche en Genomique Humaine, CEA - Institut de Biologie Francois Jacob, Evry, France
| | - Bradley M Broom
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Khayat
- Department of Medical Oncology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Jean-Philippe Spano
- Department of Medical Oncology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel G Malouf
- Department of Medical Oncology, Groupe Hospitalier Pitié-Salpêtrière, University Pierre and Marie Curie (Paris VI), Institut Universitaire de Cancérologie, AP-HP, Paris, France.
| |
Collapse
|
63
|
Posadas EM, Limvorasak S, Figlin RA. Targeted therapies for renal cell carcinoma. Nat Rev Nephrol 2017; 13:496-511. [DOI: 10.1038/nrneph.2017.82] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
64
|
Minardi D, Lucarini G, Milanese G, Montironi R, Di Primio R. Prognostic role of BAP1 in pT1 clear cell carcinoma in partial nephrectomy specimens. Virchows Arch 2017; 471:99-105. [PMID: 28488170 DOI: 10.1007/s00428-017-2143-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/26/2017] [Accepted: 05/01/2017] [Indexed: 12/21/2022]
Abstract
BAP1 is a gene situated on chromosome 3p in a region that can be modified in renal cell carcinomas (RCCs). Mutations that cause loss of expression of BAP1 frequently occur in primary clear cell renal carcinoma (ccRCC). In a previous work, we observed that loss of nuclear BAP1 expression was crucial in ccRCC progression; in the current study, we investigated BAP1 expression in a large series of small conventional ccRCCs treated with partial nephrectomy, to assess a possible role as biomarker and the prognostic value in terms of patients' survival at long-term follow-up. One hundred sixty-two patients with single pT1 ccRCC were selected from those who underwent surgery at our Institute of Urology between 1987 and 2000. The features considered in this study were gender, age, tumor size, grade, incidence of metastasis, and patient-specific survival; they were correlated with immunohistochemical BAP1 nuclear expression in tumoral tissue. Median follow-up was 197.24 months (range 19 to 274); median survival was 125.34 months (range 5 to 274 months). None of our pT1 ccRCCs showed total loss of nuclear BAP1 staining; we found a significant negative correlation between nuclear BAP1 expression and tumor size and between nuclear BAP1 expression and grade. In small ccRCCs, nuclear BAP1 staining was not correlated with disease-specific 5-year survival.Our data confirm the established role of BAP1 as a tumor suppressor protein. This is the first report where BAP1 has been studied in pT1 ccRCC tumors, but nuclear BAP1 expression cannot help identify patients having high-risk disease in these patients.
Collapse
Affiliation(s)
- Daniele Minardi
- Dipartimento di Scienze Cliniche e Specialistiche - Sezione di Urologia, Università Politecnica delle Marche - Azienda Ospedaliero-Universitaria Ospedali Riuniti, via Conca, 71, Ancona, Italy.
| | - Guendalina Lucarini
- Dipartimento di Scienze Cliniche e Molecolari - Sezione di Istologia, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Milanese
- Dipartimento di Scienze Cliniche e Specialistiche - Sezione di Urologia, Università Politecnica delle Marche - Azienda Ospedaliero-Universitaria Ospedali Riuniti, via Conca, 71, Ancona, Italy
| | - Rodolfo Montironi
- Dipartimento di Scienze Biomediche e Sanità Pubblica - Sezione di Patologia e Istopatologia, Università Politecnica delle Marche - Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy
| | - Roberto Di Primio
- Dipartimento di Scienze Cliniche e Molecolari - Sezione di Istologia, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
65
|
Gu YF, Cohn S, Christie A, McKenzie T, Wolff N, Do QN, Madhuranthakam AJ, Pedrosa I, Wang T, Dey A, Busslinger M, Xie XJ, Hammer RE, McKay RM, Kapur P, Brugarolas J. Modeling Renal Cell Carcinoma in Mice: Bap1 and Pbrm1 Inactivation Drive Tumor Grade. Cancer Discov 2017; 7:900-917. [PMID: 28473526 DOI: 10.1158/2159-8290.cd-17-0292] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/29/2017] [Accepted: 05/02/2017] [Indexed: 12/22/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by BAP1 and PBRM1 mutations, which are associated with tumors of different grade and prognosis. However, whether BAP1 and PBRM1 loss causes ccRCC and determines tumor grade is unclear. We conditionally targeted Bap1 and Pbrm1 (with Vhl) in the mouse using several Cre drivers. Sglt2 and Villin proximal convoluted tubule drivers failed to cause tumorigenesis, challenging the conventional notion of ccRCC origins. In contrast, targeting with PAX8, a transcription factor frequently overexpressed in ccRCC, led to ccRCC of different grades. Bap1-deficient tumors were of high grade and showed greater mTORC1 activation than Pbrm1-deficient tumors, which exhibited longer latency. Disrupting one allele of the mTORC1 negative regulator, Tsc1, in Pbrm1-deficient kidneys triggered higher grade ccRCC. This study establishes Bap1 and Pbrm1 as lineage-specific drivers of ccRCC and histologic grade, implicates mTORC1 as a tumor grade rheostat, and suggests that ccRCCs arise from Bowman capsule cells.Significance: Determinants of tumor grade and aggressiveness across cancer types are poorly understood. Using ccRCC as a model, we show that Bap1 and Pbrm1 loss drives tumor grade. Furthermore, we show that the conversion from low grade to high grade can be promoted by activation of mTORC1. Cancer Discov; 7(8); 900-17. ©2017 AACR.See related commentary by Leung and Kim, p. 802This article is highlighted in the In This Issue feature, p. 783.
Collapse
Affiliation(s)
- Yi-Feng Gu
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shannon Cohn
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tiffani McKenzie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nicholas Wolff
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quyen N Do
- Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ananth J Madhuranthakam
- Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tao Wang
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anwesha Dey
- Department of Molecular Oncology, Genentech, South San Francisco, California
| | | | - Xian-Jin Xie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Robert E Hammer
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Renée M McKay
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas. .,Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas. .,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
66
|
Yigit N, Suarez LF, Roth LG, Orazi A, Tam W. CD4-Negative Variant of Cutaneous Blastic Plasmacytoid Dendritic Cell Neoplasm With a Novel PBRM1 Mutation in an 11-Year-Old Girl. Am J Clin Pathol 2017; 147:453-460. [PMID: 28419186 DOI: 10.1093/ajcp/aqx012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES We report a rare case of CD4- cutaneous blastic plasmacytoid dendritic cell neoplasm (BPDCN) with a novel PBRM1 mutation. METHODS An 11-year-old girl presented with an enlarged mass on her left arm and underwent an incisional biopsy. RESULTS Histopathologic examination and immunohistochemistry studies showed a monotonous proliferation of blasts that were CD4-, CD56+, and CD123+. There was no evidence of leukemic dissemination. Next-generation sequencing detected PBRM1 and CIC gene abnormalities. We confirmed and validated a novel PBRM1 mutation by conventional polymerase chain reaction and Sanger sequencing. CONCLUSIONS CD4- variant of BPDCN may be mistaken for myeloid sarcoma or extramedullary lymphoblastic leukemia/lymphoma because of their overlapping morphologic and immunophenotypic features; thus, a careful clinicopathologic evaluation is essential to reach the correct diagnosis. PBRM1 mutation seems to be a driver event in this case. Our study underscores the importance of alterations in chromatin remodeling in the pathogenesis of BPDCN.
Collapse
Affiliation(s)
- Nuri Yigit
- Division of Hematopathology, Department of Pathology and Laboratory Medicine
- Department of Pathology, Gulhane Military Medical Academy and School of Medicine, Kecioren, Ankara, Turkey
| | | | - Lisa Giulino Roth
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Attilio Orazi
- Division of Hematopathology, Department of Pathology and Laboratory Medicine
| | - Wayne Tam
- Division of Hematopathology, Department of Pathology and Laboratory Medicine
| |
Collapse
|
67
|
Wang Y, Gao W, Xu J, Zhu Y, Liu L. The long noncoding RNA urothelial carcinoma-associated 1 overexpression as a poor prognostic biomarker in clear cell renal cell carcinoma. Tumour Biol 2017; 39:1010428317698377. [PMID: 28459210 DOI: 10.1177/1010428317698377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNA urothelial carcinoma-associated 1 has previously played important roles in cancer. However, its role is still unknown in clear cell renal cell carcinoma. We utilized the most recent molecular and clinical data of clear cell renal cell carcinoma from The Cancer Genome Atlas project, and the relationship between urothelial carcinoma-associated 1 expression and the clinicopathological features was analyzed. Our results indicated that urothelial carcinoma-associated 1 overexpression was associated with male ( p = 0.003), wild-type PBRM1 ( p = 0.021), and BAP1 mutation ( p = 0.022) in clear cell renal cell carcinoma, although lower expression was found in tumors compared with normal controls, validated in tumor tissues from The Cancer Genome Atlas and 21 clear cell renal cell carcinoma patients at our hospital. Moreover, urothelial carcinoma-associated 1 overexpression indicated poor prognosis independently (Hazard Ratio [HR]: 1.92, p = 0.000) in clear cell renal cell carcinoma; it might be a potential detrimental gene considered as a predictive biomarker involved in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Yang Wang
- Department of Oncology, Nanjing Medical University, Nanjing, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jiali Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Yizhi Zhu
- Department of Oncology, Nanjing Medical University, Nanjing, China
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
68
|
Mammalian SWI/SNF complexes in cancer: emerging therapeutic opportunities. Curr Opin Genet Dev 2017; 42:56-67. [PMID: 28391084 DOI: 10.1016/j.gde.2017.02.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/23/2017] [Accepted: 02/07/2017] [Indexed: 02/08/2023]
Abstract
Mammalian SWI/SNF (BAF) chromatin remodeling complexes orchestrate a diverse set of chromatin alterations which impact transcriptional output. Recent whole-exome sequencing efforts have revealed that the genes encoding subunits of mSWI/SNF complexes are mutated in over 20% of cancers, spanning a wide range of tissue types. The majority of mutations result in loss of subunit protein expression, implicating mSWI/SNF subunits as tumor suppressors. mSWI/SNF-deficient cancers remain a therapeutic challenge, owing to a lack of potent and selective agents which target complexes or unique pathway dependencies generated by mSWI/SNF subunit perturbations. Here, we review the current landscape of mechanistic insights and emerging therapeutic opportunities for human malignancies driven by mSWI/SNF complex perturbation.
Collapse
|
69
|
Liu L, Guo R, Zhang X, Liang Y, Kong F, Wang J, Xu Z. Loss of SETD2, but not H3K36me3, correlates with aggressive clinicopathological features of clear cell renal cell carcinoma patients. Biosci Trends 2017; 11:214-220. [PMID: 28260718 DOI: 10.5582/bst.2016.01228] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recent studies facilitated by DNA sequencing identified the histone modifying gene SETD2 as the second most frequent mutant gene in sporadic clear cell renal cell carcinoma (ccRCC) patients. SETD2 functions as a tumor suppressor in ccRCC. However, its clinical association and biological functions are not fully delineated. The aim of this study is to evaluate the clinical significance of SETD2 in ccRCC patients. SETD2 and its canonical histone modification product H3K36me3 were analyzed by immunohistochemistry (IHC) in 155 ccRCC patients from two independent cohorts retrospectively. Both SETD2 and H3K36me3 were heterogeneously stained and down-regulated in ccRCC tissues, compared with normal controls. The SETD2 protein deficiency rate was 34.07%, which is much higher than the reported SETD2 gene inactive mutation rate. Furthermore, low SETD2 protein expression, but not H3K36me3 expression, was associated with the aggressive phenotype of ccRCC patients. In addition, cox multivariate analysis identified low SETD2 protein expression as an independent prognostic factor for overall survival of ccRCC patients. Consistently, using RNA-Seq data of ccRCC patients from The Cancer Genome Atlas, we validated our findings that low SETD2 mRNA expression is significantly associated with the aggressive phenotypes, and predicted a worse outcome for ccRCC patients. In conclusion, our study demonstrated a massive down-regulation of SETD2 protein in ccRCC, and identified SETD2 protein, but not H3K36me3, as an independent good prognostic marker, which warrants further study focusing on the non-methyltransferase role of SETD2 in kidney tumor biology.
Collapse
Affiliation(s)
- Lei Liu
- Institute of Basic Medical Science and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University.,Department of Urology, Weihai Municipal Hospital
| | - Renbo Guo
- Institute of Basic Medical Science and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University.,Department of Urology, Shandong Cancer Hospital and Institute
| | - Xiang Zhang
- Institute of Basic Medical Science and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University.,Department of Urology, Qilu Hospital of Shandong University
| | - Yiran Liang
- Institute of Basic Medical Science and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University.,Department of Breast Surgery, Qilu Hospital of Shandong University
| | - Feng Kong
- Central Laboratory, The Second Hospital of Shandong University
| | - Jue Wang
- Central Laboratory, The Second Hospital of Shandong University
| | - Zhonghua Xu
- Institute of Basic Medical Science and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University.,Department of Urology, Qilu Hospital of Shandong University
| |
Collapse
|
70
|
Poplawski P, Rybicka B, Boguslawska J, Rodzik K, Visser TJ, Nauman A, Piekielko-Witkowska A. Induction of type 1 iodothyronine deiodinase expression inhibits proliferation and migration of renal cancer cells. Mol Cell Endocrinol 2017; 442:58-67. [PMID: 27940296 DOI: 10.1016/j.mce.2016.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 11/29/2016] [Accepted: 12/07/2016] [Indexed: 01/12/2023]
Abstract
Type 1 iodothyronine deiodinase (DIO1) regulates peripheral metabolism of thyroid hormones that control cellular proliferation, differentiation and metabolism. The significance of DIO1 in cancer is unknown. In this study we hypothesized that diminished expression of DIO1, observed in renal cancer, contributes to the carcinogenic process in the kidney. Here, we demonstrate that ectopic expression of DIO1 in renal cancer cells changes the expression of genes controlling cell cycle, including cyclin E1 and E2F5, and results in inhibition of proliferation. The expression of genes encoding collagens (COL1A1, COL4A2, COL5A1), integrins (ITGA4, ITGA5, ITGB3) and transforming growth factor-β-induced (TGFBI) is significantly altered in renal cancer cells with induced expression of DIO1. Finally, we show that overexpression of DIO1 inhibits migration of renal cancer cells. In conclusion, we demonstrate for the first time that loss of DIO1 contributes to renal carcinogenesis and that its induced expression protects cells against cancerous proliferation and migration.
Collapse
Affiliation(s)
- Piotr Poplawski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Beata Rybicka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Joanna Boguslawska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Katarzyna Rodzik
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland
| | - Theo J Visser
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Alicja Nauman
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland; Laboratory of Human Cancer Genetics, Centre of New Technologies, CENT, University of Warsaw, 02-089, Warsaw, Poland
| | - Agnieszka Piekielko-Witkowska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813, Warsaw, Poland.
| |
Collapse
|
71
|
Misumi K, Hayashi A, Shibahara J, Arita J, Sakamoto Y, Hasegawa K, Kokudo N, Fukayama M. Intrahepatic cholangiocarcinoma frequently shows loss of BAP1 and PBRM1 expression, and demonstrates specific clinicopathological and genetic characteristics with BAP1 loss. Histopathology 2017; 70:766-774. [PMID: 27864835 DOI: 10.1111/his.13127] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/16/2016] [Indexed: 12/15/2022]
Abstract
AIMS BAP1 and PBRM1 expression loss has been observed in multiple cancers, including intrahepatic cholangiocarcinoma (ICC). We investigated BAP1 and PBRM1 expression in ICC using immunohistochemistry, and analysed its association with clinicopathological and genetic features, including two histological subtypes. METHODS AND RESULTS Whole-section slides of 108 consecutive primary ICC cases were immunostained against BAP1 and PBRM1. Complete loss of BAP1 and PBRM1 was observed in 21 (19.4%) and 25 (23.1%) cases, respectively, and partial loss was identified in four (3.7%) and nine (8.4%) cases. In all cases, normal bile ducts were strongly and diffusely positive for both BAP1 and PBRM1. ICC with BAP1 loss showed lower serum CA19-9 levels, less perineural invasion, rare mucin production, weaker immunoreactivity against S-100P and stronger immunoreactivity against N-cadherin and NCAM. IDH mutations were identified more frequently in ICCs with BAP1 loss. All ICC with BAP1 loss corresponded to small-duct type ICC. Multivariate Cox regression analysis showed that BAP1 loss was an independent prognostic factor for both overall and recurrence-free survival (P < 0.05). Conversely, PBRM1 loss was found in both small-duct type and large-duct type ICC, and was not associated significantly with any specific characteristics, including prognosis. CONCLUSION BAP1 and PBRM1 loss is seen frequently in ICC. ICC with BAP1 loss shares features of small-duct type ICC.
Collapse
Affiliation(s)
- Kento Misumi
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akimasa Hayashi
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junji Shibahara
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Arita
- Hepato-Biliary-Pancreatic Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Sakamoto
- Hepato-Biliary-Pancreatic Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norihiro Kokudo
- Hepato-Biliary-Pancreatic Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
72
|
Prognostic and Predictive Markers, and Stratifications Tables, for the Detection and Treatment of Renal Cell Carcinoma. Urol Oncol 2017. [DOI: 10.1007/978-3-319-42603-7_57-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
73
|
Marquez-Vilendrer SB, Rai SK, Gramling SJ, Lu L, Reisman DN. BRG1 and BRM loss selectively impacts RB and P53, respectively: BRG1 and BRM have differential functions in vivo. Oncoscience 2016; 3:337-350. [PMID: 28105458 PMCID: PMC5235922 DOI: 10.18632/oncoscience.333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/23/2016] [Indexed: 12/11/2022] Open
Abstract
The SWI/SNF complex is an important regulator of gene expression that functions by interacting with a diverse array of cellular proteins. The catalytic subunits of SWI/SNF, BRG1 and BRM, are frequently lost alone or concomitantly in a range of different cancer types. This loss abrogates SWI/SNF complex function as well as the functions of proteins that are required for SWI/SNF function, such as RB1 and TP53. Yet while both proteins are known to be dependent on SWI/SNF, we found that BRG1, but not BRM, is functionally linked to RB1, such that loss of BRG1 can directly or indirectly inactivate the RB1 pathway. This newly discovered dependence of RB1 on BRG1 is important because it explains why BRG1 loss can blunt the growth-inhibitory effect of tyrosine kinase inhibitors (TKIs). We also observed that selection for Trp53 mutations occurred in Brm-positive tumors but did not occur in Brm-negative tumors. Hence, these data indicate that, during cancer development, Trp53 is functionally dependent on Brm but not Brg1. Our findings show for the first time the key differences in Brm- and Brg1-specific SWI/SNF complexes and help explain why concomitant loss of Brg1 and Brm frequently occurs in cancer, as well as how their loss impacts cancer development.
Collapse
Affiliation(s)
| | - Sudhir K Rai
- Department of Hematology/Oncology, Medicine, University of Florida, Gainesville, FL, USA
| | - Sarah Jb Gramling
- Department of Hematology/Oncology, Medicine, University of Florida, Gainesville, FL, USA
| | - Li Lu
- Department of Hematology/Oncology, Medicine, University of Florida, Gainesville, FL, USA; Department of Pathology, University of Florida, Gainesville, FL, USA
| | - David N Reisman
- Department of Hematology/Oncology, Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
74
|
Brodaczewska KK, Szczylik C, Fiedorowicz M, Porta C, Czarnecka AM. Choosing the right cell line for renal cell cancer research. Mol Cancer 2016; 15:83. [PMID: 27993170 PMCID: PMC5168717 DOI: 10.1186/s12943-016-0565-8] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 11/30/2016] [Indexed: 01/08/2023] Open
Abstract
Cell lines are still a tool of choice for many fields of biomedical research, including oncology. Although cancer is a very complex disease, many discoveries have been made using monocultures of established cell lines. Therefore, the proper use of in vitro models is crucial to enhance our understanding of cancer. Therapeutics against renal cell cancer (RCC) are also screened with the use of cell lines. Multiple RCC in vitro cultures are available, allowing in vivo heterogeneity in the laboratory, but at the same time, these can be a source of errors. In this review, we tried to sum up the data on the RCC cell lines used currently. An increasing amount of data on RCC shed new light on the molecular background of the disease; however, it revealed how much still needs to be done. As new types of RCC are being distinguished, novel cell lines and the re-exploration of old ones seems to be indispensable to create effective in vitro tools for drug screening and more.
Collapse
Affiliation(s)
- Klaudia K Brodaczewska
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Michal Fiedorowicz
- Department of Experimental Pharmacology, Polish Academy of Science Medical Research Centre, Warsaw, Poland
| | - Camillo Porta
- Department of Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| |
Collapse
|
75
|
Razafinjatovo C, Bihr S, Mischo A, Vogl U, Schmidinger M, Moch H, Schraml P. Characterization of VHL missense mutations in sporadic clear cell renal cell carcinoma: hotspots, affected binding domains, functional impact on pVHL and therapeutic relevance. BMC Cancer 2016; 16:638. [PMID: 27530247 PMCID: PMC4987997 DOI: 10.1186/s12885-016-2688-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 08/08/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The VHL protein (pVHL) is a multiadaptor protein that interacts with more than 30 different binding partners involved in many oncogenic processes. About 70 % of clear cell renal cell carcinoma (ccRCC) have VHL mutations with varying impact on pVHL function. Loss of pVHL function leads to the accumulation of Hypoxia Inducible Factor (HIF), which is targeted by current targeted treatments. In contrast to nonsense and frameshift mutations that highly likely nullify pVHL multipurpose functions, missense mutations may rather specifically influence the binding capability of pVHL to its partners. The affected pathways may offer predictive clues to therapy and response to treatment. In this study we focused on the VHL missense mutation pattern in ccRCC, and studied their potential effects on pVHL protein stability and binding partners and discussed treatment options. METHODS We sequenced VHL in 360 sporadic ccRCC FFPE samples and compared observed and expected frequency of missense mutations in 32 different binding domains. The prediction of the impact of those mutations on protein stability and function was assessed in silico. The response to HIF-related, anti-angiogenic treatment of 30 patients with known VHL mutation status was also investigated. RESULTS We identified 254 VHL mutations (68.3 % of the cases) including 89 missense mutations (35 %). Codons Ser65, Asn78, Ser80, Trp117 and Leu184 represented hotspots and missense mutations in Trp117 and Leu 184 were predicted to highly destabilize pVHL. About 40 % of VHL missense mutations were predicted to cause severe protein malfunction. The pVHL binding domains for HIF1AN, BCL2L11, HIF1/2α, RPB1, PRKCZ, aPKC-λ/ι, EEF1A1, CCT-ζ-2, and Cullin2 were preferentially affected. These binding partners are mainly acting in transcriptional regulation, apoptosis and ubiquitin ligation. There was no correlation between VHL mutation status and response to treatment. CONCLUSIONS VHL missense mutations may exert mild, moderate or strong impact on pVHL stability. Besides the HIF binding domain, other pVHL binding sites seem to be non-randomly altered by missense mutations. In contrast to LOF mutations that affect all the different pathways normally controlled by pVHL, missense mutations may be rather appropriate for designing tailor-made treatment strategies for ccRCC.
Collapse
Affiliation(s)
| | - Svenja Bihr
- Oncology Clinic, University Hospital Zurich, Zurich, Switzerland
| | - Axel Mischo
- Oncology Clinic, University Hospital Zurich, Zurich, Switzerland
| | - Ursula Vogl
- Department of Medicine, St. Joseph Hospital Vienna, Vienna, Austria
| | - Manuela Schmidinger
- Department of Medicine I, Clinical Division of Oncology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Peter Schraml
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
76
|
Paulucci DJ, Sfakianos JP, Yadav SS, Badani KK. BAP1 is overexpressed in black compared with white patients with Mx-M1 clear cell renal cell carcinoma: A report from the cancer genome atlas. Urol Oncol 2016; 34:259.e9-259.e14. [DOI: 10.1016/j.urolonc.2015.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/15/2015] [Accepted: 12/30/2015] [Indexed: 12/17/2022]
|
77
|
Minardi D, Lucarini G, Milanese G, Di Primio R, Montironi R, Muzzonigro G. Loss of nuclear BAP1 protein expression is a marker of poor prognosis in patients with clear cell renal cell carcinoma. Urol Oncol 2016; 34:338.e11-8. [PMID: 27085487 DOI: 10.1016/j.urolonc.2016.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/24/2016] [Accepted: 03/11/2016] [Indexed: 01/12/2023]
Abstract
INTRODUCTION BRCA1-associated protein 1 (BAP1) is a gene situated on chromosome 3p in a region that is deleted in more than 90% of renal cell carcinomas (RCCs). In the present study, we studied BAP1 immunohistochemical expression in a large series of conventional clear cell RCCs (ccRCCs) treated with radical nephrectomy; we assessed the prognostic value of their expression in terms of patients׳ survival at long-term follow-up. MATERIALS AND METHODS A total of 154 consecutive patients with ccRCC were selected from a prospective database and considered for the study purpose; all patients were treated with radical nephrectomy and lymphadenectomy at our Institute of Urology between 1983 and 1985. The features considered in this study were tumor size, grade and stage, vascular and capsular invasion, incidence of metastasis, and patient-specific survival; all these parameters were correlated with immunohistochemical cytoplasmic and nuclear expression of BAP1 in tumoral tissue. RESULTS Median follow-up was 196.18 months and median survival was 125.34 months. Nuclear BAP1 expression showed a high frequency of loss in tumoral cells; nuclear BAP1-negative tumors had higher tumor size, higher Fuhrman grade, and higher stage, a greater amount of vascular and capsular invasion and a higher incidence of metastases. In multivariate analysis, pathological stage and nuclear BAP1 expression resulted independent prognostic factors. CONCLUSION We have demonstrated that nuclear BAP1 expression is a marker of prognosis in ccRCC, having an influence on cancer-specific survival. The clinical importance for BAP1 will be realized with the identification and application of targeted therapies and with individualized approaches in the adjuvant setting or in the metastatic setting or in both the settings.
Collapse
Affiliation(s)
- Daniele Minardi
- Dipartimento di Scienze Cliniche e Specialistiche-Sezione di Urologia, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy.
| | - Guendalina Lucarini
- Dipartimento di Scienze Cliniche e Molecolari-Sezione di Istologia, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Milanese
- Dipartimento di Scienze Cliniche e Specialistiche-Sezione di Urologia, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy
| | - Roberto Di Primio
- Dipartimento di Scienze Cliniche e Molecolari-Sezione di Istologia, Università Politecnica delle Marche, Ancona, Italy
| | - Rodolfo Montironi
- Dipartimento di Scienze Biomediche e Sanità Pubblica-Sezione di Patologia e Istopatologia, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy
| | - Giovanni Muzzonigro
- Dipartimento di Scienze Cliniche e Specialistiche-Sezione di Urologia, Università Politecnica delle Marche-Azienda Ospedaliero-Universitaria Ospedali Riuniti, Ancona, Italy
| |
Collapse
|
78
|
Abstract
The diagnosis and management of renal cell carcinoma have changed remarkably rapidly. Although the incidence of renal cell carcinoma has been increasing, survival has improved substantially. As incidental diagnosis of small indolent cancers has become more frequent, active surveillance, robot-assisted nephron-sparing surgical techniques, and minimally invasive procedures, such as thermal ablation, have gained popularity. Despite progression in cancer control and survival, locally advanced disease and distant metastases are still diagnosed in a notable proportion of patients. An integrated management strategy that includes surgical debulking and systemic treatment with well established targeted biological drugs has improved the care of patients. Nevertheless, uncertainties, controversies, and research questions remain. Further advances are expected from translational and clinical studies.
Collapse
Affiliation(s)
- Umberto Capitanio
- Department of Urology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Francesco Montorsi
- Division of Experimental Oncology, URI, Urological Research Institute, Renal Cancer Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
79
|
Stanculeanu DL, Lazescu A, Zob DD, Bunghez R, Anghel R, Poteca TD. Metastatic clear cell renal carcinoma - an unusual response to Temsirolimus in second line therapy. J Med Life 2016; 9:193-8. [PMID: 27453754 PMCID: PMC4863514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Renal cell carcinoma (RCC) represents 3% of all cancers, with the highest incidence occurring in the most developed countries and representing the seventh most common cancer in men and the ninth most common cancer in women. The understanding of the tumor molecular biology and the discovery of new drugs that target molecular pathways have increased the arsenal against advanced renal cell carcinoma and improved the outcomes in the patients suffering from these affections. Studying the molecular signaling that controls the tumor growth and the progression has led to the development of molecular therapies targeting the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways, resulting in a significant improvement in the overall survival and quality of life. Sunitinib represents an inhibitor of VEGFR 1-3, c-kit, FLT-3 and PDGFR. We present the case of a patient with metastatic clear cell RCC with a treatment effect following sequential VEGF and mTOR inhibitor treatment. Under sunitinib treatment, the patient had a progression free survival (PFS) of approximately 9 months, similar to the PFS observed in clinical trials. Sunitinib was well tolerated by this patient. Temsirolimus, an mTOR inhibitor, is currently only approved for the first-line treatment of mRCC patients with poor prognosis. This study analyzes a treatment effect of second line temsirolimus in a patient with metastatic renal cell carcinoma (mRCC).
Collapse
Affiliation(s)
| | - A Lazescu
- Institute of Oncology, Bucharest, Romania
| | - DD Zob
- Institute of Oncology, Bucharest, Romania
| | - R Bunghez
- Institute of Oncology, Bucharest, Romania
| | - R Anghel
- Institute of Oncology, Bucharest, Romania
| | - TD Poteca
- Institute of Oncology, Bucharest, Romania
| |
Collapse
|
80
|
Li H, Samawi H, Heng DY. The use of prognostic factors in metastatic renal cell carcinoma. Urol Oncol 2015; 33:509-16. [DOI: 10.1016/j.urolonc.2015.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/28/2015] [Accepted: 08/05/2015] [Indexed: 11/25/2022]
|
81
|
Abstract
Cancer is widely recognized as a genetic disease in which somatic mutations are sequentially accumulated to drive tumor progression. Although genomic landscape studies are informative for individual cancer types, a comprehensive comparative study of tumorigenic mutations across cancer types based on integrative data sources is still a pressing need. We systematically analyzed ~10(6) non-synonymous mutations extracted from COSMIC, involving ~8000 genome-wide screened samples across 23 major human cancers at both the amino acid and gene levels. Our analysis identified cancer-specific heterogeneity that traditional nucleotide variation analysis alone usually overlooked. Particularly, the amino acid arginine (R) turns out to be the most favorable target of amino acid alteration in most cancer types studied (P < 10(-9), binomial test), reflecting its important role in cellular physiology. The tumor suppressor gene TP53 is mutated exclusively with the HYDIN, KRAS, and PTEN genes in large intestine, lung, and endometrial cancers respectively, indicating that TP53 takes part in different signaling pathways in different cancers. While some of our analyses corroborated previous observations, others indicated relevant candidates with high priority for further experimental validation. Our findings have many ramifications in understanding the etiology of cancer and the underlying molecular mechanisms in particular cancers.
Collapse
|
82
|
Christinat Y, Krek W. Integrated genomic analysis identifies subclasses and prognosis signatures of kidney cancer. Oncotarget 2015; 6:10521-31. [PMID: 25826081 PMCID: PMC4496372 DOI: 10.18632/oncotarget.3294] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/08/2015] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To define robust miRNA-based molecular classifiers for human clear cell renal cell carcinoma (ccRCC) subgrouping and prognostication. EXPERIMENTAL DESIGN Multidimensional data of over 500 clear cell renal cell carcinoma (ccRCC) patients were retrieved from The Cancer Genome Atlas (TCGA) archive. Data analysis was based on a novel computational approach that selectively considers patients with extreme expression values of miRNAs to detect survival-associated molecular signatures. RESULTS Our in silico analysis unveiled a novel ccRCC-specific 5-miRNA (miR-10b, miR-21, miR-143, miR-183, and miR-192) signature able, when combined with information from conventional TNM staging and the age of the patient, to prognosticate ccRCC outcome more accurately than known ccRCC miRNA signatures or TNM staging alone. Furthermore, our approach revealed the existence of 6 distinct subgroups of ccRCC characterized by discrete differences in overall survival, tumor stage, and mutational spectra in key ccRCC tumor suppressor genes. It also demonstrated that BAP1 mutations correlate with tumor progression rather than overall survival. CONCLUSION Integrated analysis of multidimensional data from the TCGA archive allowed to draw a portrait of distinct molecular subclasses of human ccRCC and to define signatures for prognosticating disease outcome. Together, these results offer new prospects for more accurate stratification and prognostication of ccRCC.
Collapse
Affiliation(s)
- Yann Christinat
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Wilhelm Krek
- Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
83
|
Benusiglio PR, Couvé S, Gilbert-Dussardier B, Deveaux S, Le Jeune H, Da Costa M, Fromont G, Memeteau F, Yacoub M, Coupier I, Leroux D, Méjean A, Escudier B, Giraud S, Gimenez-Roqueplo AP, Blondel C, Frouin E, Teh BT, Ferlicot S, Bressac-de Paillerets B, Richard S, Gad S. A germline mutation inPBRM1predisposes to renal cell carcinoma. J Med Genet 2015; 52:426-30. [DOI: 10.1136/jmedgenet-2014-102912] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/04/2015] [Indexed: 12/22/2022]
|
84
|
Liao L, Testa JR, Yang H. The roles of chromatin-remodelers and epigenetic modifiers in kidney cancer. Cancer Genet 2015; 208:206-14. [PMID: 25873528 DOI: 10.1016/j.cancergen.2015.02.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/05/2015] [Accepted: 02/17/2015] [Indexed: 10/24/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the major subtype of kidney cancer that is characterized by frequent inactivation of the von Hippel-Lindau (VHL) gene in 80-90% of the tumors. Recent reports using massive parallel sequencing technologies have discovered additional cancer driver genes. PBRM1 was found to be mutated in about 40% of ccRCC tumors, whereas BAP1 and SETD2 were each mutated in about 10-15% of ccRCC tumors. JARID1C and UTX, two histone H3 demethylases, were also found to harbor mutations in ccRCC, albeit at lower rates. ccRCC tumors display a high degree of intra-tumoral heterogeneity, with some mutations present in all cancer cells (ubiquitous), whereas others are subclonal. The VHL mutations were always ubiquitous in the tumors; PBRM1 mutations were also ubiquitous but to a lesser extent. On the contrary, mutations in BAP1, SETD2, JARID1C, and UTX were all subclonal, meaning that they were present in a subset of cancer cells in a tumor. The prognostic value of PBRM1 mutations in ccRCC is still controversial, whereas BAP1 mutations were tightly linked to worse clinical outcomes in multiple studies. The molecular functions of these newly identified cancer driver genes are discussed, and they were known readers, writers, or erasers of histone marks on histone H2 and H3 tails that are very close to each other, suggesting that these factors might functionally interact and affect common pathways. The studies on these newly identified tumor suppressors will shed light on ccRCC tumorigenesis and development, and will likely lead to development of novel therapeutic interventions for ccRCC patients.
Collapse
Affiliation(s)
- Lili Liao
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Haifeng Yang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA.
| |
Collapse
|
85
|
Ho TH, Kapur P, Joseph RW, Serie DJ, Eckel-Passow JE, Parasramka M, Cheville JC, Wu KJ, Frenkel E, Rakheja D, Stefanius K, Brugarolas J, Parker AS. Loss of PBRM1 and BAP1 expression is less common in non-clear cell renal cell carcinoma than in clear cell renal cell carcinoma. Urol Oncol 2015; 33:23.e9-23.e14. [PMID: 25465300 PMCID: PMC4274200 DOI: 10.1016/j.urolonc.2014.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/02/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Recurrent mutations in polybromo-1 (PBRM1, ~40%) and BRCA1-associated protein-1 (BAP1, ~10%) occur in clear cell renal cell carcinoma (ccRCC), but their prevalence in non-ccRCC or renal oncocytoma (RO) is unknown. We evaluated loss of PBRM1 and BAP1 staining in ccRCC, papillary RCC (pRCC), chromophobe RCC (chRCC), and RO tumors using an immunohistochemistry assay in which negative staining was associated with loss-of-function mutations. METHODS We identified 458 patients treated surgically for ccRCC, pRCC, chRCC, and RO between 2004 and 2012. We performed immunohistochemistry assays to evaluate PBRM1 and BAP1 protein expression to classify tumors as PBRM1 or BAP1 negative. We compared loss of staining of these 2 proteins in ccRCC and non-ccRCC using the Fisher exact test. RESULTS For the total cohort of 458 patients, we successfully stained both PBRM1 and BAP1 in 408 tumor samples. Consistent with the mutation rate, loss of PBRM1 and BAP1 staining occurred in 43% (80/187) and 10% (18/187) of ccRCC cases, respectively. However, loss of PBRM1 staining occurred in only 3% (2/59), 6% (1/17), and 0% (0/34) of pRCC, chRCC, and RO tumors, respectively (P<0.0001). BAP1 loss was not observed in any of the pRCC (n = 61), chRCC (n = 17), or RO (n = 34) tumors, (P = 0.00021). CONCLUSION Our data suggest that biallelic inactivation of PBRM1 or BAP1 is less common in non-ccRCC when compared with ccRCC tumors. These findings suggest that loss of PBRM1 or BAP1 are key events in ccRCC, whereas other pathways may support tumorigenesis in non-ccRCC subtypes.
Collapse
Affiliation(s)
- Thai H Ho
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ; Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN.
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX; Kidney Cancer Program, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Richard W Joseph
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL
| | | | | | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Kevin J Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL
| | - Eugene Frenkel
- Kidney Cancer Program, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX; Kidney Cancer Program, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Karoliina Stefanius
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX; Kidney Cancer Program, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - James Brugarolas
- Kidney Cancer Program, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX; Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Alexander S Parker
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
86
|
Bhatt JR, Finelli A. Landmarks in the diagnosis and treatment of renal cell carcinoma. Nat Rev Urol 2014; 11:517-25. [PMID: 25112856 DOI: 10.1038/nrurol.2014.194] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common renal cancer is renal cell carcinoma (RCC), which arises from the renal parenchyma. The global incidence of RCC has increased over the past two decades by 2% per year. RCC is the most lethal of the common urological cancers: despite diagnostic advances, 20-30% of patients present with metastatic disease. A clearer understanding of the genetic basis of RCC has led to immune-based and targeted treatments for this chemoresistant cancer. Despite promising results in advanced disease, overall response rates and durable complete responses are rare. Surgery remains the main treatment modality, especially for organ-confined disease, with a selective role in advanced and metastatic disease. Smaller tumours are increasingly managed with biopsy, minimally invasive interventions and surveillance. The future promises multimodal, integrated and personalized care, with further understanding of the disease leading to new treatment options.
Collapse
Affiliation(s)
- Jaimin R Bhatt
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| | - Antonio Finelli
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
87
|
Lee-Ying R, Lester R, Heng DYC. Current management and future perspectives of metastatic renal cell carcinoma. Int J Urol 2014; 21:847-55. [PMID: 24862210 DOI: 10.1111/iju.12502] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/14/2014] [Indexed: 12/13/2022]
Abstract
Over the last number of years, the treatment of metastatic renal cell cancer has evolved tremendously with the advent of targeted therapy. Previously, immunotherapies, such as interferon alpha and interleukin-2, were the only treatment options available for this chemoresistant malignancy. Currently, seven additional agents, including sunitinib, sorafenib, axitinib, pazopanib, bevacizumab, everolimus and temsirolimus, have been approved for use in metastatic renal cell cancer, with several more in development. The efficacy of these agents depends primarily on inhibition of the vascular endothelial growth factor and mammalian target of rapamycin pathways, and have drastically improved the outcomes of patients diagnosed with metastatic renal cell cancer. This article reviews the major treatment advances that have occurred for metastatic renal cell cancer with the advent of targeted treatments, summarizes the evidence to support their use and addresses clinical issues that have arisen with them. To help guide clinicians in their decision-making with these emerging therapeutic choices, the evidence for sequencing and combining these agents, and the need for biomarkers will be addressed. The role of surgical management options, such as cytoreductive nephrectomy and metastectomy, in the era of targeted treatment is also reviewed. Several novel treatments are also on the horizon, which might serve as future avenues for treatment advancement in metastatic renal cell cancer.
Collapse
|
88
|
Abstract
Renal cell carcinoma of clear-cell type (ccRCC) is an enigmatic tumor type, characterized by frequent inactivation of the VHL gene (infrequently mutated in other tumor types), responsiveness to angiogenesis inhibitors, and resistance to both chemotherapy and conventional radiation therapy. ccRCC tumors exhibit substantial mutation heterogeneity. Recent studies using massively parallel sequencing technologies have implicated several novel driver genes. In VHL wild-type tumors, mutations were discovered in TCEB1, which encodes Elongin C, a protein that binds to VHL and is required for its function. Several additional tumor suppressor genes have been identified near the VHL gene, within a region that is frequently deleted in ccRCC on chromosome 3p: SETD2, BAP1, and PBRM1. Mutations in BAP1 and PBRM1 are largely mutually exclusive and are associated with different tumor biology and patient outcomes. In addition, the mTORC1 pathway is deregulated by mutations in MTOR, TSC1, PIK3CA, and PTEN in approximately 20% of ccRCCs. Mutations in TSC1, and possibly other genes, may predict for sensitivity to mTORC1 inhibitors. These discoveries provide insight into ccRCC development and set the foundation for the first molecular genetic classification of the disease, paving the way for subtype-specific therapies.
Collapse
Affiliation(s)
- James Brugarolas
- From the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
89
|
Joseph RW, Kapur P, Serie DJ, Eckel-Passow JE, Parasramka M, Ho T, Cheville JC, Frenkel E, Rakheja D, Brugarolas J, Parker A. Loss of BAP1 protein expression is an independent marker of poor prognosis in patients with low-risk clear cell renal cell carcinoma. Cancer 2014; 120:1059-67. [PMID: 24382589 PMCID: PMC4075029 DOI: 10.1002/cncr.28521] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/12/2013] [Accepted: 10/23/2013] [Indexed: 02/06/2023]
Abstract
BACKGROUND The majority of patients diagnosed with clear cell renal cell carcinoma (ccRCC) have low-risk disease with a < 10% chance of ccRCC-specific death. DNA sequencing revealed that mutations in BAP1 (BRCA1 associated protein-1) occur in 5% to 15% of ccRCC cases and are associated with poor outcomes. The vast majority of BAP1 mutations abolish protein expression. In this study, we used a highly sensitive and specific immunohistochemistry (IHC) assay to test whether BAP1 expression is an independent marker of ccRCC-specific survival, particularly in patients with low-risk disease. METHODS BAP1 expression was assessed, using IHC, in 1479 patients who underwent nephrectomy to treat clinically localized ccRCC. A centralized pathologist dichotomized patients as either BAP1-positive or BAP1-negative. The authors employed Kaplan-Meier and Cox regression models to associate BAP1 expression with cancer-specific survival. RESULTS A total of 10.5% of tumors were BAP1-negative, 84.8% of tumors were BAP1-positive, and 4.6% of tumors had ambiguous staining for BAP1. Patients with BAP1-negative tumors have an increased risk of ccRCC-related death (hazard ratio [HR] = 3.06; 95% confidence interval [CI] = 2.28-4.10; P = 6.77 × 10(-14) ). BAP1 expression remained an independent marker of prognosis after adjusting for the UCLA integrated staging system (UISS) (HR = 1.67; 95% CI = 1.24-2.25; P < .001). Finally, BAP1 was an independent prognostic marker in low-risk patients with a Mayo Clinic stage, size, grade, and necrosis (SSIGN) score of ≤ 3 (HR = 3.24; 95% CI = 1.26-8.33; P = .015). CONCLUSIONS This study used a large patient cohort to demonstrate that BAP1 expression is an independent marker of prognosis in patients with low-risk (SSIGN≤ 3) ccRCC.
Collapse
Affiliation(s)
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center
| | - Daniel J. Serie
- Departments of Health Sciences Research and Medicine, Mayo Clinic Florida
| | | | | | - Thai Ho
- Division of Medical Oncology, Mayo Clinic Arizona
| | | | - Eugene Frenkel
- Division of Hematology-Oncology, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center
| | - James Brugarolas
- Division of Hematology-Oncology, Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center
- Department of Developmental Biology, University of Texas Southwestern Medical Center
| | - Alexander Parker
- Departments of Health Sciences Research and Medicine, Mayo Clinic Florida
| |
Collapse
|
90
|
Upregulated microRNA-16 as an oncogene in renal cell carcinoma. Mol Med Rep 2012; 12:1399-404. [DOI: 10.3892/mmr.2015.3496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 02/11/2015] [Indexed: 11/05/2022] Open
|