51
|
Tarade D, Ohh M. The HIF and other quandaries in VHL disease. Oncogene 2017; 37:139-147. [PMID: 28925400 DOI: 10.1038/onc.2017.338] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022]
Abstract
Mutations in VHL underlie von Hippel-Lindau (VHL) disease, a hereditary cancer syndrome with several subtypes depending on the risk of developing certain combination of classic features, such as clear cell renal cell carcinoma (ccRCC), hemangioblastoma and pheochromocytoma. Although numerous potential substrates and functions of pVHL have been described over the past decade, the best-defined role of pVHL has remained as the negative regulator of the heterodimeric hypoxia-inducible factor (HIF) transcription factor via the oxygen-dependent ubiquitin-mediated degradation of HIF-α subunit. Despite the seminal discoveries that led to the molecular elucidation of the mammalian oxygen-sensing VHL-HIF axis, which have provided several rational therapies, the mechanisms underlying the complex genotype-phenotype correlation in VHL disease are unclear. This review will discuss and highlight the studies that have provided interesting insights as well as uncertainties to the underlying mechanisms governing VHL disease.
Collapse
Affiliation(s)
- D Tarade
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - M Ohh
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, MaRS Centre West Tower, Toronto, Ontario, Canada
| |
Collapse
|
52
|
Al-Khallaf H. Isocitrate dehydrogenases in physiology and cancer: biochemical and molecular insight. Cell Biosci 2017; 7:37. [PMID: 28785398 PMCID: PMC5543436 DOI: 10.1186/s13578-017-0165-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 01/31/2023] Open
Abstract
Isocitrate dehydrogenases play important roles in cellular metabolism and cancer. This review will discuss how the roles of isoforms 1 and 2 in normal cell and cancer metabolism are distinct from those of isoform 3. It will also explain why, unlike 1 and 2, mutations in isoform 3 in tumor are not likely to be driver ones. A model explaining two important features of isocitrate dehydrogenases 1 and 2 mutations, their dominant negative effect and their mutual exclusivity, will be provided. The importance of targeting these mutations and the possibility of augmenting such therapy by targeting other cancer-related pathways will also be discussed.
Collapse
Affiliation(s)
- Hamoud Al-Khallaf
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, 6830 Ammar Bin Thabit St, Al Muraikabat, Dammam, 32253 Saudi Arabia
| |
Collapse
|
53
|
Wang X, Cheng Y, Zhu Y, Li H, Ge W, Wu X, Zhao K, Yuan J, Li Z, Jiang S, Han Z, Jiang Q, Wu Q, Liu T, Zhang C, Yu M, Hu Y. Epigenetic silencing of ASPP1 confers 5‐FU resistance in clear cell renal cell carcinoma by preventing p53 activation. Int J Cancer 2017; 141:1422-1433. [DOI: 10.1002/ijc.30852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/07/2017] [Accepted: 06/21/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Xingwen Wang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Yiwei Cheng
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - YiFu Zhu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Huayi Li
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Wenjie Ge
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| | - Xiaoliang Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Kunming Zhao
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Jinyang Yuan
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Zhenglin Li
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Shijian Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Zhengbin Han
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qinghua Jiang
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Qiong Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Tao Liu
- Shenzhen Luohu People's Hospital, Shenzhen Zhongxun Precision Medicine Research InstituteShenzhen Guangdong518001 China
| | - Cheng Zhang
- The First Affiliated HospitalHarbin Medical UniversityHarbin Heilongjiang150081 China
| | - Miao Yu
- School of Chemical Engineering and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
| | - Ying Hu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin Heilongjiang150001 China
- Shenzhen Graduate School of Harbin Institute of TechnologyXili University CityNanshanShenzhen Guangdong518055 China
| |
Collapse
|
54
|
Tanimoto K. Genetics of the hypoxia-inducible factors in human cancers. Exp Cell Res 2017; 356:166-172. [DOI: 10.1016/j.yexcr.2017.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/16/2017] [Indexed: 12/12/2022]
|
55
|
Bailey ST, Smith AM, Kardos J, Wobker SE, Wilson HL, Krishnan B, Saito R, Lee HJ, Zhang J, Eaton SC, Williams LA, Manocha U, Peters DJ, Pan X, Carroll TJ, Felsher DW, Walter V, Zhang Q, Parker JS, Yeh JJ, Moffitt RA, Leung JY, Kim WY. MYC activation cooperates with Vhl and Ink4a/Arf loss to induce clear cell renal cell carcinoma. Nat Commun 2017; 8:15770. [PMID: 28593993 PMCID: PMC5472759 DOI: 10.1038/ncomms15770] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 04/26/2017] [Indexed: 11/17/2022] Open
Abstract
Renal carcinoma is a common and aggressive malignancy whose histopathogenesis is incompletely understood and that is largely resistant to cytotoxic chemotherapy. We present two mouse models of kidney cancer that recapitulate the genomic alterations found in human papillary (pRCC) and clear cell RCC (ccRCC), the most common RCC subtypes. MYC activation results in highly penetrant pRCC tumours (MYC), while MYC activation, when combined with Vhl and Cdkn2a (Ink4a/Arf) deletion (VIM), produce kidney tumours that approximate human ccRCC. RNAseq of the mouse tumours demonstrate that MYC tumours resemble Type 2 pRCC, which are known to harbour MYC activation. Furthermore, VIM tumours more closely simulate human ccRCC. Based on their high penetrance, short latency, and histologic fidelity, these models of papillary and clear cell RCC should be significant contributions to the field of kidney cancer research.
Collapse
Affiliation(s)
- Sean T. Bailey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Aleisha M. Smith
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jordan Kardos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Sara E. Wobker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Harper L. Wilson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Bhavani Krishnan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ryoichi Saito
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Hyo Jin Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Samuel C. Eaton
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Lindsay A. Williams
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ujjawal Manocha
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Dorien J. Peters
- Department of Pathology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Xinchao Pan
- Departments of Internal Medicine and Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Thomas J. Carroll
- Departments of Internal Medicine and Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Dean W. Felsher
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California 94305-5151, USA
| | - Vonn Walter
- Department of Biochemistry and Molecular Biology, Penn State Milton S. Hershey College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Joel S. Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard A. Moffitt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Janet Y. Leung
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
56
|
Massari F, Ciccarese C, Bria E, Porta C, La Russa F, Knuutila S, Artibani W, Porcaro AB, Bimbatti D, Modena A, Sava T, Tortora G, Cheng L, Eccher A, Cima L, Pedron S, Ghimenton C, Martignoni G, Brunelli M. Reprofiling Metastatic Samples for Chromosome 9p and 14q Aberrations as a Strategy to Overcome Tumor Heterogeneity in Clear-cell Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2017; 25:39-43. [PMID: 26509904 DOI: 10.1097/pai.0000000000000257] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Losses of chromosomes 9p and 14q are associated with worse outcomes in patients affected by clear-cell renal cell carcinoma (RCC) and are helpful for prognostic risk stratification. Both chromosomal loci harbor several hot-spot molecular pathways suitable for targeted therapeutic interventions. Intratumor heterogeneity may foster tumor adaptation and therapeutic failure. We sought to investigate the presence of losses of the hot spots of chromosomal loci 9p and 14q in primary clear-cell RCC and matched metastatic tissues. CD10 and CD13 were performed on 7 cases of clear-cell RCC with hematogenous tissue metastases. Cytogenetic fluorescence in situ hybridization analysis was performed on primary and matched metastatic tissues using specific probes mapping the 9p and the 14q loci. The loss of chromosome 9p was observed in 85% of both primary clear-cell RCCs and in matched metastases; 14% showed discordance between primary and matched metastases showing gains. The loss of chromosome 14q was observed in 58% of both primary and matched metastases. Only 3/7 (42%) did show an equal status of loss of chromosome 14q. Heterogeneity of the cytogenetic status between metastatic and primary clear-cell RCCs is observed for the loss of chromosome 14q rather than chromosome 9p. The impact of chromosome 14q cytogenetic status, harboring the HIF1 gene, a major driver for the angiogenenic switch, may drive the efficacy of targeted inhibitors, whereas the loss of chromosome 9p, harboring other hot-spot genes, seems to be related to the metastatic behavior per se, without cytogenetic modulation. Reprofiling the metastatic tissue, as compared with the primary tumor, in patients affected by metastatic RCC could be a novel approach to overcome resistance to VEGF(Rs)-targeting agents.
Collapse
Affiliation(s)
- Francesco Massari
- *Department of Medical Oncology §Urologic Clinic ¶Department of Pathology and Diagnostic, University and Hospital Trust, Verona †Medical Oncology, I.R.C.C.S. San Matteo University Hospital Foundation, Pavia, Italy ‡Department of Pathology, Laboratory of Molecular Cytogenetic, University of Helsinki, Helsinki, Finland ∥Department of Pathology and Laboratory Medicine, Indiana University, Bloomington, IN
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Offermann A, Kuempers C, Perner S. Histological (Sub)Classifications and Their Prognostic Impact in Renal Cell Carcinoma. Urol Oncol 2017. [DOI: 10.1007/978-3-319-42603-7_60-1] [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]
|
58
|
Wallace EM, Rizzi JP, Han G, Wehn PM, Cao Z, Du X, Cheng T, Czerwinski RM, Dixon DD, Goggin BS, Grina JA, Halfmann MM, Maddie MA, Olive SR, Schlachter ST, Tan H, Wang B, Wang K, Xie S, Xu R, Yang H, Josey JA. A Small-Molecule Antagonist of HIF2α Is Efficacious in Preclinical Models of Renal Cell Carcinoma. Cancer Res 2016; 76:5491-500. [PMID: 27635045 DOI: 10.1158/0008-5472.can-16-0473] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
Abstract
More than 90% of clear cell renal cell carcinomas (ccRCC) exhibit inactivation of the von Hippel-Lindau (pVHL) tumor suppressor, establishing it as the major underlying cause of this malignancy. pVHL inactivation results in stabilization of the hypoxia-inducible transcription factors, HIF1α and HIF2α, leading to expression of a genetic program essential for the initiation and progression of ccRCC. Herein, we describe the potent, selective, and orally active small-molecule inhibitor PT2385 as a specific antagonist of HIF2α that allosterically blocks its dimerization with the HIF1α/2α transcriptional dimerization partner ARNT/HIF1β. PT2385 inhibited the expression of HIF2α-dependent genes, including VEGF-A, PAI-1, and cyclin D1 in ccRCC cell lines and tumor xenografts. Treatment of tumor-bearing mice with PT2385 caused dramatic tumor regressions, validating HIF2α as a pivotal oncogenic driver in ccRCC. Notably, unlike other anticancer agents that inhibit VEGF receptor signaling, PT2385 exhibited no adverse effect on cardiovascular performance. Thus, PT2385 represents a novel class of therapeutics for the treatment of RCC with potent preclincal efficacy as well as improved tolerability relative to current agents that target the VEGF pathway. Cancer Res; 76(18); 5491-500. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | - Xinlin Du
- Peloton Therapeutics, Inc., Dallas, Texas
| | | | | | | | | | | | | | | | | | | | | | - Bin Wang
- Peloton Therapeutics, Inc., Dallas, Texas
| | - Keshi Wang
- Peloton Therapeutics, Inc., Dallas, Texas
| | | | - Rui Xu
- Peloton Therapeutics, Inc., Dallas, Texas
| | | | | |
Collapse
|
59
|
Schödel J, Grampp S, Maher ER, Moch H, Ratcliffe PJ, Russo P, Mole DR. Hypoxia, Hypoxia-inducible Transcription Factors, and Renal Cancer. Eur Urol 2016; 69:646-657. [PMID: 26298207 PMCID: PMC5012644 DOI: 10.1016/j.eururo.2015.08.007] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/05/2015] [Indexed: 12/18/2022]
Abstract
CONTEXT Renal cancer is a common urologic malignancy, and therapeutic options for metastatic disease are limited. Most clear cell renal cell carcinomas (ccRCC) are associated with loss of von Hippel-Lindau tumor suppressor (pVHL) function and deregulation of hypoxia pathways. OBJECTIVE This review summarizes recent evidence from genetic and biological studies showing that hypoxia and hypoxia-related pathways play critical roles in the development and progress of renal cancer. EVIDENCE ACQUISITION We used a systematic search for articles using the keywords hypoxia, HIF, renal cancer, and VHL. EVIDENCE SYNTHESIS Identification of the tumor suppressor pVHL has allowed the characterization of important ccRCC-associated pathways. pVHL targets α-subunits of hypoxia-inducible transcription factors (HIF) for proteasomal degradation. The two main HIF-α isoforms have opposing effects on RCC biology, possibly through distinct interactions with additional oncogenes. Furthermore, HIF-1α activity is commonly diminished by chromosomal deletion in ccRCCs, and increased HIF-1 activity reduces tumor burden in xenograft tumor models. Conversely, polymorphisms at the HIF-2α gene locus predispose to the development of ccRCCs, and HIF-2α promotes tumor growth. Genetic studies have revealed a prominent role for chromatin-modifying enzyme genes in ccRCC, and these may further modulate specific aspects of the HIF response. This suggests that, rather than global activation of HIF, specific components of the response are important in promoting kidney cancer. Some of these processes are already targets for current therapeutic strategies, and further dissection of this pathway might yield novel methods of treating RCC. CONCLUSIONS In contrast to many tumor types, HIF-1α and HIF-2α have opposing effects in ccRCC biology, with HIF-1α acting as a tumor suppressor and HIF-2α acting as an oncogene. The overall effect of VHL inactivation will depend on fine-tuning of the HIF response. PATIENT SUMMARY High levels of hypoxia-inducible transcription factors (HIF) are particularly important in the clear cell type of kidney cancer, in which they are no longer properly regulated by the von Hippel-Lindau protein. The two HIF-α proteins have opposing effects on tumor evolution.
Collapse
Affiliation(s)
- Johannes Schödel
- Medizinische Klinik 4 and Translational Research Center, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| | - Steffen Grampp
- Medizinische Klinik 4 and Translational Research Center, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Cambridge NIHR Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, UK
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter J Ratcliffe
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford, UK
| | - Paul Russo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, NY, NY, USA; Weill Medical College, Cornell University, Memorial Sloan Kettering Cancer Center, NY, NY, USA
| | - David R Mole
- Henry Wellcome Building for Molecular Physiology, University of Oxford, Oxford, UK
| |
Collapse
|
60
|
Specific genomic aberrations predict survival, but low mutation rate in cancer hot spots, in clear cell renal cell carcinoma. Appl Immunohistochem Mol Morphol 2016; 23:334-42. [PMID: 24992170 PMCID: PMC4431677 DOI: 10.1097/pai.0000000000000087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Detailed genetic profiling of clear cell renal cell carcinoma (ccRCC) has revealed genomic regions commonly affected by structural changes and a general genetic heterogeneity. VHL and PBRM1, both located at chromosome 3p, are 2 major genes mutated at high frequency but apart from these aberrations, the mutational landscape in ccRCC is largely undefined. Potential prognostic information given by the genomic changes appears to depend on the particular cohort studied. We analyzed a Swedish ccRCC cohort of 74 patients and found common changes (loss or gain occurring in >20% of the tumors) in 12 chromosomal regions (1p, 3p, 3q, 5q, 6q, 7p, 7q 8p, 9p, 9q, 10q, and 14q). A poor outcome was associated with gain of 7q and losses on 9p, 9q, and 14q. These aberrations were more frequent in metastasized tumors, suggesting alterations of genes important for tumor progression. Sequencing of 48 genes implicated in cancer revealed that only VHL, TP53, and PTEN were mutated at a noticeable frequency (51%, 9%, and 9%, respectively). Shorter relative telomere length (RTL) has been associated with loss of specific chromosomal regions in ccRCC tumors, but we could not verify this finding. However, a significantly lower tumor/nontumor (T/N) RTL ratio was detected for tumors with losses in 4q or 9p. In conclusion, poor outcome in ccRCC was associated with gain of 7q and loss on 9p, 9q, and 14q, whereas the mutation rate overall was low in a screen of cancer-associated genes.
Collapse
|
61
|
Schönenberger D, Harlander S, Rajski M, Jacobs RA, Lundby AK, Adlesic M, Hejhal T, Wild PJ, Lundby C, Frew IJ. Formation of Renal Cysts and Tumors in Vhl/Trp53-Deficient Mice Requires HIF1α and HIF2α. Cancer Res 2016; 76:2025-36. [PMID: 26759234 DOI: 10.1158/0008-5472.can-15-1859] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/07/2016] [Indexed: 11/16/2022]
Abstract
The von Hippel-Lindau (VHL) tumor suppressor gene is inactivated in the majority of clear cell renal cell carcinomas (ccRCC), but genetic ablation of Vhl alone in mouse models is insufficient to recapitulate human tumorigenesis. One function of pVHL is to regulate the stability of the hypoxia-inducible factors (HIF), which become constitutively activated in the absence of pVHL. In established ccRCC, HIF1α has been implicated as a renal tumor suppressor, whereas HIF2α is considered an oncoprotein. In this study, we investigated the contributions of HIF1α and HIF2α to ccRCC initiation in the context of Vhl deficiency. We found that deleting Vhl plus Hif1a or Hif2a specifically in the renal epithelium did not induce tumor formation. However, HIF1α and HIF2α differentially regulated cell proliferation, mitochondrial abundance and oxidative capacity, glycogen accumulation, and acquisition of a clear cell phenotype in Vhl-deficient renal epithelial cells. HIF1α, but not HIF2α, induced Warburg-like metabolism characterized by increased glycolysis, decreased oxygen consumption, and decreased ATP production in mouse embryonic fibroblasts, providing insights into the cellular changes potentially occurring in Vhl mutant renal cells before ccRCC formation. Importantly, deletion of either Hif1a or Hif2a completely prevented the formation of renal cysts and tumors in Vhl/Trp53 mutant mice. These findings argue that both HIF1α and HIF2α exert protumorigenic functions during the earliest stages of cyst and tumor formation in the kidney. Cancer Res; 76(7); 2025-36. ©2016 AACR.
Collapse
Affiliation(s)
| | - Sabine Harlander
- Institute of Physiology, University of Zurich, Zurich, Switzerland. Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Michal Rajski
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Robert A Jacobs
- Institute of Physiology, University of Zurich, Zurich, Switzerland. Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland. Health and Physical Education, School of Teaching and Learning, Western Carolina University, Cullowhee, North Carolina
| | - Anne-Kristine Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland. Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mojca Adlesic
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Tomas Hejhal
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Peter J Wild
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Carsten Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland. Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Ian J Frew
- Institute of Physiology, University of Zurich, Zurich, Switzerland. Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
62
|
Hamilton HH, McDermott A, Smith MT, Savage SJ, Wolff DJ. Clinical utility of concurrent single-nucleotide polymorphism microarray on fresh tissue as a supplementary test in the diagnosis of renal epithelial neoplasms. Am J Clin Pathol 2015; 144:731-7. [PMID: 26486737 DOI: 10.1309/ajcpjt7f5vnrxxpf] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVES The histologic and immunohistochemical variability of renal epithelial tumors makes classification difficult; with significant clinical implications, efforts to make the proper diagnoses are necessary. Single-nucleotide polymorphism (SNP) microarray analysis has been proposed as a supplementary study for the classification of renal epithelial neoplasms; however, its practical use in the routine clinical setting has not been explored. METHODS Surgical pathology cases that were classified histologically as renal epithelial tumor subtypes and had concurrent SNP microarray were retrospectively reviewed to correlate tumor morphology and SNP microarray results. RESULTS Of the 99 cases reviewed, 88 (89%) had concordant histologic and microarray results. Four (4%) cases were unclassifiable by microarray due to uncharacteristic chromosomal abnormalities. Seven (7%) of the 99 cases had discordant microarray and histologic diagnoses, and following review of the histology, the diagnoses in two of these cases were subsequently changed. CONCLUSIONS For most cases, concurrent SNP microarray confirmed the histologic diagnosis. However, discrepant microarray results prompted review of morphology and further ancillary studies, resulting in amendment of the final diagnosis in 29% of discrepant cases. SNP microarray analysis can be used to assist with the diagnosis of renal epithelial tumors, particularly those with atypical morphologic features.
Collapse
|
63
|
Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM. New strategies in renal cell carcinoma: targeting the genetic and metabolic basis of disease. Clin Cancer Res 2015; 21:10-7. [PMID: 25564569 DOI: 10.1158/1078-0432.ccr-13-2993] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The development of new forms of treatment of advanced renal cell carcinoma over the past two decades has been primarily focused on targeting the VHL/HIF pathway. The recent identification of mutations of chromatin-remodeling genes in clear-cell renal carcinoma (ccRCC), of genomic heterogeneity, and of a Warburg-like metabolic phenotype in advanced disease has had a profound effect on our understanding of the evolution of ccRCC and on potential approaches to personalized therapy. Early approaches to therapy for patients with advanced type I papillary RCC that have centered around the MET/HGF pathway will expand as more genomic information becomes available. Sporadic and familial type II papillary renal cell carcinoma are characterized by enhanced aerobic glycolysis and share an antioxidant response phenotype. In fumarate hydratase-deficient RCC, fumarate-induced succination of KEAP1 activates Nrf2 signaling. CUL3 and Nrf2 mutations as well as an Nrf2 activation phenotype are found in sporadic type II papillary RCC. Therapeutic approaches designed to target the Nrf2 pathway as well as to impair blood flow and glucose delivery in these cancers that are highly dependent on a robust tumor vasculature and on ready availability of glucose for energy production and glycolysis are in development.
Collapse
Affiliation(s)
- Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
64
|
Genetic and Chromosomal Aberrations and Their Clinical Significance in Renal Neoplasms. BIOMED RESEARCH INTERNATIONAL 2015; 2015:476508. [PMID: 26448938 PMCID: PMC4584050 DOI: 10.1155/2015/476508] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/23/2015] [Accepted: 05/25/2015] [Indexed: 12/16/2022]
Abstract
The most common form of malignant renal neoplasms is renal cell carcinoma (RCC), which is classified into several different subtypes based on the histomorphological features. However, overlaps in these characteristics may present difficulties in the accurate diagnosis of these subtypes, which have different clinical outcomes. Genomic and molecular studies have revealed unique genetic aberrations in each subtype. Knowledge of these genetic changes in hereditary and sporadic renal neoplasms has given an insight into the various proteins and signalling pathways involved in tumour formation and progression. In this review, the genetic aberrations characteristic to each renal neoplasm subtype are evaluated along with the associated protein products and affected pathways. The potential applications of these genetic aberrations and proteins as diagnostic tools, prognostic markers, or therapeutic targets are also assessed.
Collapse
|
65
|
Klinke OK, Mizani T, Baldwin G, Bancel B, Devouassoux-Shisheboran M, Scoazec JY, Bringuier PP, Feederle R, Jauch A, Hinderhofer K, Taniere P, Delecluse HJ. KIT Mutation and Loss of 14q May Be Sufficient for the Development of Clinically Symptomatic Very Low-Risk GIST. PLoS One 2015; 10:e0130149. [PMID: 26102504 PMCID: PMC4477893 DOI: 10.1371/journal.pone.0130149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/18/2015] [Indexed: 01/13/2023] Open
Abstract
The aim of this study was to determine the minimal set of genetic alterations required for the development of a very low risk clinically symptomatic gastro-intestinal stromal tumour within the stomach wall. We studied the genome of a very low-risk gastric gastro-intestinal stromal tumour by whole-genome sequencing, comparative genomic hybridisation and methylation profiling. The studied tumour harboured two typical genomic lesions: loss of the long arm of chromosome 14 and an activating mutation in exon 11 of KIT. Besides these genetic lesions, only two point mutations that may affect tumour progression were identified: A frame-shift deletion in RNF146 and a missense mutation in a zinc finger of ZNF407. Whilst the frameshift deletion in RNF146 seemed to be restricted to this particular tumour, a similar yet germline mutation in ZNF407 was found in a panel of 52 gastro-intestinal stromal tumours from different anatomical sites and different categories. Germline polymorphisms in the mitotic checkpoint proteins Aurora kinase A and BUB1 kinase B may have furthered tumour growth. The epigenetic profile of the tumour matches that of other KIT-mutant tumours. We have identified mutations in three genes and loss of the long arm of chromosome 14 as the so far minimal set of genetic abnormalities sufficient for the development of a very low risk clinically symptomatic gastric stromal tumour.
Collapse
Affiliation(s)
- Olaf Karl Klinke
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany
- Inserm unit U1074, Heidelberg, Germany
| | - Tuba Mizani
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany
- Inserm unit U1074, Heidelberg, Germany
| | - Gouri Baldwin
- Histopathology Cellular Pathology–University Hospitals Birmingham, NHS Foundation, Trust Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston Birmingham, B15 2WB, England
| | - Brigitte Bancel
- Service d’Anatomie et Cytologie pathologiques, Hôpital de la Croix Rousse, 103 Grande-Rue-de-la-Croix-Rousse, Lyon cedex 04, France
| | - Mojgan Devouassoux-Shisheboran
- Service d’Anatomie et Cytologie pathologiques, Hôpital de la Croix Rousse, 103 Grande-Rue-de-la-Croix-Rousse, Lyon cedex 04, France
| | - Jean-Yves Scoazec
- Service d’Anatomie Pathologique, Hôpital Édouard-Herriot, 5, place d’Arsonval, 69437 Lyon cedex 03, France
| | - Pierre-Paul Bringuier
- Service d’Anatomie Pathologique, Hôpital Édouard-Herriot, 5, place d’Arsonval, 69437 Lyon cedex 03, France
| | - Regina Feederle
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany
- Inserm unit U1074, Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University Heidelberg, Heidelberg, Germany
| | | | - Philippe Taniere
- Histopathology Cellular Pathology–University Hospitals Birmingham, NHS Foundation, Trust Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston Birmingham, B15 2WB, England
| | - Henri-Jacques Delecluse
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany
- Inserm unit U1074, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
66
|
Ho TH, Park IY, Zhao H, Tong P, Champion MD, Yan H, Monzon FA, Hoang A, Tamboli P, Parker AS, Joseph RW, Qiao W, Dykema K, Tannir NM, Castle EP, Nunez-Nateras R, Teh BT, Wang J, Walker CL, Hung MC, Jonasch E. High-resolution profiling of histone h3 lysine 36 trimethylation in metastatic renal cell carcinoma. Oncogene 2015; 35:1565-74. [PMID: 26073078 PMCID: PMC4679725 DOI: 10.1038/onc.2015.221] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 03/01/2015] [Accepted: 03/06/2015] [Indexed: 02/07/2023]
Abstract
Mutations in SETD2, a histone H3 lysine trimethyltransferase, have been identified in clear cell renal cell carcinoma (ccRCC); however it is unclear if loss of SETD2 function alters the genomic distribution of histone 3 lysine 36 trimethylation (H3K36me3) in ccRCC. Furthermore, published epigenomic profiles are not specific to H3K36me3 or metastatic tumors. To determine if progressive SETD2 and H3K36me3 dysregulation occurs in metastatic tumors, H3K36me3, SETD2 copy number (CN) or SETD2 mRNA abundance was assessed in two independent cohorts: metastatic ccRCC (n=71) and the Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma data set (n=413). Although SETD2 CN loss occurs with high frequency (>90%), H3K36me3 is not significantly impacted by monoallelic loss of SETD2. H3K36me3-positive nuclei were reduced an average of ~20% in primary ccRCC (90% positive nuclei in uninvolved vs 70% positive nuclei in ccRCC) and reduced by ~60% in metastases (90% positive in uninvolved kidney vs 30% positive in metastases) (P<0.001). To define a kidney-specific H3K36me3 profile, we generated genome-wide H3K36me3 profiles from four cytoreductive nephrectomies and SETD2 isogenic renal cell carcinoma (RCC) cell lines using chromatin immunoprecipitation coupled with high-throughput DNA sequencing and RNA sequencing. SETD2 loss of methyltransferase activity leads to regional alterations of H3K36me3 associated with aberrant RNA splicing in a SETD2 mutant RCC and SETD2 knockout cell line. These data suggest that during progression of ccRCC, a decline in H3K36me3 is observed in distant metastases, and regional H3K36me3 alterations influence alternative splicing in ccRCC.
Collapse
Affiliation(s)
- T H Ho
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - I Y Park
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - H Zhao
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Tong
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M D Champion
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Biomedical Statistics and Informatics, Mayo Clinic, Scottsdale, AZ, USA
| | - H Yan
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Biomedical Statistics and Informatics, Rochester, MN, USA
| | - F A Monzon
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - A Hoang
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Tamboli
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A S Parker
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - R W Joseph
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - W Qiao
- Division of Quantitative Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Dykema
- Center for Cancer Genomics and Computational Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - N M Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E P Castle
- Department of Urology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - B T Teh
- Center for Cancer Genomics and Computational Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - J Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C L Walker
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - M-C Hung
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - E Jonasch
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
67
|
White-Al Habeeb NM, Di Meo A, Scorilas A, Rotondo F, Masui O, Seivwright A, Gabril M, Girgis AHA, Jewett MA, Yousef GM. Alpha-enolase is a potential prognostic marker in clear cell renal cell carcinoma. Clin Exp Metastasis 2015; 32:531-41. [DOI: 10.1007/s10585-015-9725-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/27/2015] [Indexed: 01/20/2023]
|
68
|
Shim EH, Sudarshan S. Another small molecule in the oncometabolite mix: L-2-Hydroxyglutarate in kidney cancer. Oncoscience 2015; 2:483-6. [PMID: 26097881 PMCID: PMC4468334 DOI: 10.18632/oncoscience.165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/06/2015] [Indexed: 12/18/2022] Open
Abstract
Alterations in metabolism are now considered a hallmark of cancer. One of the clearest links between metabolism and malignancy are oncometabolites. To date, several putative oncometabolites with transforming properties have been identified in the context of tumors due to both gain and loss of function mutations in genes encoding enzymes of intermediary metabolism. Through an unbiased metabolomics approach, we identified elevations of the metabolite 2-hydroxyglutarate (2-HG) in the most common histology of kidney cancer that is among the most common malignancies in both men and women. Subsequent analyses demonstrate that the predominant enantiomer of 2-HG elevated in renal cancer is the L(S) form. Notably, elevations of L-2HG are due in part to loss of expression of the L-2HG dehydrogenase (L2HGDH) which normally serves as an enzyme of “metabolite repair” to keep levels of this metabolite from accumulating. Lowering L-2HG levels in RCC through re-expression of L2HGDH mitigates tumor phenotypes and reverses epigenetic alterations known to be targeted by oncometabolites. These data add to the growing body of evidence that metabolites, similarly to oncogenes and oncoproteins, can play a role in tumor development and/or progression. As such, they represent a unique opportunity to utilize these findings in the clinic setting.
Collapse
Affiliation(s)
- Eun-Hee Shim
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sunil Sudarshan
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
69
|
Wei H, Li Z, Wang X, Wang J, Pang W, Yang G, Shen QW. microRNA-151-3p regulates slow muscle gene expression by targeting ATP2a2 in skeletal muscle cells. J Cell Physiol 2015; 230:1003-12. [PMID: 25200835 DOI: 10.1002/jcp.24793] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNAs that regulate the stability or translation of cognate mRNAs at the post-transcriptional level. Accumulating evidence indicates that miRNAs play important roles in many aspects of muscle function, including muscle growth and development, regeneration, contractility, and muscle fiber type plasticity. In the current study, we examined the function of miR-151-3p in myoblast proliferation and differentiation. Results show that overexpression of miR-151-3p not only upregulates myoblast proliferation, but also decreases slow muscle gene expression (such as MHC-β/slow and slow muscle troponin I) in both C2C12 myotubes and in primary cultures. Alternatively, inhibition of miR-151-3p by antisense RNA was found to upregulate MHC-β/slow expression, indicating that miR-151-3p plays a role in muscle fiber type determination. Further investigation into the underlying mechanisms revealed for the first time that miR-151-3p directly targets ATP2a2, a gene encoding for a slow skeletal and cardiac muscle specific Ca(2+) ATPase, SERCA2 thus downregulating slow muscle gene expression. Mechanisms by which the alteration in SERCA2 expression induces changes in other slow muscle gene expression levels needs to be defined in future research.
Collapse
Affiliation(s)
- Huan Wei
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
70
|
Beuselinck B, Job S, Becht E, Karadimou A, Verkarre V, Couchy G, Giraldo N, Rioux-Leclercq N, Molinié V, Sibony M, Elaidi R, Teghom C, Patard JJ, Méjean A, Fridman WH, Sautès-Fridman C, de Reyniès A, Oudard S, Zucman-Rossi J. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin Cancer Res 2015; 21:1329-39. [PMID: 25583177 DOI: 10.1158/1078-0432.ccr-14-1128] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Selecting patients with metastatic clear-cell renal cell carcinoma (m-ccRCC) who might benefit from treatment with targeted tyrosine kinase inhibitors (TKI) is a challenge. Our aim was to identify molecular markers associated with outcome in patients with m-ccRCC treated with sunitinib. EXPERIMENTAL DESIGN We performed global transcriptome analyses on 53 primary resected ccRCC tumors from patients who developed metastatic disease and were treated with first-line sunitinib. We also determined chromosome copy-number aberrations, methylation status, and gene mutations in von Hippel-Lindau and PBRM1. Molecular data were analyzed in relation with response rate (RR), progression-free survival (PFS), and overall survival (OS). Validation was performed in 47 additional ccRCC samples treated in first-line metastatic setting with sunitinib. RESULTS Unsupervised transcriptome analysis identified 4 robust ccRCC subtypes (ccrcc1 to 4) related to previous molecular classifications that were associated with different responses to sunitinib treatment. ccrcc1/ccrcc4 tumors had a lower RR (P = 0.005) and a shorter PFS and OS than ccrcc2/ccrcc3 tumors (P = 0.001 and 0.0003, respectively). These subtypes were the only significant covariate in the multivariate Cox model for PFS and OS (P = 0.017 and 0.006, respectively). ccrcc1/ccrcc4 tumors were characterized by a stem-cell polycomb signature and CpG hypermethylation, whereas ccrcc3 tumors, sensitive to sunitinib, did not exhibit cellular response to hypoxia. Moreover, ccrcc4 tumors exhibited sarcomatoid differentiation with a strong inflammatory, Th1-oriented but suppressive immune microenvironment, with high expression of PDCD1 (PD-1) and its ligands. CONCLUSIONS ccRCC molecular subtypes are predictive of sunitinib response in metastatic patients, and could be used for personalized mRCC treatment with TKIs, demethylating or immunomodulatory drugs.
Collapse
Affiliation(s)
- Benoit Beuselinck
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France
| | - Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Etienne Becht
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France. UMR_S1138, Centre de Recherche des Cordeliers, Paris, France
| | - Alexandra Karadimou
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France
| | - Virginie Verkarre
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants malades, Department of Pathology, Paris, France
| | - Gabrielle Couchy
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France
| | - Nicolas Giraldo
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. UMR_S1138, Centre de Recherche des Cordeliers, Paris, France
| | | | | | - Mathilde Sibony
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Department of Pathology, Paris, France
| | - Reza Elaidi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France
| | - Corinne Teghom
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France
| | | | - Arnaud Méjean
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France
| | - Wolf Herman Fridman
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France. UMR_S1138, Centre de Recherche des Cordeliers, Paris, France
| | - Catherine Sautès-Fridman
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France. UMR_S1138, Centre de Recherche des Cordeliers, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Stéphane Oudard
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France
| | - Jessica Zucman-Rossi
- Inserm, UMR-1162, Génomique fonctionnelle des tumeurs solides, Paris, France. Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France. Labex Immuno-oncology, Paris, France. Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Oncology, Paris, France.
| |
Collapse
|
71
|
|
72
|
Koh MY, Nguyen V, Lemos R, Darnay BG, Kiriakova G, Abdelmelek M, Ho TH, Karam J, Monzon FA, Jonasch E, Powis G. Hypoxia-induced SUMOylation of E3 ligase HAF determines specific activation of HIF2 in clear-cell renal cell carcinoma. Cancer Res 2014; 75:316-29. [PMID: 25421578 DOI: 10.1158/0008-5472.can-13-2190] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Clear-cell renal cell cancer (CRCC) is initiated typically by loss of the tumor-suppressor VHL, driving constitutive activation of hypoxia-inducible factor-1 (HIF1) and HIF2. However, whereas HIF1 has a tumor-suppressor role, HIF2 plays a distinct role in driving CRCC. In this study, we show that the HIF1α E3 ligase hypoxia-associated factor (HAF) complexes with HIF2α at DNA to promote HIF2-dependent transcription through a mechanism relying upon HAF SUMOylation. HAF SUMOylation was induced by hypoxia, whereas HAF-mediated HIF1α degradation was SUMOylation independent. HAF overexpression in mice increased CRCC growth and metastasis. Clinically, HAF overexpression was associated with poor prognosis. Taken together, our results show that HAF is a specific mediator of HIF2 activation that is critical for CRCC development and morbidity.
Collapse
Affiliation(s)
- Mei Yee Koh
- Sanford-Burnham Medical Research Institute, La Jolla, California.
| | - Vuvi Nguyen
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Lemos
- Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Bryant G Darnay
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Galina Kiriakova
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mena Abdelmelek
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thai H Ho
- Department of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Jose Karam
- Department of GU Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Federico A Monzon
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Eric Jonasch
- Department of GU Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Garth Powis
- Sanford-Burnham Medical Research Institute, La Jolla, California
| |
Collapse
|
73
|
Gulati S, Martinez P, Joshi T, Birkbak NJ, Santos CR, Rowan AJ, Pickering L, Gore M, Larkin J, Szallasi Z, Bates PA, Swanton C, Gerlinger M. Systematic evaluation of the prognostic impact and intratumour heterogeneity of clear cell renal cell carcinoma biomarkers. Eur Urol 2014; 66:936-48. [PMID: 25047176 PMCID: PMC4410302 DOI: 10.1016/j.eururo.2014.06.053] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/30/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Candidate biomarkers have been identified for clear cell renal cell carcinoma (ccRCC) patients, but most have not been validated. OBJECTIVE To validate published ccRCC prognostic biomarkers in an independent patient cohort and to assess intratumour heterogeneity (ITH) of the most promising markers to guide biomarker optimisation. DESIGN, SETTING, AND PARTICIPANTS Cancer-specific survival (CSS) for each of 28 identified genetic or transcriptomic biomarkers was assessed in 350 ccRCC patients. ITH was interrogated in a multiregion biopsy data set of 10 ccRCCs. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Biomarker association with CSS was analysed by univariate and multivariate analyses. RESULTS AND LIMITATIONS A total of 17 of 28 biomarkers (TP53 mutations; amplifications of chromosomes 8q, 12, 20q11.21q13.32, and 20 and deletions of 4p, 9p, 9p21.3p24.1, and 22q; low EDNRB and TSPAN7 expression and six gene expression signatures) were validated as predictors of poor CSS in univariate analysis. Tumour stage and the ccB expression signature were the only independent predictors in multivariate analysis. ITH of the ccB signature was identified in 8 of 10 tumours. Several genetic alterations that were significant in univariate analysis were enriched, and chromosomal instability indices were increased in samples expressing the ccB signature. The study may be underpowered to validate low-prevalence biomarkers. CONCLUSIONS The ccB signature was the only independent prognostic biomarker. Enrichment of multiple poor prognosis genetic alterations in ccB samples indicated that several events may be required to establish this aggressive phenotype, catalysed in some tumours by chromosomal instability. Multiregion assessment may improve the precision of this biomarker. PATIENT SUMMARY We evaluated the ability of published biomarkers to predict the survival of patients with clear cell kidney cancer in an independent patient cohort. Only one molecular test adds prognostic information to routine clinical assessments. This marker showed good and poor prognosis results within most individual cancers. Future biomarkers need to consider variation within tumours to improve accuracy.
Collapse
Affiliation(s)
- Sakshi Gulati
- Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Pierre Martinez
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Tejal Joshi
- Centre for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - Nicolai Juul Birkbak
- Centre for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - Claudio R Santos
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, London, UK
| | - Andrew J Rowan
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, London, UK
| | | | | | | | - Zoltan Szallasi
- Centre for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark; Children's Hospital Informatics Program at the Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Paul A Bates
- Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, London, UK.
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, London, UK; UCL Cancer Institute, London, UK.
| | - Marco Gerlinger
- Translational Cancer Therapeutics Laboratory, Cancer Research UK London Research Institute, London, UK; Present address: Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| |
Collapse
|
74
|
Frew IJ, Moch H. A clearer view of the molecular complexity of clear cell renal cell carcinoma. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2014; 10:263-89. [PMID: 25387056 DOI: 10.1146/annurev-pathol-012414-040306] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The von Hippel-Lindau (VHL) tumor suppressor gene is mutated as an early event in almost all cases of clear cell renal cell carcinoma (ccRCC), the most frequent form of kidney cancer. In this review we discuss recent advances in understanding how dysregulation of the many hypoxia-inducible factor α-dependent and -independent functions of the VHL tumor suppressor protein (pVHL) can contribute to tumor initiation and progression. Recent evidence showing extensive inter- and intratumoral genetic diversity has given rise to the idea that ccRCC should actually be considered as a series of molecularly related, yet distinct, diseases defined by the pattern of combinatorial genetic alterations present within the cells of the tumor. We highlight the range of genetic and epigenetic alterations that recur in ccRCC and discuss the mechanisms through which these events appear to function cooperatively with a loss of pVHL function in tumorigenesis.
Collapse
Affiliation(s)
- Ian J Frew
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich CH-8057, Switzerland;
| | | |
Collapse
|
75
|
Hakimi AA, Mano R, Ciriello G, Gonen M, Mikkilineni N, Sfakianos JP, Kim PH, Motzer RJ, Russo P, Reuter VE, Hsieh JJ, Ostrovnaya I. Impact of recurrent copy number alterations and cancer gene mutations on the predictive accuracy of prognostic models in clear cell renal cell carcinoma. J Urol 2014; 192:24-9. [PMID: 24518768 PMCID: PMC4146751 DOI: 10.1016/j.juro.2014.01.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2014] [Indexed: 11/30/2022]
Abstract
PURPOSE Several recently reported recurrent genomic alterations in clear cell renal cell carcinoma are linked to pathological and clinical outcomes. We determined whether any recurrent cancer gene mutations or copy number alterations identified in the TCGA (The Cancer Genome Atlas) clear cell renal cell carcinoma data set could add to the predictive accuracy of current prognostic models. MATERIALS AND METHODS In 413 patients who underwent nephrectomy/partial nephrectomy we investigated whole exome, copy number array analyses and clinical variables. We identified 65 recurrent genomic alterations based on prevalence and combined them into 35 alterations, including 12 cancer gene mutations. Genomic markers were modeled using the elastic net algorithm with preoperative variables (tumor size plus patient age) and in the postoperative setting using the externally validated Mayo Clinic SSIGN (stage, size, grade and necrosis) prognostic scoring system. These models were subjected to internal validation using bootstrap. RESULTS Median followup in survivors was 45 months. Several markers correlated with adverse cancer specific survival and time to recurrence on univariate analysis. However, most of them lost significance when controlling for tumor size with or without age in the preoperative models or for SSIGN score in the postoperative setting. Adding multiple genomic markers selected by the elastic net algorithm failed to substantially add to the predictive accuracy of any preoperative or postoperative model for cancer specific survival or time to recurrence. CONCLUSIONS While recurrent copy number alterations and cancer gene mutations are biologically significant, they do not appear to improve the predictive accuracy of existing models of clinical cancer specific survival or time to recurrence for clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York.
| | - Roy Mano
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Giovanni Ciriello
- Department of Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Nina Mikkilineni
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - John P Sfakianos
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Philip H Kim
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Robert J Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - James J Hsieh
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
76
|
Gutenberg A, Nischwitz MD, Gunawan B, Enders C, Jung K, Bergmann M, Feiden W, Egensperger R, Keyvani K, Stolke D, Sure U, Schroeder HWS, Warzok R, Schober R, Meixensberger J, Paulus W, Wassmann H, Stummer W, Blumcke I, Buchfelder M, van Landeghem FKH, Vajkoczy P, Günther M, Bedke J, Giese A, Rohde V, Brück W, Füzesi L, Sander B. Predictive chromosomal clusters of synchronous and metachronous brain metastases in clear cell renal cell carcinoma. Cancer Genet 2014; 207:206-13. [PMID: 25027636 DOI: 10.1016/j.cancergen.2014.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/01/2014] [Accepted: 05/10/2014] [Indexed: 01/21/2023]
Abstract
Synchronous (early) and metachronous (late) brain metastasis (BM) events of sporadic clear cell renal cell carcinoma (ccRCC) (n = 148) were retrospectively analyzed using comparative genomic hybridization (CGH). Using oncogenetic tree models and cluster analyses, chromosomal imbalances related to recurrence-free survival until BM (RFS-BM) were analyzed. Losses at 9p and 9q appeared to be hallmarks of metachronous BM events, whereas an absence of detectable chromosomal changes at 3p was often associated with synchronous BM events. Correspondingly, k-means clustering showed that cluster 1 cases generally exhibited low copy number chromosomal changes that did not involve 3p. Cluster 2 cases had a high occurrence of -9p/-9q (94-98%) deletions, whereas cluster 3 cases had a higher frequency of copy number changes, including loss at chromosome 14 (80%). The higher number of synchronous cases in cluster 1 was also associated with a significantly shorter RFS-BM compared with clusters 2 and 3 (P = 0.02). Conversely, a significantly longer RFS-BM was observed for cluster 2 versus clusters 1 and 3 (P = 0.02). Taken together, these data suggest that metachronous BM events of ccRCC are characterized by loss of chromosome 9, whereas synchronous BM events may form independently of detectable genetic changes at chromosomes 9 and 3p.
Collapse
Affiliation(s)
- Angelika Gutenberg
- Department of Neurosurgery, Georg August University, Göttingen, Germany; Department of Neurosurgery, Johannes Gutenberg University, Mainz, Germany.
| | - Martin D Nischwitz
- Department of Gastroenteropathology, Georg August University, Göttingen, Germany
| | - Bastian Gunawan
- Department of Gastroenteropathology, Georg August University, Göttingen, Germany
| | - Christina Enders
- Department of Gastroenteropathology, Georg August University, Göttingen, Germany
| | - Klaus Jung
- Department of Medical Statistics, Georg August University, Göttingen, Germany
| | - Markus Bergmann
- Department of Neuropathology, Klinikum Bremen-Mitte, Bremen, Germany
| | - Wolfgang Feiden
- Departments of Gastroenteropathology and Neuropathology, University of the Saarland, Homburg, Germany
| | - Rupert Egensperger
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Dietmar Stolke
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Sure
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Henry W S Schroeder
- Department of Neurosurgery, Ernst Moritz Arndt University, Greifswald, Germany
| | - Rolf Warzok
- Department of Neuropathology, Ernst Moritz Arndt University, Greifswald, Germany
| | - Ralf Schober
- Department of Neuropathology, University of Leipzig, Leipzig, Germany
| | | | - Werner Paulus
- Department of Neuropathology, Westphalian Wilhelm University, Münster, Germany
| | - Hansdetlef Wassmann
- Department of Neurosurgery, Westphalian Wilhelm University, Münster, Germany
| | - Wolfgang Stummer
- Department of Neurosurgery, Westphalian Wilhelm University, Münster, Germany
| | - Ingmar Blumcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Frank K H van Landeghem
- Department of Neuropathology, Charité University Medicine, Berlin, Germany; Department of Neuropathology, University of Alberta, Edmonton, Canada
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Marlis Günther
- Department of Pathology, Health Care Center Brandenburg, Brandenburg, Germany
| | - Jens Bedke
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Alf Giese
- Department of Neurosurgery, Johannes Gutenberg University, Mainz, Germany
| | - Veit Rohde
- Department of Neurosurgery, Georg August University, Göttingen, Germany
| | - Wolfgang Brück
- Department of Neuropathology, Georg August University, Göttingen, Germany
| | - Laszlo Füzesi
- Department of Gastroenteropathology, Georg August University, Göttingen, Germany
| | - Bjoern Sander
- Stereology and Electron Microscopy Laboratory, Department of Clinical Medicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|
77
|
Alisoltani A, Fallahi H, Ebrahimi M, Ebrahimi M, Ebrahimie E. Prediction of potential cancer-risk regions based on transcriptome data: towards a comprehensive view. PLoS One 2014; 9:e96320. [PMID: 24796549 PMCID: PMC4010480 DOI: 10.1371/journal.pone.0096320] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022] Open
Abstract
A novel integrative pipeline is presented for discovery of potential cancer-susceptibility regions (PCSRs) by calculating the number of altered genes at each chromosomal region, using expression microarray datasets of different human cancers (HCs). Our novel approach comprises primarily predicting PCSRs followed by identification of key genes in these regions to obtain potential regions harboring new cancer-associated variants. In addition to finding new cancer causal variants, another advantage in prediction of such risk regions is simultaneous study of different types of genomic variants in line with focusing on specific chromosomal regions. Using this pipeline we extracted numbers of regions with highly altered expression levels in cancer condition. Regulatory networks were also constructed for different types of cancers following the identification of altered mRNA and microRNAs. Interestingly, results showed that GAPDH, LIFR, ZEB2, mir-21, mir-30a, mir-141 and mir-200c, all located at PCSRs, are common altered factors in constructed networks. We found a number of clusters of altered mRNAs and miRNAs on predicted PCSRs (e.g.12p13.31) and their common regulators including KLF4 and SOX10. Large scale prediction of risk regions based on transcriptome data can open a window in comprehensive study of cancer risk factors and the other human diseases.
Collapse
Affiliation(s)
- Arghavan Alisoltani
- Department of Plant Breeding and Biotechnology, University of Shahrekord, Shahrekord, Iran
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Mahdi Ebrahimi
- Department of Informatics, Saarland University, Saarbrucken, Germany
| | - Mansour Ebrahimi
- Bioinformatics Research Group and Department of Biology, University of Qom, Qom, Iran
| | - Esmaeil Ebrahimie
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
78
|
Hartmann A, Schlomm T, Bertz S, Heinzelmann J, Hölters S, Simon R, Stoehr R, Junker K. [Prognostic and predictive molecular markers for urologic cancers]. Urologe A 2014; 53:491-500. [PMID: 24700189 DOI: 10.1007/s00120-014-3442-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Molecular prognostic factors and genetic alterations as predictive markers for cancer-specific targeted therapies are used today in the clinic for many malignancies. In recent years, many molecular markers for urogenital cancers have also been identified. However, these markers are not clinically used yet. In prostate cancer, novel next-generation sequencing methods revealed a detailed picture of the molecular changes. There is growing evidence that a combination of classical histopathological and validated molecular markers could lead to a more precise estimation of prognosis, thus, resulting in an increasing number of patients with active surveillance as a possible treatment option. In patients with urothelial carcinoma, histopathological factors but also the proliferation of the tumor, mutations in oncogenes leading to an increasing proliferation rate and changes in genes responsible for invasion and metastasis are important. In addition, gene expression profiles which could distinguish aggressive tumors with high risk of metastasis from nonmetastasizing tumors have been recently identified. In the future, this could potentially allow better selection of patients needing systemic perioperative treatment. In renal cell carcinoma, many molecular markers that are associated with metastasis and survival have been identified. Some of these markers were also validated as independent prognostic markers. Selection of patients with primarily organ-confined tumors and increased risk of metastasis for adjuvant systemic therapy could be clinically relevant in the future.
Collapse
Affiliation(s)
- A Hartmann
- Institut für Pathologie, Universität Erlangen, Erlangen, Deutschland
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Currently approved treatments for metastatic renal cell carcinoma (RCC) include vascular endothelial growth factor (VEGF)-blocking agents, mammalian target of rapamycin (mTOR) inhibitors, and cytokine therapy. In the near future, we are likely to add immune checkpoint blocking agents to this list. As we develop treatment platforms around each therapeutic class, determining which drug is best for a particular patient becomes increasingly important. At this point, we do not have validated predictive biomarkers for patients with RCC. Here, we discuss the logistical challenges surrounding biomarker development, summarize the current crop of biomarker candidates, and explore potential avenues for the development of more effective predictive tools for patients with advanced RCC.
Collapse
Affiliation(s)
- Jesus Garcia-Donas
- Genitourinary Tumors Programme Centro Integral Oncologico Clara Campal CIOCC, Madrid, Spain
| | | | | |
Collapse
|
80
|
Histologic prognostic factors associated with chromosomal imbalances in a contemporary series of 89 clear cell renal cell carcinomas. Hum Pathol 2013; 44:2106-15. [PMID: 23806527 DOI: 10.1016/j.humpath.2013.03.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 03/27/2013] [Accepted: 03/28/2013] [Indexed: 11/21/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cancer. The aim of this study was to define specific chromosomal imbalances in ccRCC that could be related to clinical or histologic prognostic factors. Tumors and karyotypes of 89 patients who underwent nephrectomy for ccRCC were analyzed from April 2009 to July 2012. The mean number of chromosomal aberrations was significantly higher (7.8; P < .05) in Fuhrman grade 4 (F4) than in F3 (4) and F2 (3.4) cases. The results were similar, considering separately the mean number of chromosomal losses and gains. The F4 cases had a distinct pattern with more frequent losses of chromosomes 9, 13, 14, 18, 21, 22, and Y and gains of chromosome 20. Necrosis was associated with losses of chromosomes 7, 9, 18, and 22; sarcomatoid component, losses of chromosomes 7, 9, and 14 and gains of 20; and T stage, losses of chromosomes 18 and Y. After multivariate analysis, renal fat invasion, renal vein emboli, and microscopic vascular invasion were, respectively, associated with losses of chromosomes 13 and Y, loss of chromosome 13, and loss of chromosome 14 and gains of chromosomes 7 and 20. F4 was independently associated with losses of chromosomes 9 and Y; sarcomatoid component, loss of chromosome 9 and gain of 20; necrosis, loss of chromosome 18; and T stage, loss of chromosome Y. These chromosomal imbalances can be detected routinely by karyotype or fluorescence in situ hybridization analyses to stratify patients for risk of progression.
Collapse
|
81
|
Albers J, Rajski M, Schönenberger D, Harlander S, Schraml P, von Teichman A, Georgiev S, Wild PJ, Moch H, Krek W, Frew IJ. Combined mutation of Vhl and Trp53 causes renal cysts and tumours in mice. EMBO Mol Med 2013; 5:949-64. [PMID: 23606570 PMCID: PMC3779454 DOI: 10.1002/emmm.201202231] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 03/08/2013] [Accepted: 03/12/2013] [Indexed: 12/03/2022] Open
Abstract
The combinations of genetic alterations that cooperate with von Hippel–Lindau (VHL) mutation to cause clear cell renal cell carcinoma (ccRCC) remain poorly understood. We show that the TP53 tumour suppressor gene is mutated in approximately 9% of human ccRCCs. Combined deletion of Vhl and Trp53 in primary mouse embryo fibroblasts causes proliferative dysregulation and high rates of aneuploidy. Deletion of these genes in the epithelium of the kidney induces the formation of simple cysts, atypical cysts and neoplasms, and deletion in the epithelia of the genital urinary tract leads to dysplasia and tumour formation. Kidney cysts display a reduced frequency of primary cilia and atypical cysts and neoplasms exhibit a pro-proliferative signature including activation of mTORC1 and high expression of Myc, mimicking several cellular and molecular alterations seen in human ccRCC and its precursor lesions. As the majority of ccRCC is associated with functional inactivation of VHL, our findings suggest that for a subset of ccRCC, loss of p53 function represents a critical event in tumour development.
Collapse
Affiliation(s)
- Joachim Albers
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Kroeger N, Klatte T, Chamie K, Rao PN, Birkhäuser FD, Sonn GA, Riss J, Kabbinavar FF, Belldegrun AS, Pantuck AJ. Deletions of chromosomes 3p and 14q molecularly subclassify clear cell renal cell carcinoma. Cancer 2013; 119:1547-54. [PMID: 23335244 DOI: 10.1002/cncr.27947] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/05/2012] [Accepted: 12/10/2012] [Indexed: 11/08/2022]
Abstract
BACKGROUND The short arm of chromosome 3 (3p) harbors the von Hippel-Lindau (VHL) tumor suppressor gene, and the long arm of chromosome 14 (14q) harbors the hypoxia-inducible factor 1α (HIF-1α) gene. The objective of this study was to evaluate the significance of 3p loss (loss VHL gene) and 14q loss (loss HIF-1α gene) in clear cell renal cell carcinoma (ccRCC). METHODS In total, 288 ccRCC tumors underwent a prospective cytogenetic analysis for alterations in chromosomes 3p and 14q. Tumors were assigned to 1 of 4 possible chromosomal alterations: VHL +3p/+14q (VHL wild type [VHL-WT]), VHL +3p/-14q (VHL-WT plus HIF2α [WT/H2]), -3p/+14q (HIF1α and HIF2α [H1H2]), and -3p/-14q (HIF2α [H2]). RESULTS Among patients who had loss of 3p, tumors with -3p/-14q (H2) alterations were larger (P = .002), had higher grade (P = .002) and stage (P = .001), and more often were metastatic (P = .029) than tumors that retained 14q (H1H2). All patients who had tumors with -3p/-14q (H2) had worse cancer-specific survival (P = .014), and patients who had localized disease (P = .012) and primary T1 (pT1) tumors (P = .008) had worse recurrence-free survival. In patients who had pT1 tumors, combined 3p/14q loss was an independent predictor of recurrence-free survival (hazard ratio, 11.19; 95% confidence interval, 1.91-65.63) and cancer-specific survival (hazard ratio, 15.93; 95% confidence interval, 3.09-82.16). The current investigation was limited by its retrospective design, single-center experience, and a lack of confirmatory protein analyses. CONCLUSIONS Loss of chromosome 3p (the VHL gene) was associated with improved survival in patients with ccRCC, whereas loss of chromosome 14q (the HIF-1α gene) was associated with worse outcomes. The results of the current study support the hypothesis that HIF-1α functions as an important tumor suppressor gene in ccRCC.
Collapse
Affiliation(s)
- Nils Kroeger
- Institute of Urologic Oncology, Department of Urology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Junker K, Ficarra V, Kwon ED, Leibovich BC, Thompson RH, Oosterwijk E. Potential role of genetic markers in the management of kidney cancer. Eur Urol 2012; 63:333-40. [PMID: 23040205 DOI: 10.1016/j.eururo.2012.09.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 09/14/2012] [Indexed: 02/07/2023]
Abstract
CONTEXT Kidney cancer is not a single entity but comprises a number of different types of cancer that occur in the kidney including renal cell tumours as the most common type. Four major renal cell tumour subtypes can be distinguished based on morphologic and genetic characteristics. To individualise therapy and to improve the prognosis in patients with renal cell tumours, accurate subtyping, definition of individual course of disease, and the prediction of therapy response are necessary. OBJECTIVE To discuss the potential role of genetic markers in the management of kidney cancer. EVIDENCE ACQUISITION A Medline search was conducted to identify original articles, review articles, and editorials addressing the role of genetic alterations in kidney cancer management. Keywords included kidney neoplasms, genetics, SNP, gene expression, miRNA, classification, diagnosis, drug therapy, prognosis, and therapy. The articles with the highest level of evidence were identified and critically reviewed. This review is the result of an interactive peer-reviewing process by an expert panel of co-authors. EVIDENCE SYNTHESIS Each subtype is characterised by specific genetic, epigenetic, and expression patterns that potentially can be used to subclassify renal cell tumours in cases of ambivalent histopathology. Molecular signatures and single alterations in primary tumours are associated with aggressiveness and prognosis. Germline polymorphisms in specific genes encoding for metabolizing enzymes, efflux transporters, and drug targets seem to be associated with toxicity and response in patients receiving targeted therapy. CONCLUSIONS Significant advances have been achieved in the molecular analysis of renal cancer. Validation of findings is greatly needed to implement genetic markers in the management of renal cancer. This should lead to improved diagnosis, prognosis, and personalised therapy in this heterogeneous disease.
Collapse
Affiliation(s)
- Kerstin Junker
- Clinic of Urology and Pediatric Urology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.
| | | | | | | | | | | |
Collapse
|
84
|
Girgis AH, Iakovlev VV, Beheshti B, Bayani J, Squire JA, Bui A, Mankaruos M, Youssef Y, Khalil B, Khella H, Pasic M, Yousef GM. Multilevel whole-genome analysis reveals candidate biomarkers in clear cell renal cell carcinoma. Cancer Res 2012; 72:5273-84. [PMID: 22926558 DOI: 10.1158/0008-5472.can-12-0656] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC) is the most common neoplasm of the kidney. We conducted an integrated analysis of copy number, gene expression (mRNA and miRNA), protein expression, and methylation changes in clear cell renal cell carcinoma (ccRCC). We used a stepwise approach to identify the most significant copy number aberrations (CNA) and identified regions of peak and broad copy number gain and loss, including peak gains (3q21, 5q32, 5q34-q35, 7p11, 7q21, 8q24, 11q13, and 12q14) and deletions (1p36, 2q34-q37, 3p25, 4q33-q35, 6q23-q27, and 9p21). These regions harbor novel tumor-related genes and miRNAs not previously reported in renal carcinoma. Integration of genome-wide expression data and gene set enrichment analysis revealed 75 gene sets significantly altered in tumors with CNAs compared with tumors without aberration. We also identified genes located in peak CNAs with concordant methylation changes (hypomethylated in copy number gains such as STC2 and CCND1 and hypermethylated in deletions such as CLCNKB, VHL, and CDKN2A/2B). For other genes, such as CA9, expression represents the net outcome of opposing forces (deletion and hypomethylation) that also significantly influences patient survival. We also validated the prognostic value of miRNA let-7i in RCCs. miR-138, located in chromosome 3p deletion, was also found to have suppressive effects on tumor proliferation and migration abilities. Our findings provide a significant advance in the delineation of the ccRCC genome by better defining the impact of CNAs in conjunction with methylation changes on the expression of cancer-related genes, miRNAs, and proteins and their influence on patient survival.
Collapse
Affiliation(s)
- Andrew H Girgis
- Department of Laboratory Medicine, and the Keenan Research Centre in the Li Ka Shing Knowledge Institute St. Michael's Hospital, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Chiyomaru T, Yamamura S, Zaman MS, Majid S, Deng G, Shahryari V, Saini S, Hirata H, Ueno K, Chang I, Tanaka Y, Tabatabai ZL, Enokida H, Nakagawa M, Dahiya R. Genistein suppresses prostate cancer growth through inhibition of oncogenic microRNA-151. PLoS One 2012; 7:e43812. [PMID: 22928040 PMCID: PMC3426544 DOI: 10.1371/journal.pone.0043812] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/26/2012] [Indexed: 01/12/2023] Open
Abstract
Genistein has been shown to suppress the growth of several cancers through modulation of various pathways. However, the effects of genistein on the regulation of oncogenic microRNA-151 (miR-151) have not been reported. In this study, we investigated whether genistein could alter the expression of oncogenic miR-151 and its target genes that are involved in the progression and metastasis of prostate cancer (PCa). Real-time RT-PCR showed that the expression of miR-151 was higher in PC3 and DU145 cells compared with RWPE-1 cells. Treatment of PC3 and DU145 cells with 25 µM genistein down-regulated the expression of miR-151 compared with vehicle control. Inhibition of miR-151 in PCa cells by genistein significantly inhibited cell migration and invasion. In-silico analysis showed that several genes (CASZ1, IL1RAPL1, SOX17, N4BP1 and ARHGDIA) suggested to have tumor suppressive functions were target genes of miR-151. Luciferase reporter assays indicated that miR-151 directly binds to specific sites on the 3′UTR of target genes. Quantitative real-time PCR analysis showed that the mRNA expression levels of the five target genes in PC3 and DU145 were markedly changed with miR-151 mimics and inhibitor. Kaplan-Meier curves and log-rank tests revealed that high expression levels of miR-151 had an adverse effect on survival rate. This study suggests that genistein mediated suppression of oncogenic miRNAs can be an important dietary therapeutic strategy for the treatment of PCa.
Collapse
Affiliation(s)
- Takeshi Chiyomaru
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Soichiro Yamamura
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Mohd Saif Zaman
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Shahana Majid
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Guoren Deng
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Varahram Shahryari
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Sharanjot Saini
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Hiroshi Hirata
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Koji Ueno
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Inik Chang
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Yuichiro Tanaka
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Z. Laura Tabatabai
- Department of Pathology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Hideki Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masayuki Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Rajvir Dahiya
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
86
|
Jonasch E, Futreal A, Davis I, Bailey S, Kim WY, Brugarolas J, Giaccia A, Kurban G, Pause A, Frydman J, Zurita A, Rini BI, Sharma P, Atkins M, Walker C, Rathmell WK. State of the science: an update on renal cell carcinoma. Mol Cancer Res 2012; 10:859-80. [PMID: 22638109 PMCID: PMC3399969 DOI: 10.1158/1541-7786.mcr-12-0117] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Renal cell carcinomas (RCC) are emerging as a complex set of diseases that are having a major socioeconomic impact and showing a continued rise in incidence throughout the world. As the field of urologic oncology faces these trends, several major genomic and mechanistic discoveries are altering our core understanding of this multitude of cancers, including several new rare subtypes of renal cancers. In this review, these new findings are examined and placed in the context of the well-established association of clear cell RCC (ccRCC) with mutations in the von Hippel-Lindau (VHL) gene and resultant aberrant hypoxia inducible factor (HIF) signaling. The impact of novel ccRCC-associated genetic lesions on chromatin remodeling and epigenetic regulation is explored. The effects of VHL mutation on primary ciliary function, extracellular matrix homeostasis, and tumor metabolism are discussed. Studies of VHL proteostasis, with the goal of harnessing the proteostatic machinery to refunctionalize mutant VHL, are reviewed. Translational efforts using molecular tools to elucidate discriminating features of ccRCC tumors and develop improved prognostic and predictive algorithms are presented, and new therapeutics arising from the earliest molecular discoveries in ccRCC are summarized. By creating an integrated review of the key genomic and molecular biological disease characteristics of ccRCC and placing these data in the context of the evolving therapeutic landscape, we intend to facilitate interaction among basic, translational, and clinical researchers involved in the treatment of this devastating disease, and accelerate progress toward its ultimate eradication.
Collapse
Affiliation(s)
| | | | - Ian Davis
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Sean Bailey
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - William Y. Kim
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | | | | | | | | | | | - Brian I. Rini
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH
| | - Pam Sharma
- University of Texas-Houston Medical Center, Houston, TX
| | | | - Cheryl Walker
- University of Texas-Houston Medical Center, Houston, TX
| | | |
Collapse
|
87
|
Shen C, Kaelin WG. The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol 2012; 23:18-25. [PMID: 22705278 DOI: 10.1016/j.semcancer.2012.06.001] [Citation(s) in RCA: 298] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 06/05/2012] [Indexed: 02/08/2023]
Abstract
Inactivation of the VHL tumor suppressor protein (pVHL) is a common event in clear cell renal carcinoma, which is the most common form of kidney cancer. pVHL performs many functions, including serving as the substrate recognition module of an ubiquitin ligase complex that targets the alpha subunits of the heterodimeric HIF transcription factor for proteasomal degradation. Deregulation of HIF2α appears to be a driving force in pVHL-defective clear cell renal carcinomas. In contrast, genetic and functional studies suggest that HIF1α serves as a tumor suppressor and is a likely target of the 14q deletions that are characteristic of this tumor type. Drugs that inhibit HIF2α, or its downstream targets such as VEGF, are in various stages of clinical testing. Indeed, clear cell renal carcinomas are exquisitely sensitive to VEGF deprivation and four VEGF inhibitors have now been approved for the treatment of this disease.
Collapse
Affiliation(s)
- Chuan Shen
- Howard Hughes Medical Insititute, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, United States
| | | |
Collapse
|
88
|
Hu X, Chen H, Jin M, Wang X, Lee J, Xu W, Zhang R, Li S, Niu J. Molecular cytogenetic characterization of undifferentiated embryonal sarcoma of the liver: a case report and literature review. Mol Cytogenet 2012; 5:26. [PMID: 22551002 PMCID: PMC3478990 DOI: 10.1186/1755-8166-5-26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 02/26/2012] [Indexed: 12/14/2022] Open
Abstract
Undifferentiated embryonal sarcoma of the liver (UESL) represents a heterogeneous group of tumors derived from mesenchymal tissues. Earlier cytogenetic studies in limited cases demonstrated that UESL is associated with a recurrent translocation t(11;19)(q11;q13.3-q13.4) or add(19)(q13.4). In this report, we present our array comparative genomic hybridization (aCGH), fluorescence in situ hybridization (FISH) findings, and a missense mutation of TP53 gene by DNA sequencing in a 19-year-old patient with UESL. The data were compared to laboratory findings reported by previous studies.
Collapse
Affiliation(s)
- Xiaoxia Hu
- Department of Internal Medicine, The First Hospital of Jilin University, Jilin 130021, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Farber LJ, Furge K, Teh BT. Renal Cell Carcinoma Deep Sequencing: Recent Developments. Curr Oncol Rep 2012; 14:240-8. [DOI: 10.1007/s11912-012-0230-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
90
|
Quiroga-Garza G, Piña-Oviedo S, Cuevas-Ocampo K, Goldfarb R, Schwartz MR, Ayala AG, Monzon FA. Synchronous clear cell renal cell carcinoma and tubulocystic carcinoma: genetic evidence of independent ontogenesis and implications of chromosomal imbalances in tumor progression. Diagn Pathol 2012; 7:21. [PMID: 22369180 PMCID: PMC3313901 DOI: 10.1186/1746-1596-7-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 02/27/2012] [Indexed: 12/13/2022] Open
Abstract
Seven percent of renal cell carcinoma (RCC) cases are diagnosed as "unclassified" RCC by morphology. Genetic profiling of RCCs helps define renal tumor subtypes, especially in cases where morphologic diagnosis is inconclusive. This report describes a patient with synchronous clear cell RCC (ccRCC) and a tubulocystic renal carcinoma (TCRC) in the same kidney, and discusses the pathologic features and genetic profile of both tumors. A 67 year-old male underwent CT scans for an unrelated medical event. Two incidental renal lesions were found and ultimately removed by radical nephrectomy. The smaller lesion had multiple small cystic spaces lined by hobnail cells with high nuclear grade separated by fibrous stroma. This morphology and the expression of proximal (CD10, AMACR) and distal tubule cell (CK19) markers by immunohistochemistry supported the diagnosis of TCRC. The larger lesion was a typical ccRCC, with Fuhrman's nuclear grade 3 and confined to the kidney. Molecular characterization of both neoplasms using virtual karyotyping was performed to assess relatedness of these tumors. Low grade areas (Fuhrman grade 2) of the ccRCC showed loss of 3p and gains in chromosomes 5 and 7, whereas oncocytic areas displayed additional gain of 2p and loss of 10q; the high grade areas (Fuhrman grade 3) showed several additional imbalances. In contrast, the TCRC demonstrated a distinct profile with gains of chromosomes 8 and 17 and loss of 9. In conclusion, ccRCC and TCRC show distinct genomic copy number profiles and chromosomal imbalances in TCRC might be implicated in the pathogenesis of this tumor. Second, the presence of a ccRCC with varying degrees of differentiation exemplifies the sequence of chromosomal imbalances acquired during tumor progression.
Collapse
Affiliation(s)
- Gabriela Quiroga-Garza
- Department of Pathology and Genomic Medicine, The Methodist Hospital, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|