1
|
Stephen SA, Wainman LM, Lefferts JA, Pettus JR. Shared chromosome 21q loss in a mixed subtype renal cell carcinoma: composite or collision tumor? Virchows Arch 2024:10.1007/s00428-024-03820-w. [PMID: 38780616 DOI: 10.1007/s00428-024-03820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
Clear cell renal cell carcinoma (CCRCC) and papillary renal cell carcinoma (PRCC) are the two most frequently encountered subtypes of renal cell carcinoma (RCC). Rarely, these two entities are identified intermingled within the same mass and have been labeled either collision tumors juxtaposed by random chance or composite tumors that have arisen from a common tumorigenic precursor cell. Regarding this distinction, authors have commonly relied upon macroscopic, histologic, and clinicopathologic findings, which may be prone to subjectivity. Objective molecular evidence has been lacking. We present a renal tumor showing a mixed CCRCC and PRCC with corroborating histologic, immunophenotypic, chromosomal microarray analysis (CMA), and next-generation sequencing (NGS) analysis for the respective tumor components, including classic findings of chromosome 3p loss and VHL mutation within the CCRCC component and gain of chromosomes 7 and 17 within the PRCC component. Of novel interest, CMA revealed a shared loss of chromosome 21q in both components with no other identifiable shared or overlapping mutations. This report adds unique evidence supporting the possibility of a true composite renal cell carcinoma composed of two commonly recognized subtypes. This finding may help to inform early molecular pathogenetic mechanism of RCC tumorigenesis.
Collapse
Affiliation(s)
| | - Lauren M Wainman
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Joel A Lefferts
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jason R Pettus
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
2
|
Jia L, Cowell LG, Kapur P. Understanding Factors that Influence Prognosis and Response to Therapy in Clear Cell Renal Cell Carcinoma. Adv Anat Pathol 2024; 31:96-104. [PMID: 38179997 DOI: 10.1097/pap.0000000000000428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
In this review, we highlight and contextualize emerging morphologic prognostic and predictive factors in renal cell carcinoma. We focus on clear cell renal cell carcinoma (ccRCC), the most common histologic subtype. Our understanding of the molecular characterization of ccRCC has dramatically improved in the last decade. Herein, we highlight how these discoveries have laid the foundation for new approaches to prognosis and therapeutic decision-making for patients with ccRCC. We explore the clinical relevance of common mutations, established gene expression signatures, intratumoral heterogeneity, sarcomatoid/rhabdoid morphology and PD-L1 expression, and discuss their impact on predicting response to therapy.
Collapse
Affiliation(s)
| | - Lindsay G Cowell
- Peter O'Donnell School of Public Health
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| | - Payal Kapur
- Department of Pathology
- Department of Urology, University of Texas Southwestern Medical Center
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| |
Collapse
|
3
|
Batavia AA, Rutishauser D, Sobottka B, Schraml P, Beerenwinkel N, Moch H. Biallelic ELOC-Inactivated Renal Cell Carcinoma: Molecular Features Supporting Classification as a Distinct Entity. Mod Pathol 2023; 36:100194. [PMID: 37088333 DOI: 10.1016/j.modpat.2023.100194] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/28/2023] [Accepted: 04/16/2023] [Indexed: 04/25/2023]
Abstract
Approximately 70% of clear cell renal cell carcinoma (ccRCC) is characterized by the biallelic inactivation of von Hippel-Lindau (VHL) on chromosome 3p. ELOC-mutated (Elongin C-mutated) renal cell carcinoma containing biallelic ELOC inactivations with chromosome 8q deletions is considered a novel subtype of renal cancer possessing a morphologic overlap with ccRCC, renal cell carcinoma (RCC) with fibromyomatous stroma exhibiting Tuberous Sclerosis Complex (TSC)/mammalian Target of Rapamycin (mTOR) mutations, and clear cell papillary tumor. However, the frequency and consequences of ELOC alterations in wild-type VHL and mutated VHL RCC are unclear. In this study, we characterize 123 renal tumors with clear cell morphology and known VHL mutation status to assess the morphologic and molecular consequences of ELOC inactivation. Using OncoScan and whole-exome sequencing, we identify 18 ELOC-deleted RCCs, 3 of which contain ELOC mutations resulting in the biallelic inactivation of ELOC. Biallelic ELOC and biallelic VHL aberrations were mutually exclusive; however, 2 ELOC-mutated RCCs showed monoallelic VHL alterations. Furthermore, no mutations in TSC1, TSC2, or mTOR were identified in ELOC-mutated RCC with biallelic ELOC inactivation. Using High Ambiguity Driven biomolecular DOCKing, we report a novel ELOC variant containing a duplication event disrupting ELOC-VHL interaction alongside the frequently seen Y79C alteration. Using hyper reaction monitoring mass spectrometry, we show RCCs with biallelic ELOC alterations have significantly reduced ELOC expression but similar carbonic anhydrase 9 and vascular endothelial growth factor A expression compared with classical ccRCC with biallelic VHL inactivation. The absence of biallelic VHL and TSC1, TSC2, or mTOR inactivation in RCC with biallelic ELOC inactivation (ELOC mutation in combination with ELOC deletions on chromosome 8q) supports the notion of a novel, molecularly defined tumor entity.
Collapse
Affiliation(s)
- Aashil A Batavia
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dorothea Rutishauser
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bettina Sobottka
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Kapur P, Rajaram S, Brugarolas J. The expanding role of BAP1 in clear cell renal cell carcinoma. Hum Pathol 2023; 133:22-31. [PMID: 35932824 PMCID: PMC9898467 DOI: 10.1016/j.humpath.2022.07.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Mutations drive renal cell carcinoma biology and tumor growth. The BRCA1-associated protein-1 (BAP1) gene is frequently mutated in clear cell renal cell carcinoma (ccRCC) and has emerged as a prognostic and putative predictive biomarker. In this review, we discuss the role of BAP1 as a signature event of a subtype of ccRCC marked by aggressiveness, inflammation, and possibly a heightened response to immunotherapy.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA.
| | - Satwik Rajaram
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA; Department of Internal Medicine (Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
5
|
Onal B, Gultekin MH, Simsekoglu MF, Selcuk B, Gurbuz A. Biomarkers in Urological Cancers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Urological tumours have become one of the most common cancers in the
last decade. It is important to apply an approach that evaluates many factors related to
the patient and the disease carefully to minimize cancer-associated morbidity and
mortality. The clinical use of cancer biomarkers is a valuable part of the clinical
management of urological cancers. These biomarkers may lead to optimized detection,
treatment, and follow-up of urological cancers. With the development of molecular
research, newly developed biomarkers and next-generation sequencing have also
contributed to patient management. In this chapter, we will present biomarkers in the
most common urological cancers under subheadings of bladder cancer, prostate cancer,
kidney cancer, and testicular cancer. Additionally, due to the development that
occurred in the next-generation sequencing (NGS), all the above-mentioned
malignancies are evaluated with regard to NGS.
Collapse
Affiliation(s)
- Bulent Onal
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Mehmet Hamza Gultekin
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Muhammed Fatih Simsekoglu
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Berin Selcuk
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| | - Ahmet Gurbuz
- Department of Urology, Cerrahpasa School of Medicine, Istanbul University - Cerrahpasa,
Istanbul, Turkey
| |
Collapse
|
6
|
Millet-Boureima C, He S, Le TBU, Gamberi C. Modeling Neoplastic Growth in Renal Cell Carcinoma and Polycystic Kidney Disease. Int J Mol Sci 2021; 22:3918. [PMID: 33920158 PMCID: PMC8070407 DOI: 10.3390/ijms22083918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) and autosomal dominant polycystic kidney disease (ADPKD) share several characteristics, including neoplastic cell growth, kidney cysts, and limited therapeutics. As well, both exhibit impaired vasculature and compensatory VEGF activation of angiogenesis. The PI3K/AKT/mTOR and Ras/Raf/ERK pathways play important roles in regulating cystic and tumor cell proliferation and growth. Both RCC and ADPKD result in hypoxia, where HIF-α signaling is activated in response to oxygen deprivation. Primary cilia and altered cell metabolism may play a role in disease progression. Non-coding RNAs may regulate RCC carcinogenesis and ADPKD through their varied effects. Drosophila exhibits remarkable conservation of the pathways involved in RCC and ADPKD. Here, we review the progress towards understanding disease mechanisms, partially overlapping cellular and molecular dysfunctions in RCC and ADPKD and reflect on the potential for the agile Drosophila genetic model to accelerate discovery science, address unresolved mechanistic aspects of these diseases, and perform rapid pharmacological screens.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Thi Bich Uyen Le
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore 119228, Singapore
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29528-6054, USA
| |
Collapse
|
7
|
Liu K, Gao R, Wu H, Wang Z, Han G. Single-cell analysis reveals metastatic cell heterogeneity in clear cell renal cell carcinoma. J Cell Mol Med 2021; 25:4260-4274. [PMID: 33759378 PMCID: PMC8093989 DOI: 10.1111/jcmm.16479] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the leading causes of cancer-related death worldwide. Tumour metastasis and heterogeneity lead to poor survival outcomes and drug resistance in patients with metastatic RCC (mRCC). In this study, we aimed to assess intratumoural heterogeneity (ITH) in mRCC cells by performing a combined analysis of bulk data and single-cell RNA-sequencing data, and develop novel biomarkers for prognosis prediction on the basis of the potential molecular mechanisms underlying tumorigenesis. Eligible single-cell cohorts related to mRCC were acquired using the Gene Expression Omnibus (GEO) dataset to identify potential mRCC subpopulations. We then performed gene set variation analysis to understand the differential function in primary RCC and mRCC samples. Subsequently, we applied weighted correlation network analysis to identify coexpressing gene modules that were related to the external trait of metastasis. Protein-protein interactions were used to screen hub subpopulation-difference (sub-dif) markers (ACTG1, IL6, CASP3, ACTB and RAP1B) that might be involved in the regulation of RCC metastasis and progression. Cox regression analysis revealed that ACTG1 was a protective factor (HR < 1), whereas the other four genes (IL6, CASP3, ACTB and RAP1B) were risk factors (HR > 1). Kaplan-Meier survival analysis suggested the potential prognostic value of these sub-dif markers. The expression of sub-dif markers in mRCC was further evaluated in clinical samples by immunohistochemistry (IHC). Additionally, the genetic features of sub-dif marker expression patterns, such as genetic variation profiles, correlations with tumour-infiltrating lymphocytes (TILs), and targeted signalling pathway activities, were assessed in bulk RNA-seq datasets. In conclusion, we established novel subpopulation markers as key prognostic factors affecting EMT-related signalling pathway activation in mRCC, which could facilitate the implementation of a treatment for mRCC patients.
Collapse
Affiliation(s)
- Kun Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Gao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hao Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhe Wang
- Department of Gastrointestinal Oncology, Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Wang L, Li Y, Lyu Y, Wen H, Feng C. Association between copy-number alteration of +20q, -14q and -18p and cross-sensitivity to tyrosine kinase inhibitors in clear-cell renal cell carcinoma. Cancer Cell Int 2020; 20:482. [PMID: 33041663 PMCID: PMC7541266 DOI: 10.1186/s12935-020-01585-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
Background We aim to explore association between copy number alteration (CNA) and sensitivity to common tyrosine kinase inhibitors (TKIs) used in clear-cell renal cell carcinoma (ccRCC) treatment. Methods CNA with related sensitivity profiles were extracted from the Genomics of Drug Sensitivity in Cancer (GDSC) dataset and was cross-referenced with common CNA in ccRCC in the Cancer Genome Atlas (TCGA) dataset. Functional annotation was profiled using GSEA and NET-GE. Target genes within cytobands of interest were screened in silico and validated in vitro using proliferation assays in A498 and 786-O ccRCC cells. Results Four TKIs (Sunitinib, Cabozantinib, Axitinib and Sorafenib) that were clinically used in ccRCC were selected. In silico analysis showed gain of 20q (+20q) occurred in ~ 23% of cases and was associated with resistance to all four TKIs; loss of 14q (−14q) occurred in ~ 39% of cases and was associated with resistance to Sunitinib and Sorafenib; loss of 18p (−18p) occurred in ~ 39% of cases and was associated with sensitivity to Sunitinib and Sorafenib. All 3 CNAs were associated with worsened prognosis, respectively. Candidate target genes included of RBL1 on 20q, KLHL33 on 14q and ARHGAP28 on18q. In vitro validation showed RBL1 overexpression induced resistance to Sunitinib and Cabozantinib; KLHL33 silencing induced resistance to Sunitinib; ARHGAP28 silencing induced sensitivity to Cabozantinib. Functional annotation indicated FoxO signaling, hypoxic response and Wnt pathway, and Rho-related cellular adhesion were mechanistically associated with +20q, −14q and −18p, respectively. Conclusion Common CNAs in ccRCC are associated with cancer-intrinsic cross-sensitivity to common TKIs. Further validation and functional analyses are therefore needed.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, 300052 People's Republic of China
| | - Yuqing Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Yinfeng Lyu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Hui Wen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| |
Collapse
|
9
|
Chromosome 17p13 deletion is associated with an aggressive tumor phenotype in clear cell renal cell carcinoma. World J Surg Oncol 2020; 18:128. [PMID: 32534597 PMCID: PMC7293794 DOI: 10.1186/s12957-020-01902-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Deletions of 17p13 recurrently occur in renal cell carcinoma (RCC) but their prognostic role seems to be uncertain. Methods To determine prevalence, relationship with tumor phenotype, and patient prognosis, a tissue microarray containing samples from 1809 RCCs was evaluated using dual labeling fluorescence in situ hybridization (FISH) with 17p13 and chromosome 17 centromere probes. Results A 17p13 deletion was found in 72 of 1429 interpretable tumors. The frequency of 17p13 deletions varied greatly between RCC subtypes and was highest in chromophobe RCC (24/72; 33.3%). 17p13 deletions were also found in 35 (3.7%) of 946 clear cell RCC, 9 (4.3%) of 208 papillary RCC, 1 of 121 oncocytomas (0.8%), as well as in several rare cases of comprising 1 of 7 Xp11.2 translocation cancers, 1 of 3 collecting duct carcinomas, and 1 of 20 not otherwise specified (NOS) carcinomas. In clear cell carcinomas, 17p13 deletions revealed a strong and consistent association with higher Fuhrman, ISUP, and Thoenes grade (p < 0.0001 each), and linked to advanced tumor stage (p = 0.0168), large tumor diameter (p = 0.0004), distant metastases (p = 0.0077), cancer-specific survival (p = 0.0391), and recurrence-free survival (p = 0.0072). In multivariate analysis, 17p13 deletions showed in clear cell RCC a dependent prognostic role for established clinical-pathological parameters. Conclusion 17p13 deletions have a dual role in RCC. They are associated with disease progression in clear cell RCC and possibly other subtypes and they are linked to the development of chromophobe RCC—a subtype with a particularly favorable prognosis.
Collapse
|
10
|
Tumkur Sitaram R, Landström M, Roos G, Ljungberg B. Significance of PI3K signalling pathway in clear cell renal cell carcinoma in relation to VHL and HIF status. J Clin Pathol 2020; 74:216-222. [PMID: 32467322 DOI: 10.1136/jclinpath-2020-206693] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/10/2020] [Indexed: 12/30/2022]
Abstract
Renal cell carcinoma (RCC) includes diverse tumour types characterised by various genetic abnormalities. The genetic changes, like mutations, deletions and epigenetic alterations, play a crucial role in the modification of signalling networks, tumour pathogenesis and prognosis. The most prevalent RCC type, clear cell RCC (ccRCC), is asymptomatic in the early stages and has a poorer prognosis compared with the papillary and the chromophobe types RCCs. Generally, ccRCC is refractory to chemotherapy and radiation therapy. Loss of von Hippel-Lindau (VHL) gene and upregulation of hypoxia-inducible factors (HIF), the signature of most sporadic ccRCC, promote multiple growth factors. Hence, VHL/HIF and a variety of pathways, including phosphatase and TEnsin homolog on chromosome 10/phosphatidylinositol-3-kinase (PI3K)/AKT, are closely connected and contribute to the ontogeny of ccRCC. In the recent decade, multiple targeting agents have been developed based on blocking major signalling pathways directly or indirectly involved in ccRCC tumour progression, metastasis, angiogenesis and survival. However, most of these drugs have limitations; either metastatic ccRCC develops resistance to these agents, or despite blocking receptors, tumour cells use alternate signalling pathways. This review compiles the state of knowledge about the PI3K/AKT signalling pathway confined to ccRCC and its cross-talks with VHL/HIF pathway.
Collapse
Affiliation(s)
- Raviprakash Tumkur Sitaram
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Maréne Landström
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Göran Roos
- Department of Medical Biosciences, Pathology, Translational Research Center (TRC), Umeå Universitet, Umeå, Väasterbotten, Sweden
| | - Börje Ljungberg
- Department of Surgical and Preoperative Sciences, Urology and Andrology, Umeå Universitet, Umea, Västerbotten, Sweden
| |
Collapse
|
11
|
Liu YJ, Houldsworth J, Emmadi R, Dyer L, Wolff DJ. Assessing Genomic Copy Number Alterations as Best Practice for Renal Cell Neoplasia: An Evidence-Based Review from the Cancer Genomics Consortium Workgroup. Cancer Genet 2020; 244:40-54. [PMID: 32434132 DOI: 10.1016/j.cancergen.2020.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/05/2023]
Abstract
Renal cell neoplasia are heterogeneous with diverse histology, genetic alterations, and clinical behavior that are diagnosed mostly on morphologic features. The Renal Cell Neoplasia Workgroup of the Cancer Genomics Consortium systematically evaluated peer-reviewed literature on genomic studies of renal cell carcinoma (RCC), including clear cell RCC, papillary RCC, chromophobe RCC, and the translocation RCC involving TFE3, TFEB and MITF rearrangements, as well as benign oncocytoma, which together comprise about 95% of all renal cell neoplasia. The Workgroup curated recurrent copy number alterations (CNAs), copy-neutral loss-of-heterozygosity (cnLOH), rearrangements, and mutations, found in each subtype and assigned clinical relevance according to established criteria. In clear cell RCC, loss of 3p has a disease-initiating role and most likely also in progression with mutations detected in VHL and other genes mapped to this arm, and loss of 9p and/or 14q has well-substantiated prognostic utility. Gain of chromosomes 7 and 17 are hallmark CNAs of papillary RCC, but patterns of other CNAs as detected by chromosomal microarray analysis (CMA) afford sub-classification into Type 1 and 2 with prognostic value, and for further sub-stratification of Type 2. Inherent chromosome loss in chromophobe RCC as detected by CMA is useful for distinguishing the eosinophilic variant from benign oncocytoma which in contrast exhibits few CNAs or rearranged CCND1, but share mitochondrial DNA mutations. In morphologically atypical RCCs, rearrangement of TFE3 and TFEB should be considered in the differential diagnosis, portending an aggressive RCC subtype. Overall, this evidence-based review provides a validated role for assessment of CNAs in renal cell neoplasia in the clinical setting to assist in renal cell neoplasm diagnosis and sub-classification within subtypes that is integral to the management of patients, from small incidentally found renal masses to larger surgically resected specimens, and simultaneously identify the presence of key alterations portending outcome in malignant RCC subtypes.
Collapse
Affiliation(s)
- Yajuan J Liu
- Departments of Pathology and Laboratory Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA 98195.
| | - Jane Houldsworth
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Health System, 1 Gustave Levy Place, New York, NY 10029.
| | - Rajyasree Emmadi
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood Street, Chicago, IL 60612
| | - Lisa Dyer
- Department of Pediatrics, Division of Human Genetics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, MLC 4006, Cincinnati, OH 45229-3039
| | - Daynna J Wolff
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 165 Ashley Avenue, MSC 908, Charleston, SC 29425
| |
Collapse
|
12
|
Herrscher H, Boilève A, Lindner V, Barthélémy P, Hutt É, Pierard L, Kurtz JE, Rioux-Leclercq N, Lang H, Malouf GG. [MiT family translocation renal cell carcinomas: Natural history, molecular features and multidisciplinary management]. Bull Cancer 2020; 107:272-280. [PMID: 32044098 DOI: 10.1016/j.bulcan.2019.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
MiT family translocation renal cell carcinomas (tRCC) represent a rare subtype of renal cell carcinomas. These tumors have been introduced for the first time in the World Health Classification (WHO) classification of kidney cancers in 2004. tRCC are characterized by reccurent translocations involving members of the MiT family transcription factors, mainly TFE3 and TFEB. The estimated incidence of these tumors is ∼1-5 % among all renal cell carcinomas, with female prodominance. tRCC were initially described in children, and the spectrum has been expanded over time to encompass adolescents and adults. TFE3- and TFEB-rearranged RCC harbor characteristic clinicopathological and immunohistochemical features and fluorescent hybridization in situ is considered the gold standard for their diagnosis, although it has some limitations especially when the partners are located in the vicinity of TFE3. Nephron-sparing surgery is an efficient treatment of localized cases when achievable. In metastatic setting, targeted agents and immunotherapy showed modest efficacy, with response rates and median overall survival inferior to those observed in clear-cell renal cell carcinomas. Management of tRCC necessite a multidisciplinary team and accrual in clinical trials have to be encouraged when possible. Novel biological insights are urgently awaited to better understand the mechanisms associated with kidney oncogenesis in this setting, and ultimately help to identify therapeutic targets.
Collapse
Affiliation(s)
- Hugo Herrscher
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Alice Boilève
- Institut Gustave-Roussy, département de médecine, 67200 Villejuif, France
| | - Véronique Lindner
- Hôpitaux universitaires de Strasbourg, département de pathologie, 67200 Strasbourg, France
| | - Philippe Barthélémy
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Émilie Hutt
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Laure Pierard
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Jean-Emmanuel Kurtz
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Nathalie Rioux-Leclercq
- Université de Rennes, service d'anatomie et cytologie pathologiques, Centre Hospitalier Universitaire de Rennes, IRSET, 67200 Rennes, France
| | - Hervé Lang
- Hôpitaux universitaires de Strasbourg, service d'urologie, 35033 Strasbourg, France
| | - Gabriel G Malouf
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France.
| |
Collapse
|
13
|
Xiong Y, Qi Y, Bai Q, Xia Y, Liu L, Guo J. Relevance of arm somatic copy number alterations for oncologic outcomes and tumor immune microenvironment in clear cell renal cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:646. [PMID: 31930047 DOI: 10.21037/atm.2019.10.54] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Prognostic value of arm somatic copy number alterations (SCNAs) in clear cell renal cell carcinoma (ccRCC) have not been systematically evaluated in a large cohort. Its association with tumor microenvironment remained unknown. Methods We retrospectively correlated arm SCNAs with OS and recurrence free survival (RFS) in a cohort of 524 ccRCC patients. The prognostic landscape of arm SCNA was depicted by bubble heatmap. Associations between arm SCNAs and tumor microenvironment were evaluated by CIBERSORT and Gene Set Enrichment Analysis (GSEA). Results We found that amplifications of 1p, 3p and loss of 4p, 4q, 5p, 5q, 11p, 11q, 11q, 13q, 19p were independent adverse risk factor for OS, while amplification of 1q and deletions of 4p, 4q, 9p, 9q associated with worse RFS. Loss of 4q were independent adverse risk factor for OS (P=0.012, HR =1.614) and RFS (P=0.001, HR =2.005). It could identify a subset of early stage ccRCC patients with high risk of death and recurrence. CXCL9, CXCL10, CXCL11 mRNA level and CD8+ T cell infiltration were downregulated in ccRCC with 4q deletion. Patients with high arm SCNA level had shorter OS (P=0.005) and RFS (P=0.001). Markers, immune cells and pathways referring to immune suppression were elevated in tumors with high arm SCNA level. Conclusions In conclusion, loss of 4q was an independent adverse risk factor for OS and RFS in ccRCC patients and contributed to cytotoxic cell exclusion via downregulation of CXCL9, CXCL10 and CXCL11. Patients with higher arm SCNAs had worse survival and a more immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
14
|
Mendoza-Alvarez A, Guillen-Guio B, Baez-Ortega A, Hernandez-Perez C, Lakhwani-Lakhwani S, Maeso MDC, Lorenzo-Salazar JM, Morales M, Flores C. Whole-Exome Sequencing Identifies Somatic Mutations Associated With Mortality in Metastatic Clear Cell Kidney Carcinoma. Front Genet 2019; 10:439. [PMID: 31156702 PMCID: PMC6529576 DOI: 10.3389/fgene.2019.00439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/29/2019] [Indexed: 11/16/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is among the most aggressive histologic subtypes of kidney cancer, representing about 3% of all human cancers. Patients at stage IV have nearly 60% of mortality in 2–3 years after diagnosis. To date, most ccRCC studies have used DNA microarrays and targeted sequencing of a small set of well-established, commonly altered genes. An exception is the large multi-omics study of The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC), which identified new ccRCC genes based on whole exome-sequencing (WES) data, and molecular prognostic signatures based on transcriptomics, epigenetics and proteomics data. Applying WES to simultaneously interrogate virtually all exons in the human genome for somatic variation, here we analyzed the burden of coding somatic mutations in metastatic ccRCC primary tumors, and its association with patient mortality from cancer, in patients who received VEGF receptor-targeting drugs as the first-line therapy. To this end, we sequenced the exomes of ten tumor–normal pairs of ccRCC patient tissues from primary biopsies at >100× mean depth and called somatic coding variation. Mutation burden analysis prioritized 138 genes linked to patient mortality. A gene set enrichment analysis evidenced strong statistical support for the abundance of genes involved in the development of kidney cancer (p = 2.31 × 10−9) and carcinoma (p = 1.22 × 10−5), with 49 genes having direct links with kidney cancer according to the published records. Two of these genes, SIPA1L2 and EIF3A, demonstrated independent associations with mortality in TCGA-KIRC project data. Besides, three mutational signatures were found to be operative in the tumor exomes, one of which (COSMIC signature 12) has not been previously reported in ccRCC. Selection analysis yielded no detectable evidence of overall positive or negative selection, with the exome-wide number of nonsynonymous substitutions per synonymous site reflecting largely neutral tumor evolution. Despite the limited sample size, our results provide evidence for candidate genes where somatic mutation burden is tentatively associated with patient mortality in metastatic ccRCC, offering new potential pharmacological targets and a basis for further validation studies.
Collapse
Affiliation(s)
- Alejandro Mendoza-Alvarez
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Beatriz Guillen-Guio
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Adrian Baez-Ortega
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Carolina Hernandez-Perez
- Service of Medical Oncology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Sita Lakhwani-Lakhwani
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Maria-Del-Carmen Maeso
- Department of Pathology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Jose M Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Manuel Morales
- Service of Medical Oncology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Batavia AA, Schraml P, Moch H. Clear cell renal cell carcinoma with wild-type von Hippel-Lindau gene: a non-existent or new tumour entity? Histopathology 2019; 74:60-67. [PMID: 30565303 DOI: 10.1111/his.13749] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/30/2018] [Indexed: 01/01/2023]
Abstract
The current World Health Organisation (WHO) classification of renal tumours is based on characteristic histological features or specific molecular alterations. von Hippel-Lindau (VHL) alteration is the hallmark of clear cell renal cell carcinoma (RCC). After identification of the MiT translocation family of tumours, clear cell papillary renal cancer and others, the group of ccRCC with wild-type VHL is small. TCEB1 mutation combined with chromosome 8q loss is an emerging tumour entity with wild-type VHL. Inactivation of TCEB1 increases HIF stabilisation via the same mechanism as VHL inactivation. Importantly, recent molecular analyses suggest the existence of another 'VHL wild-type' evolutionary subtype of clear cell RCC in addition to TCEB1 mutated RCC and clear cell papillary renal cancer. These tumours are characterised by an aggressive behaviour, high tumour cell proliferation rate, elevated chromosomal instability and frequent presence of sarcomatoid differentiation. Future clinicopathological studies will have to provide data to determine whether TCEB1 tumours and clear cell RCC with wild-type VHL are separate tumour entities or represent variants of a clear cell RCC tumour family.
Collapse
Affiliation(s)
- Aashil A Batavia
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Evelönn EA, Landfors M, Haider Z, Köhn L, Ljungberg B, Roos G, Degerman S. DNA methylation associates with survival in non-metastatic clear cell renal cell carcinoma. BMC Cancer 2019; 19:65. [PMID: 30642274 PMCID: PMC6332661 DOI: 10.1186/s12885-019-5291-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is the most common subtype among renal cancer and is associated with poor prognosis if metastasized. Up to one third of patients with local disease at diagnosis will develop metastasis after nephrectomy, and there is a need for new molecular markers to identify patients with high risk of tumor progression. In the present study, we performed genome-wide promoter DNA methylation analysis at diagnosis to identify DNA methylation profiles associated with risk for progress. METHOD Diagnostic tissue samples from 115 ccRCC patients were analysed by Illumina HumanMethylation450K arrays and methylation status of 155,931 promoter associated CpGs were related to genetic aberrations, gene expression and clinicopathological parameters. RESULTS The ccRCC samples separated into two clusters (cluster A/B) based on genome-wide promoter methylation status. The samples in these clusters differed in tumor diameter (p < 0.001), TNM stage (p < 0.001), morphological grade (p < 0.001), and patients outcome (5 year cancer specific survival (pCSS5yr) p < 0.001 and cumulative incidence of progress (pCIP5yr) p < 0.001. An integrated genomic and epigenomic analysis in the ccRCCs, revealed significant correlations between the total number of genetic aberrations and total number of hypermethylated CpGs (R = 0.435, p < 0.001), and predicted mitotic age (R = 0.407, p < 0.001). We identified a promoter methylation classifier (PMC) panel consisting of 172 differently methylated CpGs accompanying progress of disease. Classifying non-metastatic patients using the PMC panel showed that PMC high tumors had a worse prognosis compared with the PMC low tumors (pCIP5yr 38% vs. 8%, p = 0.001), which was confirmed in non-metastatic ccRCCs in the publically available TCGA-KIRC dataset (pCIP5yr 39% vs. 16%, p < 0.001). CONCLUSION DNA methylation analysis at diagnosis in ccRCC has the potential to improve outcome-prediction in non-metastatic patients at diagnosis.
Collapse
Affiliation(s)
- Emma Andersson Evelönn
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Mattias Landfors
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Zahra Haider
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Linda Köhn
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and andrology, Umeå University, Umeå, Sweden
| | - Göran Roos
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| | - Sofie Degerman
- Department of Medical Biosciences, Umeå University, NUS, Blg 6M, 2nd floor, SE-90185 Umeå, Sweden
| |
Collapse
|
17
|
Elias R, Sharma A, Singla N, Brugarolas J. Next Generation Sequencing in Renal Cell Carcinoma: Towards Precision Medicine. KIDNEY CANCER JOURNAL : OFFICIAL JOURNAL OF THE KIDNEY CANCER ASSOCIATION 2019; 17:94-104. [PMID: 32206160 PMCID: PMC7089604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Affiliation(s)
- Roy Elias
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
- Department of Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - Akanksha Sharma
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - Nirmish Singla
- Department of Urology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - James Brugarolas
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
- Department of Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| |
Collapse
|
18
|
Kluzek K, Srebniak MI, Majer W, Ida A, Milecki T, Huminska K, van der Helm RM, Silesian A, Wrzesinski TM, Wojciechowicz J, Beverloo BH, Kwias Z, Bluyssen HAR, Wesoly J. Genetic characterization of Polish ccRCC patients: somatic mutation analysis of PBRM1, BAP1 and KDMC5, genomic SNP array analysis in tumor biopsy and preliminary results of chromosome aberrations analysis in plasma cell free DNA. Oncotarget 2018; 8:28558-28574. [PMID: 28212566 PMCID: PMC5438672 DOI: 10.18632/oncotarget.15331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/10/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Mutation analysis and cytogenetic testing in clear cell renal cell carcinoma (ccRCC) is not yet implemented in a routine diagnostics of ccRCC. MATERIAL AND METHODS We characterized the chromosomal alterations in 83 ccRCC tumors from Polish patients using whole genome SNP genotyping assay. Moreover, the utility of next generation sequencing of cell free DNA (cfDNA) in patients plasma as a potential tool for non-invasive cytogenetic analysis was tested. Additionally, tumor specific somatic mutations in PBRM1, BAP1 and KDM5C were determined. RESULTS We confirmed a correlation between deletions at 9p and higher tumor size, and deletion of chromosome 20 and the survival time. In Fuhrman grade 1, only aberrations of 3p and 8p deletion, gain of 5q and 13q and gains of chromosome 7 and 16 were present. The number of aberrations increased with Fuhrman grade, all chromosomes displayed cytogenetic changes in G3 and G4. ccRCC specific chromosome aberrations were observed in cfDNA, although discrepancies were found between cfDNA and tumor samples. In total 12 common and 94 rare variants were detected in PBRM1, BAP1 and KDM5C, with four potentially pathogenic variants. We observed markedly lower mutation load in PBRM1. CONCLUSIONS Cytogenetic analysis of cfDNA may allow more accurate diagnosis of tumor aberrations and therefore the correlation between the chromosome aberrations in cfDNA and clinical outcome should be studied in larger cohorts. The functional studies on in BAP1, KDM5C, PBRM1 mutations in large, independent sample set would be necessary for the assessment of their prognostic and diagnostic potential.
Collapse
Affiliation(s)
- Katarzyna Kluzek
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Malgorzata I Srebniak
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Weronika Majer
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Agnieszka Ida
- Department of Urology and Urological Oncology, Poznan University of Medical Sciences, 61-285 Poznan, Poland
| | - Tomasz Milecki
- Department of Urology and Urological Oncology, Poznan University of Medical Sciences, 61-285 Poznan, Poland
| | - Kinga Huminska
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland.,Genomic Laboratory, DNA Research Center, 61-612 Poznan, Poland
| | - Robert M van der Helm
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Adrian Silesian
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Tomasz M Wrzesinski
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | | | - Berna H Beverloo
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Zbigniew Kwias
- Department of Urology and Urological Oncology, Poznan University of Medical Sciences, 61-285 Poznan, Poland
| | - Hans A R Bluyssen
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| |
Collapse
|
19
|
Clinical Relevance of Gene Copy Number Variation in Metastatic Clear Cell Renal Cell Carcinoma. Clin Genitourin Cancer 2018; 16:e795-e805. [PMID: 29548613 DOI: 10.1016/j.clgc.2018.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/11/2018] [Accepted: 02/18/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND Gene copy number variations (CNVs) have been reported to be frequent in renal cell carcinoma (RCC), with potential prognostic value for some. However, their clinical utility, especially to guide treatment of metastatic disease remains to be established. Our objectives were to assess CNVs on a panel of selected genes and determine their clinical relevance in patients who underwent treatment of metastatic RCC. PATIENTS AND METHODS The genetic assessment was performed on frozen tissue samples of clear cell metastatic RCC using quantitative multiplex polymerase chain reaction of short fluorescent fragment method to detect CNVs on a panel of 14 genes of interest. The comparison of the electropherogram obtained from both tumor and normal renal adjacent tissue allowed for CNV identification. The clinical, biologic, and survival characteristics were assessed for their associations with the most frequent CNVs. RESULTS Fifty patients with clear cell metastatic RCC were included. The CNV rate was 21.4%. The loss of CDKN2A and PLG was associated with a higher tumor stage (P < .05). The loss of PLG and ALDOB was associated with a higher Fuhrman grade (P < .05). The loss of ALDOB was also associated with a worse Heng prognostic score (95% vs. 66%; P = .029) and lower 24-month survival rate (18% vs. 58%; P = .012). The loss of both ALDOB and PLG was frequent (32%) and was associated with a higher tumor stage and grade (P < .05). CONCLUSION As expected, we showed that several CNVs were associated with clinical relevance, especially those located on CDKN2A, PLG, and ALDOB, in a homogeneous cohort of patients with clear cell metastatic RCC.
Collapse
|
20
|
Thiesen HJ, Steinbeck F, Maruschke M, Koczan D, Ziems B, Hakenberg OW. Stratification of clear cell renal cell carcinoma (ccRCC) genomes by gene-directed copy number alteration (CNA) analysis. PLoS One 2017; 12:e0176659. [PMID: 28486536 PMCID: PMC5423597 DOI: 10.1371/journal.pone.0176659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/14/2017] [Indexed: 11/18/2022] Open
Abstract
Tumorigenic processes are understood to be driven by epi-/genetic and genomic alterations from single point mutations to chromosomal alterations such as insertions and deletions of nucleotides up to gains and losses of large chromosomal fragments including products of chromosomal rearrangements e.g. fusion genes and proteins. Overall comparisons of copy number alterations (CNAs) presented in 48 clear cell renal cell carcinoma (ccRCC) genomes resulted in ratios of gene losses versus gene gains between 26 ccRCC Fuhrman malignancy grades G1 (ratio 1.25) and 20 G3 (ratio 0.58). Gene losses and gains of 15762 CNA genes were mapped to 795 chromosomal cytoband loci including 280 KEGG pathways. CNAs were classified according to their contribution to Fuhrman tumour gradings G1 and G3. Gene gains and losses turned out to be highly structured processes in ccRCC genomes enabling the subclassification and stratification of ccRCC tumours in a genome-wide manner. CNAs of ccRCC seem to start with common tumour related gene losses flanked by CNAs specifying Fuhrman grade G1 losses and CNA gains favouring grade G3 tumours. The appearance of recurrent CNA signatures implies the presence of causal mechanisms most likely implicated in the pathogenesis and disease-outcome of ccRCC tumours distinguishing lower from higher malignant tumours. The diagnostic quality of initial 201 genes (108 genes supporting G1 and 93 genes G3 phenotypes) has been successfully validated on published Swiss data (GSE19949) leading to a restricted CNA gene set of 171 CNA genes of which 85 genes favour Fuhrman grade G1 and 86 genes Fuhrman grade G3. Regarding these gene sets overall survival decreased with the number of G3 related gene losses plus G3 related gene gains. CNA gene sets presented define an entry to a gene-directed and pathway-related functional understanding of ongoing copy number alterations within and between individual ccRCC tumours leading to CNA genes of prognostic and predictive value.
Collapse
Affiliation(s)
- H.-J. Thiesen
- Institute of Immunology, University of Rostock, Rostock, Germany
- * E-mail:
| | - F. Steinbeck
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - M. Maruschke
- Department of Urology, University of Rostock, Rostock, Germany
- Department of Urology, HELIOS Hanseklinikum Stralsund, Germany
| | - D. Koczan
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - B. Ziems
- Steinbeis Center for Proteome Analysis, Rostock, Germany
| | - O. W. Hakenberg
- Department of Urology, University of Rostock, Rostock, Germany
| |
Collapse
|
21
|
Prattichizzo C, Gigante M, Pontrelli P, Stella A, Rocchetti MT, Gigante M, Maiorano E, Herr W, Battaglia M, Gesualdo L, Ranieri E. Establishment and characterization of a highly immunogenic human renal carcinoma cell line. Int J Oncol 2016; 49:457-70. [PMID: 27278998 PMCID: PMC4922831 DOI: 10.3892/ijo.2016.3544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/02/2015] [Indexed: 11/26/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common kidney cancer, and accounts for ~3% of all adult malignancies. RCC has proven refractory to conventional treatment modalities but appears to be the only histological form that shows any consistent response to immunotherapeutic approaches. The development of a clinically effective vaccine remains a major strategic target for devising active specific immunotherapy in RCC. We aimed to identify a highly immunogenic antigenic format for immunotherapeutic approaches, so as to boost immune responses in RCC patients. We established and cloned an immunogenic cell line, RCC85#21 named Elthem, which was derived from a non-aggressive and non-metastatic clear cell carcinoma. The cell line characterization was performed by genomics (real-time PCR, genome instability), proteomics (two dimensional electrophoresis, mass spectro-metry) and immunological analysis (mixed lymphocytes tumor cell cultures). Real-time PCR confirmed the RCC85#21 cell expression of tumor antigens and cytokine genes. No difference in microsatellite instability (MSI) in RCC85#21 cell line was found as compared to control, loss of heterozygosity was observed in the RCC85#21 clone, but not in the renal cancer cell lines from which it was generated. The image analysis of RCC85#21 by two-dimensional gels showed 700±26 spots and 119 spots were identified by mass spectrometry analysis. RCC85#21 promoted a significant RCC-specific T cells activation by exhibiting a cytotoxic phenotype after mixed lymphocyte and tumor cell cultures. CD8+ T cells isolated from RCC patients displayed an elevated reactivity against RCC85#21 and efficiently lysed the RCC85#21 clone. The RCC85#21 immunogenic cell line will be suitable for immune stimulation. The identification of novel tumor associated antigens will allow the evaluation of the immune response in vitro and, subsequently, in vivo paving the way for new immunotherapeutic strategies in the RCC setting.
Collapse
Affiliation(s)
- Clelia Prattichizzo
- Department of Medical and Surgical Sciences, Section of Clinical Pathology, University of Foggia, Foggia, Italy
| | - Margherita Gigante
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Paola Pontrelli
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Alessandro Stella
- Medical Genetics Unit, Department of Biomedicine in Childhood, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Teresa Rocchetti
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maddalena Gigante
- Department of Medical and Surgical Sciences, Section of Clinical Pathology, University of Foggia, Foggia, Italy
| | - Eugenio Maiorano
- Department of Pathological Anatomy, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Wolfgang Herr
- Department of Medicine III, Johannes Gutenberg-University Mainz, Mainz, Germany
| | | | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Section of Nephrology, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Elena Ranieri
- Department of Medical and Surgical Sciences, Section of Clinical Pathology, University of Foggia, Foggia, Italy
| |
Collapse
|
22
|
Thyroid Hormones as Renal Cell Cancer Regulators. JOURNAL OF SIGNAL TRANSDUCTION 2016; 2016:1362407. [PMID: 27034829 PMCID: PMC4808550 DOI: 10.1155/2016/1362407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/23/2016] [Indexed: 12/27/2022]
Abstract
It is known that thyroid hormone is an important regulator of cancer development and metastasis. What is more, changes across the genome, as well as alternative splicing, may affect the activity of the thyroid hormone receptors. Mechanism of action of the thyroid hormone is different in every cancer; therefore in this review thyroid hormone and its receptor are presented as a regulator of renal cell carcinoma.
Collapse
|
23
|
Chen K, Zeng J, Xiao H, Huang C, Hu J, Yao W, Yu G, Xiao W, Xu H, Ye Z. Regulation of glucose metabolism by p62/SQSTM1 through HIF1α. J Cell Sci 2016; 129:817-30. [PMID: 26743088 PMCID: PMC4760374 DOI: 10.1242/jcs.178756] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/23/2015] [Indexed: 12/16/2022] Open
Abstract
The signaling adaptor sequestosome 1 (SQSTM1)/p62 is frequently overexpressed in tumors and plays an important role in the regulation of tumorigenesis. Although great progress has been made, biological roles of p62 and relevant molecular mechanisms responsible for its pro-tumor activity remain largely unknown. Here, we show that p62 knockdown reduces cell growth and the expression of glycolytic genes in a manner that depends on HIF1α activity in renal cancer cells. Knockdown of p62 decreases HIF1α levels and transcriptional activity by regulating mTORC1 activity and NF-κB nuclear translocation. Furthermore, p62 interacts directly with the von Hippel-Lindau (VHL) E3 ligase complex to modulate the stability of HIF1α. Mechanistically, p62 binds to the VHL complex and competes with HIF1α. Expression of p62 inhibits the interaction of DCNL1 (also known as DCUN1D1) with CUL2 and attenuates the neddylation of CUL2, and thus downregulates the VHL E3 ligase complex activity. Functionally, HIF1α expression is required for p62-induced glucose uptake, lactate production and soft agar colony growth. Taken together, our findings demonstrate that p62 is a crucial positive regulator of HIF1α, which is a facilitating factor in p62-enhanced tumorigenesis.
Collapse
Affiliation(s)
- Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jin Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Haibing Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chunhua Huang
- College of Basic Medicine Science, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Junhui Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Gan Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Wei Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
24
|
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: 1.3] [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
|
25
|
Ito T, Pei J, Dulaimi E, Menges C, Abbosh PH, Smaldone MC, Chen DYT, Greenberg RE, Kutikov A, Viterbo R, Uzzo RG, Testa JR. Genomic Copy Number Alterations in Renal Cell Carcinoma with Sarcomatoid Features. J Urol 2015; 195:852-8. [PMID: 26602888 DOI: 10.1016/j.juro.2015.10.180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2015] [Indexed: 01/31/2023]
Abstract
PURPOSE Sarcomatoid changes in renal cell carcinoma are associated with a poor prognosis. The identification of genetic alterations that drive this aggressive phenotype could aid in the development of more effective targeted therapies. In this study we aimed to pinpoint unique copy number alterations in sarcomatoid renal cell carcinoma compared to classical renal cell carcinoma subtypes. MATERIALS AND METHODS Genomic copy number analysis was performed using single nucleotide polymorphism based microarrays on tissue extracted from the tumors of 81 patients who underwent renal mass excision, including 17 with sarcomatoid renal cell carcinoma. RESULTS Sarcomatoid renal cell carcinoma showed a significantly higher number of copy number alterations than clear cell, papillary and chromophobe renal cell carcinoma (mean 18.0 vs 5.8, 6.5 and 7.2, respectively, p <0.0001). Copy number losses of chromosome arms 9q, 15q, 18p/q and 22q, and gains of 1q and 8q occurred in a significantly higher proportion of sarcomatoid renal cell carcinomas than in the other 3 histologies. Patients with sarcomatoid renal cell carcinoma demonstrated significantly worse overall survival compared to those without that condition on Kaplan-Meier analysis (p = 0.0001). Patients with 9 or more copy number alterations also demonstrated significantly worse overall survival than those with fewer than 9 copy number alterations (p = 0.004). CONCLUSIONS Sarcomatoid changes in renal cell carcinoma are associated with a high rate of chromosomal imbalances with losses of 9q, 15q, 18p/q and 22q, and gains of 1q and 8q occurring at significantly higher frequencies in comparison to nonsarcomatoid renal cell carcinoma. Identifying candidate driver genes or tumor suppressor loci in these chromosomal regions may help identify targets for future therapies.
Collapse
Affiliation(s)
- Timothy Ito
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jianming Pei
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Essel Dulaimi
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Craig Menges
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Philip H Abbosh
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Marc C Smaldone
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - David Y T Chen
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Richard E Greenberg
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Alexander Kutikov
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Rosalia Viterbo
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Robert G Uzzo
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Joseph R Testa
- Cancer Biology Program and Blood Cell Development and Function Program (ED), Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
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
|
27
|
Zhao SC, Zhou BW, Luo F, Mao X, Lu YJ. The structure and function of NKAIN2-a candidate tumor suppressor. Int J Clin Exp Med 2015; 8:17072-17079. [PMID: 26770299 PMCID: PMC4694199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 10/10/2015] [Indexed: 06/05/2023]
Abstract
The deletion of chromosomal region 6q was commonly found in several types of human cancers, although the tumor suppressor genes (TSGs) located within this genomic region are not well established. Our recent work detected recurrent chromosomal truncation at the Na(+)/K(+) transporting ATPase interacting 2 (NKAIN2) gene in prostate cancer, which was also found to be truncated in leukemia and lymphoma, suggesting that NKAIN2 is potentially one of the TSGs located in the 6q commonly deleted region in human cancers. NKAIN2 gene consists of eight coding exons that span approximately 1 Mb of genomic DNA on chromosome 6q and there are four main splice variants. The function of this gene is not well investigated and the limited knowledge of this gene pointed to nervous system development. The chromosomal translocations in nervous development disorders usually lead to inactivation of this gene. In human tumors, both chromosomal deletion and translocation may also inactivate this gene and consequently contribute to tumorigenesis. Further genetic and cellular functional studies are required to establish its tumor suppressor role.
Collapse
Affiliation(s)
- Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical UniversityGuangzhou, PR China
| | - Bo-Wei Zhou
- Department of Urology, Nanfang Hospital, Southern Medical UniversityGuangzhou, PR China
| | - Fei Luo
- Department of Urology, Nanfang Hospital, Southern Medical UniversityGuangzhou, PR China
| | - Xueying Mao
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondon, UK
| | - Yong-Jie Lu
- Molecular Oncology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondon, UK
| |
Collapse
|
28
|
Kang XL, Zou H, Pang LJ, Hu WH, Zhao J, Qi Y, Liu CX, Hu JM, Tang JX, LI HA, Liang WH, Yuan XL, Li F. Chromosomal imbalances revealed in primary renal cell carcinomas by comparative genomic hybridization. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3636-47. [PMID: 26097545 PMCID: PMC4466932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
Renal cell carcinoma (RCC) accounts for approximately 3% of all new cancer cases. Although the classification of RCC is based mainly on histology, this method is not always accurate. We applied comparative genomic hybridization (CGH) to determine genomic alterations in 46 cases of different RCC histological subtypes [10 cases of clear cell RCC (CCRCC), 13 cases of papillary RCC (PRCC), 12 cases of chromophobe RCC (CRCC), 9 cases of Xp11.2 translocation RCC (Xp11.2RCC), 2 cases of undifferentiated RCC (unRCC)], and investigated the relationships between clinical parameters and genomic aberrations. Changes involving one or more regions of the genome were seen in all RCC patients; DNA sequence gains were most frequently (>30%) seen in chromosomes 7q, 16p, and 20q; losses from 1p, 3p, 13q, 14q, and 8p. We conclude CGH is a useful complementary method for differential diagnosis of RCC. Loss of 3p21-25, 15q, and gain of 16p11-13 are relatively particular to CCRCC vs. other types of RCC. Gain of 7p13-22, 8q21-24, and loss of 18q12-ter, 14q13-24, and Xp11-q13/Y are more apparent in PRCC, and gain of 8q21-24 is characteristic of type 2 PRCC vs. type 1 PRCC. Loss of 2q12-32, 10p12-15, and 11p11-15, 13p are characteristic of CRCC, and gain of 3p and loss of 11p11-15 and 13p are significant differentiators between common CRCC and CRCC accompanied by sarcomatous change groups. Gain of Xp11-12 is characteristic of the Xp11.2RCC group. Based on Multivariate Cox regression analysis, aberration in 5 chromosome regions were poor prognostic markers of RCC, and include the gain of chromosome 12p12-ter (P = 0.034, RR = 3.502, 95% CI 1.097-11.182), 12q14-ter (P = 0.002, RR = 5.115, 95% CI 1.847-14.170), 16q21-24 (P = 0.044, RR = 2.629, 95% CI 1.027-6.731), 17p12-ter (P = 0.017, RR = 3.643, 95% CI 1.262-10.512) and the loss of 18q12-23 (P = 0.049, RR = 2.911, 95% CI 1.006-8.425), which may provide clues of new genes involved in RCC tumorigenesis.
Collapse
Affiliation(s)
- Xue-Ling Kang
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
- Department of Pathology and Pathophysiology, Fudan University School of MedicineShanghai, China
| | - Hong Zou
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
- Tongji Hospital Cancer Center, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Li Juan Pang
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Wen Hao Hu
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Jin Zhao
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Yan Qi
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Chun-Xia Liu
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Jian Ming Hu
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Jing-Xia Tang
- Department of Preventive Medicine, Medical College of Shihezi UniversityShihezi, Xinjiang, China
| | - Hong An LI
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Wei Hua Liang
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
| | - Xiang-Lin Yuan
- Tongji Hospital Cancer Center, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education of ChinaShihezi, Xinjiang, China
- Tongji Hospital Cancer Center, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, Hubei, China
| |
Collapse
|
29
|
Shu X, Purdue MP, Ye Y, Wood CG, Chen M, Wang Z, Albanes D, Pu X, Huang M, Stevens VL, Diver WR, Gapstur SM, Virtamo J, Chow WH, Tannir NM, Dinney CP, Rothman N, Chanock SJ, Wu X. Multilevel-analysis identify a cis-expression quantitative trait locus associated with risk of renal cell carcinoma. Oncotarget 2015; 6:4097-109. [PMID: 25784652 PMCID: PMC4414175 DOI: 10.18632/oncotarget.3001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/21/2014] [Indexed: 01/20/2023] Open
Abstract
We conducted multilevel analyses to identify potential susceptibility loci for renal cell carcinoma (RCC), which may be overlooked in traditional genome-wide association studies (GWAS). A gene set enrichment analysis was performed utilizing a GWAS dataset comprised of 894 RCC cases and 1,516 controls using GenGen, SNP ratio test, and ALIGATOR. The antigen processing and presentation pathway was consistently significant (P = 0.001, = 0.004, and < 0.001, respectively). Versatile gene-based association study approach was applied to the top-ranked pathway and identified the driven genes. By comparing the expression of the genes in RCC tumor and adjacent normal tissues, we observed significant overexpression of HLA genes in tumor tissues, which was also supported by public databases. We sought to validate genetic variants in antigen processing and presentation pathway in an independent GWAS dataset comprised of 1,311 RCC cases and 3,424 control subjects from the National Cancer Institute; one SNP, rs1063355, was significant in both populations (Pmeta-analysis = 9.15 × 10−4, Pheterogeneity = 0.427). Strong correlation indicated that rs1063355 was a cis-expression quantitative trait loci which associated with HLA-DQB1 expression (Spearman's rank r = −0.59, p = 5.61 × 10−6). The correlation was further validated using a public dataset. Our results highlighted the role of immune-related pathway and genes in the etiology of RCC.
Collapse
Affiliation(s)
- Xiang Shu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher G Wood
- Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Meng Chen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhaoming Wang
- Cancer Genomics Research Laboratory, SAIC-Frederick Inc., National Cancer Institute-Frederick, Frederick, Maryland, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Xia Pu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maosheng Huang
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA
| | - W Ryan Diver
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Wong-Ho Chow
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nizar M Tannir
- Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Colin P Dinney
- Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Marquez SB, Thompson KW, Lu L, Reisman D. Beyond Mutations: Additional Mechanisms and Implications of SWI/SNF Complex Inactivation. Front Oncol 2015; 4:372. [PMID: 25774356 PMCID: PMC4343012 DOI: 10.3389/fonc.2014.00372] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/11/2014] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED SWI/SNF is a major regulator of gene expression. Its role is to facilitate the shifting and exposure of DNA segments within the promoter and other key domains to transcription factors and other essential cellular proteins. This complex interacts with a wide range of proteins and does not function within a single, specific pathway; thus, it is involved in a multitude of cellular processes, including DNA repair, differentiation, development, cell adhesion, and growth control. Given SWI/SNF's prominent role in these processes, many of which are important for blocking cancer development, it is not surprising that the SWI/SNF complex is targeted during cancer initiation and progression both by mutations and by non-mutational mechanisms. Currently, the understanding of the types of alterations, their frequency, and their impact on the SWI/SNF subunits is an area of intense research that has been bolstered by a recent cadre of NextGen sequencing studies. These studies have revealed mutations in SWI/SNF subunits, indicating that this complex is thus important for cancer development. The purpose of this review is to put into perspective the role of mutations versus other mechanisms in the silencing of SWI/SNF subunits, in particular, BRG1 and BRM. In addition, this review explores the recent development of synthetic lethality and how it applies to this complex, as well as how BRM polymorphisms are becoming recognized as potential clinical biomarkers for cancer risk. SIGNIFICANCE Recent reviews have detailed the occurrence of mutations in nearly all SWI/SNF subunits, which indicates that this complex is an important target for cancer. However, when the frequency of mutations in a given tumor type is compared to the frequency of subunit loss, it becomes clear that other non-mutational mechanisms must play a role in the inactivation of SWI/SNF subunits. Such data indicate that epigenetic mechanisms that are known to regulate BRM may also be involved in the loss of expression of other SWI/SNF subunits. This is important since epigenetically silenced genes are inducible, and thus, the reversal of the silencing of these non-mutationally suppressed subunits may be a viable mode of targeted therapy.
Collapse
Affiliation(s)
- Stefanie B Marquez
- Department of Medicine, Division of Hematology/Oncology, University of Florida , Gainesville, FL , USA
| | - Kenneth W Thompson
- Department of Medicine, Division of Hematology/Oncology, University of Florida , Gainesville, FL , USA
| | - Li Lu
- Department of Pathology, University of Florida , Gainesville, FL , USA
| | - David Reisman
- Department of Medicine, Division of Hematology/Oncology, University of Florida , Gainesville, FL , USA
| |
Collapse
|
31
|
Czarnecka AM, Kornakiewicz A, Kukwa W, Szczylik C. Frontiers in clinical and molecular diagnostics and staging of metastatic clear cell renal cell carcinoma. Future Oncol 2015; 10:1095-111. [PMID: 24941992 DOI: 10.2217/fon.13.258] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The last few years have brought advances in the understanding of the molecular biology of metastatic clear cell renal cell carcinoma (RCC). Both preclinical research and clinical trials brought together results from the latest advancements in RCC diagnostic and staging. Understanding of the complex molecular alterations involved in the development and progression of RCC enables development of immunohistochemical and genetic diagnostic tools and is also opening the doors for experimental targeted therapies. At the same time, improvements of medical and molecular imaging improves the sensitivity and specificity of metastatic disease diagnosis. Moreover, independent validation of molecular profiles across high-throughput platforms, methods, laboratories and cancer populations has recently been successfully performed in RCC. Generation of informative, clinical diagnostic tools is likely to contribute to development of novel personalized diagnostic and treatment protocols and ensure prolonged survival of RCC patient in the near future.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141 Warsaw, Poland
| | | | | | | |
Collapse
|
32
|
|
33
|
Alexiev BA, Zou YS. Clear cell papillary renal cell carcinoma: A chromosomal microarray analysis of two cases using a novel Molecular Inversion Probe (MIP) technology. Pathol Res Pract 2014; 210:1049-53. [DOI: 10.1016/j.prp.2014.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 09/29/2014] [Accepted: 10/13/2014] [Indexed: 10/24/2022]
|
34
|
Huang G, Stock C, Bommeljé CC, Weeda VB, Shah K, Bains S, Buss E, Shaha M, Rechler W, Ramanathan SY, Singh B. SCCRO3 (DCUN1D3) antagonizes the neddylation and oncogenic activity of SCCRO (DCUN1D1). J Biol Chem 2014; 289:34728-42. [PMID: 25349211 DOI: 10.1074/jbc.m114.585505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The activity of cullin-RING type ubiquitination E3 ligases is regulated by neddylation, a process analogous to ubiquitination that culminates in covalent attachment of the ubiquitin-like protein Nedd8 to cullins. As a component of the E3 for neddylation, SCCRO/DCUN1D1 plays a key regulatory role in neddylation and, consequently, cullin-RING ligase activity. The essential contribution of SCCRO to neddylation is to promote nuclear translocation of the cullin-ROC1 complex. The presence of a myristoyl sequence in SCCRO3, one of four SCCRO paralogues present in humans that localizes to the membrane, raises questions about its function in neddylation. We found that although SCCRO3 binds to CAND1, cullins, and ROC1, it does not efficiently bind to Ubc12, promote cullin neddylation, or conform to the reaction processivity paradigms, suggesting that SCCRO3 does not have E3 activity. Expression of SCCRO3 inhibits SCCRO-promoted neddylation by sequestering cullins to the membrane, thereby blocking its nuclear translocation. Moreover, SCCRO3 inhibits SCCRO transforming activity. The inhibitory effects of SCCRO3 on SCCRO-promoted neddylation and transformation require both an intact myristoyl sequence and PONY domain, confirming that membrane localization and binding to cullins are required for in vivo functions. Taken together, our findings suggest that SCCRO3 functions as a tumor suppressor by antagonizing the neddylation activity of SCCRO.
Collapse
Affiliation(s)
- Guochang Huang
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Cameron Stock
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Claire C Bommeljé
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Víola B Weeda
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Kushyup Shah
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Sarina Bains
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Elizabeth Buss
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Manish Shaha
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Willi Rechler
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Suresh Y Ramanathan
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Bhuvanesh Singh
- From the Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
35
|
Gowrishankar B, Cahill L, Arndt AE, Al-Ahmadie H, Lin O, Chadalavada K, Chaganti S, Nanjangud GJ, Murty VV, Chaganti RSK, Reuter VE, Houldsworth J. Subtyping of renal cortical neoplasms in fine needle aspiration biopsies using a decision tree based on genomic alterations detected by fluorescence in situ hybridization. BJU Int 2014; 114:881-90. [PMID: 24467611 PMCID: PMC4257075 DOI: 10.1111/bju.12643] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To improve the overall accuracy of diagnosis in needle biopsies of renal masses, especially small renal masses (SRMs), using fluorescence in situ hybridization (FISH), and to develop a renal cortical neoplasm classification decision tree based on genomic alterations detected by FISH. PATIENTS AND METHODS Ex vivo fine needle aspiration biopsies of 122 resected renal cortical neoplasms were subjected to FISH using a series of seven-probe sets to assess gain or loss of 10 chromosomes and rearrangement of the 11q13 locus. Using specimen (nephrectomy)-histology as the 'gold standard', a genomic aberration-based decision tree was generated to classify specimens. The diagnostic potential of the decision tree was assessed by comparing the FISH-based classification and biopsy histology with specimen histology. RESULTS Of the 114 biopsies diagnostic by either method, a higher diagnostic yield was achieved by FISH (92 and 96%) than histology alone (82 and 84%) in the 65 biopsies from SRMs (<4 cm) and 49 from larger masses, respectively. An optimized decision tree was constructed based on aberrations detected in eight chromosomes, by which the maximum concordance of classification achieved by FISH was 79%, irrespective of mass size. In SRMs, the overall sensitivity of diagnosis by FISH compared with histopathology was higher for benign oncocytoma, was similar for the chromophobe renal cell carcinoma subtype, and was lower for clear-cell and papillary subtypes. The diagnostic accuracy of classification of needle biopsy specimens (from SRMs) increased from 80% obtained by histology alone to 94% when combining histology and FISH. CONCLUSION The present study suggests that a novel FISH assay developed by us has a role to play in assisting in the yield and accuracy of diagnosis of renal cortical neoplasms in needle biopsies in particular, and can help guide the clinical management of patients with SRMs that were non-diagnostic by histology.
Collapse
|
36
|
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: 0.8] [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
|
37
|
Abstract
Renal cell carcinoma of clear-cell type (ccRCC) is an enigmatic tumor type, characterized by frequent inactivation of the VHL gene (infrequently mutated in other tumor types), responsiveness to angiogenesis inhibitors, and resistance to both chemotherapy and conventional radiation therapy. ccRCC tumors exhibit substantial mutation heterogeneity. Recent studies using massively parallel sequencing technologies have implicated several novel driver genes. In VHL wild-type tumors, mutations were discovered in TCEB1, which encodes Elongin C, a protein that binds to VHL and is required for its function. Several additional tumor suppressor genes have been identified near the VHL gene, within a region that is frequently deleted in ccRCC on chromosome 3p: SETD2, BAP1, and PBRM1. Mutations in BAP1 and PBRM1 are largely mutually exclusive and are associated with different tumor biology and patient outcomes. In addition, the mTORC1 pathway is deregulated by mutations in MTOR, TSC1, PIK3CA, and PTEN in approximately 20% of ccRCCs. Mutations in TSC1, and possibly other genes, may predict for sensitivity to mTORC1 inhibitors. These discoveries provide insight into ccRCC development and set the foundation for the first molecular genetic classification of the disease, paving the way for subtype-specific therapies.
Collapse
Affiliation(s)
- James Brugarolas
- From the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
38
|
Abstract
Telomeres are ribonucleoprotein structures capping the end of every linear chromosome. In all vertebrates, they are composed of TTAGGG repeats coated with specific protecting proteins. Telomeres shorten with each mitotic cell division, but telomerase, a reverse transcriptase, elongate telomeres in very specific cells, such as embryonic and adult stem cells. Although telomere sequence is identical in mice and humans and telomeres serve the same role of protecting chromosomes and genetic information from damage and erosion in both species, abnormalities in telomere maintenance and in telomerase function do not coincide in phenotype in humans and mice. The telomeres of most laboratory mice are 5 to 10 times longer than in humans, but their lifespan is 30 times shorter. Complete absence of telomerase has little expression in phenotype over several generations in mice, whereas heterozygosity for telomerase mutations in humans is sufficient to result in organ regeneration defect and cancer development. Patients with telomerase deficiency and very short telomeres may develop aplastic anemia, pulmonary fibrosis, or cirrhosis, whereas telomerase-null murine models display only modest hematopoietic deficiency and develop emphysema when exposed to cigarette smoke. In summary, telomerase deficiency in both humans and mice accelerate telomere shortening, but its consequences in the different organs and in the organism diverge, mainly due to telomere length differences.
Collapse
|
39
|
Caldwell IR, Oei P, Ng D, Caudwell B, Fong PCC, Broom RJ. Analysis of molecular cytogenetic changes in metastatic renal cell carcinoma in the setting of everolimus treatment: a pilot project. Clin Genitourin Cancer 2014; 12:256-61. [PMID: 24524969 DOI: 10.1016/j.clgc.2014.01.002] [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] [Received: 10/26/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND The mTOR inhibitors have improved outcomes for patients with metastatic renal cell carcinoma (mRCC) but the duration of benefit is variable. Currently there are no predictive biomarkers for preselecting patients who are more likely to benefit from these agents. We undertook an exploratory translational study evaluating molecular cytogenetic changes in the context of outcomes from treatment with everolimus. PATIENTS AND METHODS Ten patients with clear cell mRCC treated with everolimus were enrolled. Pretreatment tissue specimens were analyzed for molecular cytogenetic changes using fluorescence in situ hybridization and progression-free survival (PFS) data were obtained. Gene probes chosen for this analysis were: Von Hippel Lindau, fragile histidine triad, fibroblast growth factor receptor (FGFR) 1, FGFR3, PDGFβ, PDGFRβ, epidermal growth factor receptor, and myelocytomatosis viral oncogene. RESULTS Median PFS was 8.75 months. Two patients with the longest PFS (28 months and 23 months) had gain of PDGFβ and PDGFRβ. This was also observed in 3 other patients who had a PFS of 11.5 months, 8 months, and 5.5 months, respectively. Cytogenetic evolution was observed between primary and metastatic specimens. CONCLUSION PDGFβ and PDGFRβ gene status might be of relevance to everolimus therapy. Further research evaluating the utility of these potential biomarkers is required.
Collapse
Affiliation(s)
- Imogen R Caldwell
- Adult Oncology Research Centre, Department of Medical Oncology, Auckland City Hospital, Auckland, New Zealand
| | - Paul Oei
- IGENZ Ltd, Auckland, New Zealand
| | | | - Beth Caudwell
- Adult Oncology Research Centre, Department of Medical Oncology, Auckland City Hospital, Auckland, New Zealand
| | - Peter C C Fong
- Adult Oncology Research Centre, Department of Medical Oncology, Auckland City Hospital, Auckland, New Zealand
| | - Reuben J Broom
- Adult Oncology Research Centre, Department of Medical Oncology, Auckland City Hospital, Auckland, New Zealand.
| |
Collapse
|
40
|
Li L, Shen C, Nakamura E, Ando K, Signoretti S, Beroukhim R, Cowley GS, Lizotte P, Liberzon E, Bair S, Root DE, Tamayo P, Tsherniak A, Cheng SC, Tabak B, Jacobsen A, Hakimi AA, Schultz N, Ciriello G, Sander C, Hsieh JJ, Kaelin WG. SQSTM1 is a pathogenic target of 5q copy number gains in kidney cancer. Cancer Cell 2013; 24:738-50. [PMID: 24332042 PMCID: PMC3910168 DOI: 10.1016/j.ccr.2013.10.025] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 09/02/2013] [Accepted: 10/31/2013] [Indexed: 12/22/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer and is often linked to loss of chromosome 3p, which harbors the VHL tumor suppressor gene, loss of chromosome 14q, which includes HIF1A, and gain of chromosome 5q. The relevant target(s) on chromosome 5q is not known. Here, we show that 5q amplification leads to overexpression of the SQSTM1 oncogene in ccRCC lines and tumors. Overexpression of SQSTM1 in ccRCC lines promoted resistance to redox stress and increased soft agar growth, while downregulation of SQSTM1 decreased resistance to redox stress, impaired cellular fitness, and decreased tumor formation. Therefore, the selection pressure to amplify 5q in ccRCC is driven, at least partly, by SQSTM1.
Collapse
Affiliation(s)
- Lianjie Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chuan Shen
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eijiro Nakamura
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kiyohiro Ando
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Patrick Lizotte
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Ella Liberzon
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Steven Bair
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David E Root
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Pablo Tamayo
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Aviad Tsherniak
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Su-Chun Cheng
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Barbara Tabak
- Department of Cancer Biology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Anders Jacobsen
- Department of Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - A Ari Hakimi
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - Nikolaus Schultz
- Department of Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - Giovanni Ciriello
- Department of Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - Chris Sander
- Department of Computational Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - James J Hsieh
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA; Department of Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 16605, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
41
|
White NMA, Newsted DW, Masui O, Romaschin AD, Siu KWM, Yousef GM. Identification and validation of dysregulated metabolic pathways in metastatic renal cell carcinoma. Tumour Biol 2013; 35:1833-46. [PMID: 24136743 DOI: 10.1007/s13277-013-1245-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 09/23/2013] [Indexed: 01/03/2023] Open
Abstract
Metastatic renal cell carcinoma (mRCC) is a devastating disease with a 5-year survival rate of approximately 9 % and low response to chemotherapy and radiotherapy. Targeted therapies have slightly improved patient survival, but are only effective in a small subset of patients, who eventually develop resistance. A better understanding of pathways contributing to tumor progression and metastasis will allow for the development of novel targeted therapies and accurate prognostic markers. We performed extensive bioinformatics coupled with experimental validation on proteins dysregulated in mRCC. Gene ontology analysis showed that many proteins are involved in oxidation reduction, metabolic processes, and signal transduction. Pathway analysis showed metabolic pathways are altered in mRCC including glycolysis and pyruvate metabolism, the citric acid cycle, and the pentose phosphate pathway. RT-qPCR analysis showed that genes involved in the citric acid cycle were downregulated in metastatic RCC while genes of the pentose phosphate pathway were overexpressed. Protein-protein interaction analysis showed that most of the 198 proteins altered in mRCC clustered together and many were involved in glycolysis and pyruvate metabolism. We identified 29 reported regions of chromosomal aberrations in metastatic disease that correlate with the direction of protein dysregulation in mRCC. Furthermore, 36 proteins dysregulated in mRCC are predicted to be targets of metastasis-related miRNAs. A more comprehensive understanding of the pathways dysregulated in metastasis can be useful for the development of new therapies and novel prognostic markers. Also, multileveled analyses provide a unique "snapshot" of the molecular "environment" in RCC with prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Nicole M A White
- Department of Laboratory Medicine and the Keenan Research Centre, Li Ka Shing Knowledge Institute of St. Michael's Hospital, 30 Bond Street, Toronto, M5B 1W8, Canada
| | | | | | | | | | | |
Collapse
|
42
|
Genomics and epigenomics of clear cell renal cell carcinoma: recent developments and potential applications. Cancer Lett 2013; 341:111-26. [PMID: 23933176 DOI: 10.1016/j.canlet.2013.08.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/12/2013] [Accepted: 08/02/2013] [Indexed: 12/21/2022]
Abstract
Majority of clear cell renal cell carcinomas (ccRCCs) are diagnosed in the advanced metastatic stage resulting in dramatic decrease of patient survival. Thereby, early detection and monitoring of the disease may improve prognosis and treatment results. Recent technological advances enable the identification of genetic events associated with ccRCC and reveal significant molecular heterogeneity of ccRCC tumors. This review summarizes recent findings in ccRCC genomics and epigenomics derived from chromosomal aberrations, DNA sequencing and methylation, mRNA, miRNA expression profiling experiments. We provide a molecular insight into ccRCC pathology and recapitulate possible clinical applications of genomic alterations as predictive and prognostic biomarkers.
Collapse
|
43
|
Alanee S, Shah S, Murali R, Rau-Murthy R, Schrader KA, Offit K. Absence of loss of heterozygosity of BRCA1 in a renal tumor from a BRCA1 germline mutation carrier. Fam Cancer 2013; 12:125-7. [PMID: 23086583 DOI: 10.1007/s10689-012-9572-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BRCA1 functions as a tumor suppressor gene and germline and somatic mutations in this gene have been shown to be associated with many types of cancer. We report the first tumor study of renal cell carcinoma in a carrier of the deleterious BRCA1 mutation-c.68_69delAG.
Collapse
Affiliation(s)
- Shaheen Alanee
- Department of Surgery, Urology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
44
|
Castro-Vega LJ, Jouravleva K, Liu WY, Martinez C, Gestraud P, Hupé P, Servant N, Albaud B, Gentien D, Gad S, Richard S, Bacchetti S, Londoño-Vallejo A. Telomere crisis in kidney epithelial cells promotes the acquisition of a microRNA signature retrieved in aggressive renal cell carcinomas. Carcinogenesis 2013; 34:1173-80. [PMID: 23358853 DOI: 10.1093/carcin/bgt029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Telomere shortening is a major source of chromosome instability (CIN) at early stages during carcinogenesis. However, the mechanisms through which telomere-driven CIN (T-CIN) contributes to the acquisition of tumor phenotypes remain uncharacterized. We discovered that human epithelial kidney cells undergoing T-CIN display massive microRNA (miR) expression changes that are not related to local losses or gains. This widespread miR deregulation encompasses a miR-200-dependent epithelial-to-mesenchymal transition (EMT) that confers to immortalized pre-tumoral cells phenotypic traits of metastatic potential. Remarkably, a miR signature of these cells, comprising a downregulation of miRs with conserved expression in kidney, was retrieved in poorly differentiated aggressive renal cell carcinomas. Our results reveal an unanticipated connection between telomere crisis and the activation of the EMT program that occurs at pre-invasive stages of epithelial cancers, through mechanisms that involve miR deregulation. Thus, this study provides a new rational into how telomere instability contributes to the acquisition of the malignant phenotype.
Collapse
Affiliation(s)
- Luis Jaime Castro-Vega
- UMR3244, Telomeres and Cancer Laboratory, Institut Curie, 26 rue d'Ulm, Paris 75248, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cuadros T, Trilla E, Vilà MR, de Torres I, Vilardell J, Messaoud NB, Salcedo M, Sarró E, López-Hellin J, Blanco A, Mir C, Ramón y Cajal S, Itarte E, Morote J, Meseguer A. Hepatitis A virus cellular receptor 1/kidney injury molecule-1 is a susceptibility gene for clear cell renal cell carcinoma and hepatitis A virus cellular receptor/kidney injury molecule-1 ectodomain shedding a predictive biomarker of tumour progression. Eur J Cancer 2013; 49:2034-47. [PMID: 23352434 DOI: 10.1016/j.ejca.2012.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 12/19/2012] [Accepted: 12/21/2012] [Indexed: 11/29/2022]
Abstract
AIM OF THE STUDY To correlate hepatitis A virus cellular receptor (HAVCR)/kidney injury molecule-1 (KIM-1) expression in clear cell renal cell carcinoma (ccRCC) tumours with patient outcome and study the consequences of HAVCR/KIM-1 ectodomain shedding. METHODS HAVCR/KIM-1 expression in ccRCC, oncocytomes, papillary carcinomas and unaffected tissue counterparts was evaluated. Minimal change disease and pre-clamping normal and ccRCC tissue biopsies were included. Tissue microarrays from 98 ccRCC tumours were analysed. Tumour registry data and patient outcome were retrospectivelly collected. Deletions in HAVCR/KIM-1 ectodomain and lentiviral infection of 786-O cells with HAVCR/KIM-1 mutated constructs to determine their subcellular distribution and invasive capacity were performed. RESULTS HAVCR/KIM-1 was expressed in ccRCC, papillary tumours and in tubule cells of adjacent and distal unaffected counterparts of ccRCC tumours. The latest was not related to ischemic or tumour-related paracrine effects since pre-clamping normal biopsies were positive for HAVCR/KIM-1 and unaffected counterparts of papillary tumours were negative. HAVCR/KIM-1 analyses in patients and the invasive capacity of HAVCR/KIM-1 shedding mutants in cell lines demonstrated that: (i) relative low HAVCR/KIM-1 membrane levels correlate with activated shedding in ccRCC patients and mutant cell lines; (ii) augmented shedding directly correlates with higher invasiveness and tumour malignancy. CONCLUDING STATEMENTS: Constitutive expression of HAVCR/KIM-1 in kidney might constitute a susceptibility trait for ccRCC tumour development. Enhanced HAVCR/KIM-1 ectodomain shedding promotes invasive phenotype in vitro and more aggressive tumours in vivo.
Collapse
|
46
|
Du Y, Su T, Tan X, Li X, Xie J, Wang G, Shen J, Hou J, Cao G. Polymorphism in protein tyrosine phosphatase receptor delta is associated with the risk of clear cell renal cell carcinoma. Gene 2013; 512:64-9. [DOI: 10.1016/j.gene.2012.09.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/28/2012] [Accepted: 09/27/2012] [Indexed: 10/27/2022]
|
47
|
Han J, Cao S, Zhang K, Zhao G, Xin Y, Dong Q, Yan Y, Cui J. Fluorescence in situ hybridization as adjunct to cytology improves the diagnosis and directs estimation of prognosis of malignant pleural effusions. J Cardiothorac Surg 2012; 7:121. [PMID: 23148562 PMCID: PMC3514305 DOI: 10.1186/1749-8090-7-121] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 11/03/2012] [Indexed: 11/18/2022] Open
Abstract
Background The identification of malignant cells in effusions by conventional cytology is hampered by its limited sensitivity and specificity. The aim of this study was to investigate the value of fluorescence in situ hybridization (FISH) as adjuncts to conventional cytologic examination in patients with malignant pleural effusions. Methods We conducted a retrospective cohort study of 93 inpatients with pleural effusions (72 malignant pleural effusions metastatic from 11 different organs and 21 benign) over 23 months. All the patients came from Chinese northeast areas. Aspirated pleural fluid underwent cytologic examination and fluorescence in situ hybridization (FISH) for aneuploidy. We used FISH in single-colour or if appropriate in dual-colour evaluation to detect chromosomal aberrations (chromosomes 7, 11, and 17) in effusion cells as markers of malignancy, to raise the diagnostic yield and identified the efficiency by diagnostic biopsy. Predominant cytogenetic anomalies and patterns of intratumor cytogenetic heterogeneity were brought in relation to overall survival rate. Results Cytology alone confirmed malignant pleural effusions in 45 of 72 patients (sensitivity 63%), whereas FISH alone positively identified 48 of 72 patients (sensitivity 67%). Both tests had high specificity in predicting benign effusions. If cytology and FISH were considered together, they exhibited 88% sensitivity and 94.5% specificity in discriminating benign and malignant effusions. Combined, the two assays were more sensitive than either test alone. Although the positive predictive value of each test was 94.5%, the negative predictive value of cytology and FISH combined was 78%, better than 47% and 44% for FISH and cytology alone, respectively. There was a significantly prolonged survival rate for patients with aneuploidy for chromosome 17. Conclusions FISH in combination with conventional cytology is a highly sensitive and specific diagnostic tool for detecting malignant cells in pleural effusions . The high sensitivity and specificity may be associated with geographic area and race. Simple numeric FISH anomalies may be prognostic.
Collapse
Affiliation(s)
- Jingquan Han
- Department of Thoracic Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Nan gang District, Harbin, Heilongjiang Province 150001, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hao XD, Yang Y, Song X, Zhao XK, Wang LD, He JD, Kong QP, Tang NLS, Zhang YP. Correlation of telomere length shortening with TP53 somatic mutations, polymorphisms and allelic loss in breast tumors and esophageal cancer. Oncol Rep 2012; 29:226-36. [PMID: 23124483 DOI: 10.3892/or.2012.2098] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 09/07/2012] [Indexed: 11/05/2022] Open
Abstract
Genomic instability caused by telomere erosion is an important mechanism of tumorigenesis. p53 plays a key role in cellular senescence and/or apoptosis associated with telomere erosion which positions p53 as a guard against tumorigenesis. The present study was undertaken to investigate the potential interactions between p53 functional mutations, polymorphisms, allelic loss and telomere erosion in 126 breast tumor patients and 68 esophageal cancer patients. Telomere length (TL) was measured by real-time quantitative PCR. Somatic mutations, polymorphisms and allelic loss in the TP53 gene were detected by direct sequencing of both tumor and normal tissue samples. Our results showed that telomeres were significantly shorter in tumors with somatic p53 mutations compared with tumors with wild-type p53 in both breast tumors (P=0.007) and esophageal cancer (P=0.001). Telomeres of patients with minor genotype CC of rs12951053 and GG of rs1042522 were significantly shorter compared to patients with other genotypes of this single nucleotide polymorphism in esophageal cancer tissue. Furthermore, TP53 allelic loss was detected and significantly associated with somatic mutations in both types of tumor tissues. These findings suggest that somatic p53 mutations, rs12951053 genotype CC and rs1042522 genotype GG contribute to erosion of telomeres, and TP53 allelic loss may be one of the representations of chromosomal instability caused by telomere erosion combined with somatic p53 mutations. These results support that the TP53 gene has a strong interaction with TL erosion in tumorigenesis.
Collapse
Affiliation(s)
- Xiao-Dan Hao
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sivanand S, Peña-Llopis S, Zhao H, Kucejova B, Spence P, Pavia-Jimenez A, Yamasaki T, McBride DJ, Gillen J, Wolff NC, Morlock L, Lotan Y, Raj GV, Sagalowsky A, Margulis V, Cadeddu JA, Ross MT, Bentley DR, Kabbani W, Xie XJ, Kapur P, Williams NS, Brugarolas J. A validated tumorgraft model reveals activity of dovitinib against renal cell carcinoma. Sci Transl Med 2012; 4:137ra75. [PMID: 22674553 DOI: 10.1126/scitranslmed.3003643] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most anticancer drugs entering clinical trials fail to achieve approval from the U.S. Food and Drug Administration. Drug development is hampered by the lack of preclinical models with therapeutic predictive value. Herein, we report the development and validation of a tumorgraft model of renal cell carcinoma (RCC) and its application to the evaluation of an experimental drug. Tumor samples from 94 patients were implanted in the kidneys of mice without additives or disaggregation. Tumors from 35 of these patients formed tumorgrafts, and 16 stable lines were established. Samples from metastatic sites engrafted at higher frequency than those from primary tumors, and stable engraftment of primary tumors in mice correlated with decreased patient survival. Tumorgrafts retained the histology, gene expression, DNA copy number alterations, and more than 90% of the protein-coding gene mutations of the corresponding tumors. As determined by the induction of hypercalcemia in tumorgraft-bearing mice, tumorgrafts retained the ability to induce paraneoplastic syndromes. In studies simulating drug exposures in patients, RCC tumorgraft growth was inhibited by sunitinib and sirolimus (the active metabolite of temsirolimus in humans), but not by erlotinib, which was used as a control. Dovitinib, a drug in clinical development, showed greater activity than sunitinib and sirolimus. The routine incorporation of models recapitulating the molecular genetics and drug sensitivities of human tumors into preclinical programs has the potential to improve oncology drug development.
Collapse
Affiliation(s)
- Sharanya Sivanand
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Oh EK, Kim YW, Kim IW, Liu HB, Lee KH, Chun HJ, Park DC, Oh EJ, Lee AW, Bae SM, Ahn WS. Differential DNA copy number aberrations in the progression of cervical lesions to invasive cervical carcinoma. Int J Oncol 2012; 41:2038-46. [PMID: 23023522 DOI: 10.3892/ijo.2012.1644] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/21/2012] [Indexed: 11/06/2022] Open
Abstract
Host genomic alterations in addition to human papillomavirus (HPV) are needed for cervical precursor lesions to progress to invasive cancer because only a small percentage of women infected by the virus develop disease. However, the genomic alterations during the progression of cervical lesions have not been systematically examined. The aim of this study was to identify differential genomic alterations among cervical intraepithelial neoplasia CIN1, CIN2, CIN3 and cervical squamous cell carcinoma (SCC). Genomic alterations were examined for 15 cases each of CIN1, CIN2, CIN3 and SCC by array-based comparative genomic hybridization (array CGH). The chromosomal regions showing significant differential in DNA copy number aberrations (DCNAs) among CIN1, CIN2, CIN3 and SCC were successfully identified by resampling-based t-test. The chromosomal regions of 5q35.3 and 2q14.3 showed significant DCNAs between CIN1 and CIN2, and between CIN2 and CIN3, respectively, while a significant difference in DCNAs between CIN3 and SCC was observed at 1q24.3, 3p14.1, 3p14.2, 5q13.2, 7p15.3, 7q22.1 and 13q32.3. In addition, the status of DCNAs in 1q43, 2p11.2, 6p11.2, 7p21.1, 7p14.3, 10q24.1, 13q22.3, 13q34 and 16p13.3 was conserved throughout the progression of CIN to SCC. The presence of differential and common DCNAs among CIN1, CIN2, CIN3 and SCC supports that the CIN progression may include continual clonal selection and evolution. This approach also identified 34 probe sets consistently overexpressed when amplified, suggesting an unbiased identification of candidate genes in SCC during cervical cancer progression.
Collapse
Affiliation(s)
- Eun Kyeong Oh
- Department of Obstetrics and Gynecology, The Catholic University of Korea, Seocho-ku, Seoul 137-040, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|