451
|
Turajlic S, Litchfield K, Xu H, Rosenthal R, McGranahan N, Reading JL, Wong YNS, Rowan A, Kanu N, Al Bakir M, Chambers T, Salgado R, Savas P, Loi S, Birkbak NJ, Sansregret L, Gore M, Larkin J, Quezada SA, Swanton C. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol 2017; 18:1009-1021. [PMID: 28694034 DOI: 10.1016/s1470-2045(17)30516-8] [Citation(s) in RCA: 653] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND The focus of tumour-specific antigen analyses has been on single nucleotide variants (SNVs), with the contribution of small insertions and deletions (indels) less well characterised. We investigated whether the frameshift nature of indel mutations, which create novel open reading frames and a large quantity of mutagenic peptides highly distinct from self, might contribute to the immunogenic phenotype. METHODS We analysed whole-exome sequencing data from 5777 solid tumours, spanning 19 cancer types from The Cancer Genome Atlas. We compared the proportion and number of indels across the cohort, with a subset of results replicated in two independent datasets. We assessed in-silico tumour-specific neoantigen predictions by mutation type with pan-cancer analysis, together with RNAseq profiling in renal clear cell carcinoma cases (n=392), to compare immune gene expression across patient subgroups. Associations between indel burden and treatment response were assessed across four checkpoint inhibitor datasets. FINDINGS We observed renal cell carcinomas to have the highest proportion (0·12) and number of indel mutations across the pan-cancer cohort (p<2·2 × 10-16), more than double the median proportion of indel mutations in all other cancer types examined. Analysis of tumour-specific neoantigens showed that enrichment of indel mutations for high-affinity binders was three times that of non-synonymous SNV mutations. Furthermore, neoantigens derived from indel mutations were nine times enriched for mutant specific binding, as compared with non-synonymous SNV derived neoantigens. Immune gene expression analysis in the renal clear cell carcinoma cohort showed that the presence of mutant-specific neoantigens was associated with upregulation of antigen presentation genes, which correlated (r=0·78) with T-cell activation as measured by CD8-positive expression. Finally, analysis of checkpoint inhibitor response data revealed frameshift indel count to be significantly associated with checkpoint inhibitor response across three separate melanoma cohorts (p=4·7 × 10-4). INTERPRETATION Renal cell carcinomas have the highest pan-cancer proportion and number of indel mutations. Evidence suggests indels are a highly immunogenic mutational class, which can trigger an increased abundance of neoantigens and greater mutant-binding specificity. FUNDING Cancer Research UK, UK National Institute for Health Research (NIHR) at the Royal Marsden Hospital National Health Service Foundation Trust, Institute of Cancer Research and University College London Hospitals Biomedical Research Centres, the UK Medical Research Council, the Rosetrees Trust, Novo Nordisk Foundation, the Prostate Cancer Foundation, the Breast Cancer Research Foundation, the European Research Council.
Collapse
Affiliation(s)
- Samra Turajlic
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - Kevin Litchfield
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Hang Xu
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Rachel Rosenthal
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - Nicholas McGranahan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - James L Reading
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Yien Ning S Wong
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Andrew Rowan
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK
| | - Maise Al Bakir
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Tim Chambers
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Roberto Salgado
- Department of Pathology, Gasthuiszusters, Antwerp, Belgium; Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Savas
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Sherene Loi
- Division of Research and Cancer Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Nicolai J Birkbak
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Laurent Sansregret
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Martin Gore
- Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - James Larkin
- Renal and Skin Units, The Royal Marsden Hospital National Health Service Foundation Trust, London, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, London, UK
| | - Charles Swanton
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
452
|
Keap1/Nrf2 pathway in kidney cancer: frequent methylation of KEAP1 gene promoter in clear renal cell carcinoma. Oncotarget 2017; 8:11187-11198. [PMID: 28061437 PMCID: PMC5355256 DOI: 10.18632/oncotarget.14492] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/27/2016] [Indexed: 12/21/2022] Open
Abstract
The Keap1/Nrf2 pathway is a master regulator of the cellular redox state through the induction of several antioxidant defence genes implicated in chemotherapeutic drugs resistance of tumor cells. An increasing body of evidence supports a key role for Keap1/Nrf2 pathway in kidney diseases and renal cell carcinoma (RCC), but data concerning the molecular basis and the clinical effect of its deregulation remain incomplete. Here we present a molecular profiling of the KEAP1 and NFE2L2 genes in five different Renal Cell Carcinoma histotypes by analysing 89 tumor/normal paired tissues (clear cell Renal Carcinoma, ccRCCs; Oncocytomas; Papillary Renal Cell Carcinoma Type 1, PRCC1; Papillary Renal Cell Carcinoma Type 2, PRCC2; and Chromophobe Cell Carcinoma). A tumor-specific DNA methylation of the KEAP1 gene promoter region was found as a specific feature of the ccRCC subtype (18/37, 48.6%) and a direct correlation with mRNA levels was confirmed by in vitro 5-azacytidine treatment. Analysis of an independent data set of 481 ccRCC and 265 PRCC tumors corroborates our results and multivariate analysis reveals a significant correlation among ccRCCs epigenetic KEAP1 silencing and staging, grading and overall survival. Our molecular results show for the the first time the epigenetic silencing of KEAP1 promoter as the leading mechanism for modulation of KEAP1 expression in ccRCCs and corroborate the driver role of Keap1/Nrf2 axis deregulation with potential new function as independent epigenetic prognostic marker in renal cell carcinoma.
Collapse
|
453
|
Yip SM, Ruiz Morales JM, Donskov F, Fraccon A, Basso U, Rini BI, Lee JL, Bjarnason GA, Sim HW, Beuselinck B, Kanesvaran R, Brugarolas J, Koutsoukos K, Fu SYF, Yuasa T, Davis I, Alva A, Kollmannsberger C, Choueiri TK, Heng DYC. Outcomes of Metastatic Chromophobe Renal Cell Carcinoma (chrRCC) in the Targeted Therapy Era: Results from the International Metastatic Renal Cell Cancer Database Consortium (IMDC). KIDNEY CANCER 2017; 1:41-47. [PMID: 30334003 PMCID: PMC6179119 DOI: 10.3233/kca-160002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Treatment outcomes are poorly characterized in patients with metastatic chromophobe renal cell cancer (chrRCC), a subtype of renal cell carcinoma. Objective: This retrospective series aims to determine metastatic chrRCC treatment outcomes in the targeted therapy era. Methods: A retrospective data analysis was performed using the IMDC dataset of 4970 patients to determine metastatic chrRCC treatment outcomes in the targeted therapy era. Results: 109/4970 (2.2%) patients had metastatic chrRCC out of all patients with mRCC treated with targeted therapy. These patients were compared with 4861/4970 (97.8%) clear cell mRCC (ccRCC) patients. Patients with metastatic chrRCC had a similar OS compared to patients with ccRCC (23.8 months (95% CI 16.7 – 28.1) vs 22.4 months (95% CI 21.4 – 23.4), respectively (p = 0.0908). Patients with IMDC favorable (18%), intermediate (59%) and poor risk (23%) had median overall survivals of 31.4, 27.3, and 4.8 months, respectively (p = 0.028). Conclusions: To the authors’ knowledge, this is the largest series of metastatic chrRCC patients and these results set new benchmarks for survival in clinical trial design and patient counseling. The IMDC criteria risk categories seem to stratify patients into appropriate favourable, intermediate, and poor risk groups, although larger patient numbers are required. It appears that outcomes between metastatic chrRCC and ccRCC are similar when treated with conventional targeted therapies. Patients with metastatic chrRCC can be treated with tyrosine kinase inhibitors and enrolled in clinical trials to further measure outcomes in this rare patient population.
Collapse
Affiliation(s)
- Steven M Yip
- Department of Medical Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| | | | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Anna Fraccon
- Medical Oncology, Casa di Cura Pederzoli, Peschiera Del Garda, Peschiera Del Garda (VR), Italy
| | | | - Brian I Rini
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Georg A Bjarnason
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Hao-Wen Sim
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - James Brugarolas
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kostas Koutsoukos
- Department of Clinical Therapeutics, Oncology Unit, Alexandra Hospital, University of Athens, Athens, Greece
| | | | - Takeshi Yuasa
- Department of Urology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ian Davis
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Ajjai Alva
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | | | - Toni K Choueiri
- Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | - Daniel Y C Heng
- Tom Baker Cancer Center, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
454
|
Casuscelli J, Vano YA, Fridman WH, Hsieh JJ. Molecular Classification of Renal Cell Carcinoma and Its Implication in Future Clinical Practice. KIDNEY CANCER 2017; 1:3-13. [PMID: 30334000 PMCID: PMC6179110 DOI: 10.3233/kca-170008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Renal cell carcinoma (RCC) encompasses a wide spectrum of morphologically and molecularly distinct (>10) cancer subtypes originated from the kidney epithelium. Metastatic RCC (mRCC) is lethal and refractory to conventional chemotherapeutic agents. The incorporation of targeted therapies and immune checkpoint inhibitors into the current practice of mRCC has markedly improved the median overall survival of clear cell RCC (ccRCC) patients, the most common subtype, but not rare kidney cancer (RKC or non-ccRCC, nccRCC). Varied treatment response in mRCC patients is observed, which presents clinical challenges/opportunities at the modern mRCC therapeutic landscape consisting of 12 approved drugs representing 6 different effective mechanisms. Key contributing factors include inter- and intra-RCC heterogeneity. With the advances in pan-omics technologies, we now have a better understanding of the molecular pathobiology of individual RCC subtype. Here, we attempt to classify ccRCC based on contemporary molecular features with emphasis on their respective potential significance in clinical practice.
Collapse
Affiliation(s)
| | - Yann-Alexandre Vano
- Oncologie Médicale, Hôpital Européen Georges Pompidou and Centre de Recherche des Cordeliers, Paris, France
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris 5 Descartes University, Paris, France
| | - Wolf Herve Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris 5 Descartes University, Paris, France
| | - James J. Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO, USA
| |
Collapse
|
455
|
Hong B, Yang Y, Guo S, Duoerkun S, Deng X, Chen D, Yu S, Qian W, Li Q, Li Q, Gong K, Zhang N. Intra-tumour molecular heterogeneity of clear cell renal cell carcinoma reveals the diversity of the response to targeted therapies using patient-derived xenograft models. Oncotarget 2017; 8:49839-49850. [PMID: 28548943 PMCID: PMC5564811 DOI: 10.18632/oncotarget.17765] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/26/2017] [Indexed: 01/14/2023] Open
Abstract
Inter- and intra-tumour molecular heterogeneity is increasingly recognized in clear cell renal cell carcinoma (ccRCC). It may partially explain the diversity of responses to targeted therapies and the various clinical outcomes. In this study, a 56-year-old male ccRCC patient with multiple metastases received radical nephrectomy and resection of the metastatic tumour in chest wall. The surgical specimens were implanted into nude mice to establish patient-derived xenograft (PDX) models with KI2367 model derived from the primary tumour and KI2368 model from the metastastic tumour. The two modles were treated with Sorafenib, Sunitinib, Axitinib, combined Sorafenib/Sunitinib, or alternating therapy of Sorafenib and Sunitinib. Significant anti-tumour activity was found in KI2367 treated with Sorafenib/Sunitinib monotherapy, combined Sorafenib/Sunitinib, and alternating therapy of Sorafenib/Sunitinib (P<0.05) but not in that treated with Axitinib monotherapy. In contrast, KI2368 was significantly responsive to Sunitinib monotherapy, combined Sorafenib/Sunitinib therapy and alternating therapy of Sorafenib/Sunitinib but not responsive to Sorafenib and Axitinib monotherapy (P<0.05). RNAseq of the two models demonstrated that the expression levels of 1,725 genes including the drug targeted genes of PDGFA, PDGFB and PDGFRA were >5-fold higher in KI2367 than in KI2368 and the expression levels of 994 genes were > 5-fold higher in KI2368 than in KI2367. These results suggest the presence of intra-tumour molecular heterogeneity in this patient. This heterogeneity may influence the response to targeted therapies. Multiple biopsy, liquid biopsy and genomic analysis of intra- tumour molecular heterogeneity may help guide a more precise and effective plan in selecting targeted therapies for ccRCC patients.
Collapse
Affiliation(s)
- Baoan Hong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing, P.R. China
| | - Yong Yang
- Department of Urology, Beijing Cancer Hospital, Beijing Institute for Cancer Research, Beijing, P.R. China
| | - Sheng Guo
- Division of Translational Oncology, Crown Bioscience, Taicang, Jiangsu, P.R. China
| | - Shayiremu Duoerkun
- Department of Urology, Central Hospital of HaMi Region, Xinjiang, P.R. China
| | - Xiaohu Deng
- Department of Urology, People's Hospital of Kelamayi, Xinjiang, P.R. China
| | - Dawei Chen
- Division of Translational Oncology, Crown Bioscience, Taicang, Jiangsu, P.R. China
| | - Shijun Yu
- Division of Translational Oncology, Crown Bioscience, Taicang, Jiangsu, P.R. China
| | - Wubin Qian
- Division of Translational Oncology, Crown Bioscience, Taicang, Jiangsu, P.R. China
| | - Qixiang Li
- Division of Translational Oncology, Crown Bioscience, Taicang, Jiangsu, P.R. China
| | - Qing Li
- Center for Cellular & Structural Biology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing, P.R. China
| | - Ning Zhang
- Department of Urology, Beijing Cancer Hospital, Beijing Institute for Cancer Research, Beijing, P.R. China
| |
Collapse
|
456
|
Renal Cell Carcinoma With Chromosome 6p Amplification Including the TFEB Gene: A Novel Mechanism of Tumor Pathogenesis? Am J Surg Pathol 2017; 41:287-298. [PMID: 28009604 DOI: 10.1097/pas.0000000000000776] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amplification of chromosome 6p has been implicated in aggressive behavior in several cancers, but has not been characterized in renal cell carcinoma (RCC). We identified 9 renal tumors with amplification of chromosome 6p including the TFEB gene, 3 by fluorescence in situ hybridization, and 6 from the Cancer Genome Atlas (TCGA) databases. Patients' ages were 28 to 78 years (median, 61 y). Most tumors were high stage (7/9 pT3a, 2/9 pN1). Using immunohistochemistry, 2/4 were positive for melanocytic markers and cathepsin K. Novel TFEB fusions were reported by TCGA in 2; however, due to a small composition of fusion transcripts compared with full-length transcripts (0.5/174 and 3.3/132 FPKM), we hypothesize that these represent secondary fusions due to amplification. Five specimens (4 TCGA, 1 fluorescence in situ hybridization) had concurrent chromosome 3p copy number loss or VHL deletion. However, these did not resemble clear cell RCC, had negative carbonic anhydrase IX labeling, lacked VHL mutation, and had papillary or unclassified histology (2/4 had gain of chromosome 7 or 17). One tumor each had somatic FH mutation and SMARCB1 mutation. Chromosome 6p amplification including TFEB is a previously unrecognized cytogenetic alteration in RCC, associated with heterogenous tubulopapillary eosinophilic and clear cell histology. The combined constellation of features does not fit cleanly into an existing tumor category (unclassified), most closely resembling papillary or translocation RCC. The tendency for high tumor stage, varied tubulopapillary morphology, and a subset with melanocytic marker positivity suggests the possibility of a unique tumor type, despite some variation in appearance and genetics.
Collapse
|
457
|
Wang Z, Kim TB, Peng B, Karam J, Creighton C, Joon A, Kawakami F, Trevisan P, Jonasch E, Chow CW, Canales JR, Tamboli P, Tannir N, Wood C, Monzon F, Baggerly K, Varella-Garcia M, Czerniak B, Wistuba I, Mills G, Shaw K, Chen K, Sircar K. Sarcomatoid Renal Cell Carcinoma Has a Distinct Molecular Pathogenesis, Driver Mutation Profile, and Transcriptional Landscape. Clin Cancer Res 2017; 23:6686-6696. [PMID: 28710314 DOI: 10.1158/1078-0432.ccr-17-1057] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/02/2017] [Accepted: 07/10/2017] [Indexed: 01/03/2023]
Abstract
Purpose: Sarcomatoid renal cell carcinoma (SRCC) ranks among the most aggressive clinicopathologic phenotypes of RCC. However, the paucity of high-quality, genome-wide molecular examinations of SRCC has hindered our understanding of this entity.Experimental Design: We interrogated the mutational, copy number, and transcriptional characteristics of SRCC and compared these data with those of nonsarcomatoid RCC (RCC). We evaluated whole-exome sequencing, single-nucleotide polymorphism, and RNA sequencing data from patients with SRCC (n = 65) and RCC (n = 598) across different parent RCC subtypes, including clear-cell RCC, papillary RCC, and chromophobe RCC subtypes.Results: SRCC was molecularly discrete from RCC and clustered according to its parent RCC subtype, though with upregulation of TGFβ signaling across all subtypes. The epithelioid (E-) and spindled (S-) histologic components of SRCC did not show differences in mutational load among cancer-related genes despite a higher mutational burden in S-. Notably, sarcomatoid clear-cell RCC (SccRCC) showed significantly fewer deletions at 3p21-25, a lower rate of two-hit loss for VHL and PBRM1, and more mutations in PTEN, TP53, and RELN compared with ccRCC. A two-hit loss involving VHL predicted for ccRCC and a better prognosis, whereas mutations in PTEN, TP53, or RELN predicted for SccRCC and worse prognosis.Conclusions: SRCC segregates by parent subtype, and SccRCC has a fundamentally different early molecular pathogenesis, usually lacking the classic 3p21-25 deletion and showing distinctive mutational and transcriptional profiles. These features prompt a more precise molecular classification of RCC, with diagnostic, prognostic, and therapeutic implications. Clin Cancer Res; 23(21); 6686-96. ©2017 AACRSee related commentary by Bergerot et al., p. 6381.
Collapse
Affiliation(s)
- Zixing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tae Beom Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bo Peng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jose Karam
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chad Creighton
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Aron Joon
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fumi Kawakami
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Trevisan
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chi-Wan Chow
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaime Rodriguez Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pheroze Tamboli
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nizar Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher Wood
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Keith Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon Mills
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kenna Shaw
- Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kanishka Sircar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
458
|
Robinson KM, Hawkins AS, Santana-Cruz I, Adkins RS, Shetty AC, Nagaraj S, Sadzewicz L, Tallon LJ, Rasko DA, Fraser CM, Mahurkar A, Silva JC, Dunning Hotopp JC. Aligner optimization increases accuracy and decreases compute times in multi-species sequence data. Microb Genom 2017; 3:e000122. [PMID: 29114401 PMCID: PMC5643015 DOI: 10.1099/mgen.0.000122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/04/2017] [Indexed: 01/01/2023] Open
Abstract
As sequencing technologies have evolved, the tools to analyze these sequences have made similar advances. However, for multi-species samples, we observed important and adverse differences in alignment specificity and computation time for bwa- mem (Burrows-Wheeler aligner-maximum exact matches) relative to bwa-aln. Therefore, we sought to optimize bwa-mem for alignment of data from multi-species samples in order to reduce alignment time and increase the specificity of alignments. In the multi-species cases examined, there was one majority member (i.e. Plasmodium falciparum or Brugia malayi) and one minority member (i.e. human or the Wolbachia endosymbiont wBm) of the sequence data. Increasing bwa-mem seed length from the default value reduced the number of read pairs from the majority sequence member that incorrectly aligned to the reference genome of the minority sequence member. Combining both source genomes into a single reference genome increased the specificity of mapping, while also reducing the central processing unit (CPU) time. In Plasmodium, at a seed length of 18 nt, 24.1 % of reads mapped to the human genome using 1.7±0.1 CPU hours, while 83.6 % of reads mapped to the Plasmodium genome using 0.2±0.0 CPU hours (total: 107.7 % reads mapping; in 1.9±0.1 CPU hours). In contrast, 97.1 % of the reads mapped to a combined Plasmodium-human reference in only 0.7±0.0 CPU hours. Overall, the results suggest that combining all references into a single reference database and using a 23 nt seed length reduces the computational time, while maximizing specificity. Similar results were found for simulated sequence reads from a mock metagenomic data set. We found similar improvements to computation time in a publicly available human-only data set.
Collapse
Affiliation(s)
- Kelly M Robinson
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aziah S Hawkins
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ivette Santana-Cruz
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ricky S Adkins
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amol C Shetty
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sushma Nagaraj
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lisa Sadzewicz
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luke J Tallon
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David A Rasko
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.,2Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claire M Fraser
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.,3Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anup Mahurkar
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joana C Silva
- 1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.,2Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Julie C Dunning Hotopp
- 2Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.,1Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
459
|
Colomba E, Le Teuff G, Eisen T, Stewart GD, Fife K, Larkin J, Biondo A, Pickering L, Srinivasan A, Boyle H, Derosa L, Sternberg CN, Recine F, Ralph C, Saldana C, Barthélémy P, Bernhard JC, Gurney H, Verhoest G, Vauleon E, Bigot P, Berger J, Pfister C, Gravis G, Rodier JM, Culine S, Caty A, Rolland F, Priou F, Escudier B, Albiges L. Metastatic chromophobe renal cell carcinoma treated with targeted therapies: A Renal Cross Channel Group study. Eur J Cancer 2017; 80:55-62. [DOI: 10.1016/j.ejca.2017.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/05/2017] [Accepted: 03/13/2017] [Indexed: 01/26/2023]
|
460
|
Farshidfar F, Zheng S, Gingras MC, Newton Y, Shih J, Robertson AG, Hinoue T, Hoadley KA, Gibb EA, Roszik J, Covington KR, Wu CC, Shinbrot E, Stransky N, Hegde A, Yang JD, Reznik E, Sadeghi S, Pedamallu CS, Ojesina AI, Hess JM, Auman JT, Rhie SK, Bowlby R, Borad MJ, Zhu AX, Stuart JM, Sander C, Akbani R, Cherniack AD, Deshpande V, Mounajjed T, Foo WC, Torbenson M, Kleiner DE, Laird PW, Wheeler DA, McRee AJ, Bathe OF, Andersen JB, Bardeesy N, Roberts LR, Kwong LN. Integrative Genomic Analysis of Cholangiocarcinoma Identifies Distinct IDH-Mutant Molecular Profiles. Cell Rep 2017; 19:2878-2880. [PMID: 28658632 PMCID: PMC6141445 DOI: 10.1016/j.celrep.2017.06.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy of the bile ducts, with poor prognosis and limited treatment options. Here, we describe the integrated analysis of somatic mutations, RNA expression, copy number, and DNA methylation by The Cancer Genome Atlas, of a set of predominantly intrahepatic CCA cases, and propose a molecular classification scheme. We identified an IDH -mutant enriched subtype with distinct molecular features including low expression of chromatin modifiers, elevated expression of mitochondrial genes, and increased mitochondrial DNA copy number. Leveraging the multi-platform data, we observed that ARID1A exhibited DNA hypermethylation and decreased expression in the IDH -mutant subtype. More broadly, we found that IDH mutations are associated with an expanded histological spectrum of liver tumors with molecular features that stratify with CCA. Our studies reveal insights into the molecular pathogenesis and heterogeneity of cholangiocarcinoma and provide classification information of potential therapeutic significance.
Collapse
Affiliation(s)
- Farshad Farshidfar
- Departments of Surgery and Oncology, Arnie Charbonneau
Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Siyuan Zheng
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Marie-Claude Gingras
- Human Genome Sequencing Center, Baylor College of Medicine,
Houston, TX 77030, USA
| | - Yulia Newton
- University of California Santa Cruz, Santa Cruz, CA 95064,
USA
| | - Juliann Shih
- The Eli and Edythe L. Broad Institute of Massachusetts
Institute of Technology and Harvard University, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer
Institute, Boston, MA 02215, USA
| | - A. Gordon Robertson
- Canada’s Michael Smith Genome Sciences Centre, BC
Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Toshinori Hinoue
- Center for Epigenetics, Van Andel Research Institute, Grand
Rapids, MI 49503
| | - Katherine A. Hoadley
- Departments of Genetics and Pathology and Laboratory
Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,
USA
- Lineberger Comprehensive Cancer Center, University of
North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ewan A. Gibb
- Canada’s Michael Smith Genome Sciences Centre, BC
Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Jason Roszik
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Kyle R. Covington
- Human Genome Sequencing Center, Baylor College of Medicine,
Houston, TX 77030, USA
| | - Chia-Chin Wu
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Eve Shinbrot
- Human Genome Sequencing Center, Baylor College of Medicine,
Houston, TX 77030, USA
| | | | - Apurva Hegde
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Ju Dong Yang
- Divisions of Gastroenterology and Hepatology and
Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
55905, USA
| | - Ed Reznik
- Memorial Sloan Kettering Cancer Center, New York, NY
10005, USA
| | - Sara Sadeghi
- Canada’s Michael Smith Genome Sciences Centre, BC
Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Chandra Sekhar Pedamallu
- The Eli and Edythe L. Broad Institute of Massachusetts
Institute of Technology and Harvard University, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer
Institute, Boston, MA 02215, USA
| | - Akinyemi I. Ojesina
- University of Alabama at Birmingham, Birmingham, AL
35294, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, AL
35806, USA
| | - Julian M. Hess
- The Eli and Edythe L. Broad Institute of Massachusetts
Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - J. Todd Auman
- Departments of Genetics and Pathology and Laboratory
Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599,
USA
| | - Suhn K. Rhie
- University of Southern California, USC/Norris
Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Reanne Bowlby
- Canada’s Michael Smith Genome Sciences Centre, BC
Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Mitesh J. Borad
- Division of Hematology and Oncology, Mayo Clinic,
Scottsdale, AZ 85054, USA
| | | | - Andrew X Zhu
- Departments of Hematology and Oncology, Massachusetts
General Hospital, Boston, MA 02114, USA
| | - Josh M. Stuart
- University of California Santa Cruz, Santa Cruz, CA 95064,
USA
| | - Chris Sander
- Memorial Sloan Kettering Cancer Center, New York, NY
10005, USA
| | - Rehan Akbani
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Andrew D. Cherniack
- The Eli and Edythe L. Broad Institute of Massachusetts
Institute of Technology and Harvard University, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer
Institute, Boston, MA 02215, USA
| | - Vikram Deshpande
- Departments of Pathology and Oncology, Massachusetts
General Hospital, Boston, MA 02114, USA
| | - Taofic Mounajjed
- Divisions of Gastroenterology and Hepatology and
Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
55905, USA
| | - Wai Chin Foo
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| | - Michael Torbenson
- Divisions of Gastroenterology and Hepatology and
Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
55905, USA
| | | | - Peter W. Laird
- Center for Epigenetics, Van Andel Research Institute, Grand
Rapids, MI 49503
| | - David A. Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine,
Houston, TX 77030, USA
| | - Autumn J. McRee
- Lineberger Comprehensive Cancer Center, University of
North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Oliver F. Bathe
- Departments of Surgery and Oncology, Arnie Charbonneau
Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre, University of
Copenhagen, DK-2200, Denmark
| | - Nabeel Bardeesy
- Departments of Pathology and Oncology, Massachusetts
General Hospital, Boston, MA 02114, USA
| | - Lewis R. Roberts
- Divisions of Gastroenterology and Hepatology and
Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
55905, USA
| | - Lawrence N. Kwong
- Departments of Genomic Medicine, Melanoma Medical Oncology,
Bioinformatics and Computational Biology, Pathology, and Translational Molecular
Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030,
USA
| |
Collapse
|
461
|
Hu S, Yuan H, Li Z, Zhang J, Wu J, Chen Y, Shi Q, Ren W, Shao N, Ying X. Transcriptional response profiles of paired tumor-normal samples offer novel perspectives in pan-cancer analysis. Oncotarget 2017; 8:41334-41347. [PMID: 28489584 PMCID: PMC5522216 DOI: 10.18632/oncotarget.17295] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 04/03/2017] [Indexed: 01/05/2023] Open
Abstract
Both tumor and adjacent normal tissues are valuable in cancer research. Transcriptional response profiles represent the changes of gene expression levels between paired tumor and adjacent normal tissues. In this study, we performed a pan-cancer analysis based on the transcriptional response profiles from 633 samples across 13 cancer types. We obtained two interesting results. Using consensus clustering method, we characterized ten clusters with distinct transcriptional response patterns and enriched pathways. Notably, head and neck squamous cell carcinoma was divided in two subtypes, enriched in cell cycle-related pathways and cell adhesion-related pathways respectively. The other interesting result is that we identified 92 potential pan-cancer genes that were consistently upregulated across multiple cancer types. Knockdown of FAM64A or TROAP inhibited the growth of cancer cells, suggesting that these genes may promote tumor development and are worthy of further validations. Our results suggest that transcriptional response profiles of paired tumor-normal tissues can provide novel perspectives in pan-cancer analysis.
Collapse
Affiliation(s)
- Shuofeng Hu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Hanyu Yuan
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zongcheng Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Translational Medicine Center of Stem Cells, 307-Ivy Translational Medicine Center, Laboratory of Oncology, Affiliated Hospital, Academy of Military Medical Sciences, Beijing 100071, China
| | - Jian Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jiaqi Wu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yaowen Chen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Department of Obstetrics and Gynecology, Fuzhou General Hospital of Nanjing Military Command, Fujian 350025, China
| | - Qiang Shi
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Wu Ren
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jilin University, Changchun 130021, China
| | - Ningsheng Shao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xiaomin Ying
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|
462
|
Casuscelli J, Weinhold N, Gundem G, Wang L, Zabor EC, Drill E, Wang PI, Nanjangud GJ, Redzematovic A, Nargund AM, Manley BJ, Arcila ME, Donin NM, Cheville JC, Thompson RH, Pantuck AJ, Russo P, Cheng EH, Lee W, Tickoo SK, Ostrovnaya I, Creighton CJ, Papaemmanuil E, Seshan VE, Hakimi AA, Hsieh JJ. Genomic landscape and evolution of metastatic chromophobe renal cell carcinoma. JCI Insight 2017; 2:92688. [PMID: 28614790 PMCID: PMC5470887 DOI: 10.1172/jci.insight.92688] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/16/2017] [Indexed: 12/31/2022] Open
Abstract
Chromophobe renal cell carcinoma (chRCC) typically shows ~7 chromosome losses (1, 2, 6, 10, 13, 17, and 21) and ~31 exonic somatic mutations, yet carries ~5%-10% metastatic incidence. Since extensive chromosomal losses can generate proteotoxic stress and compromise cellular proliferation, it is intriguing how chRCC, a tumor with extensive chromosome losses and a low number of somatic mutations, can develop lethal metastases. Genomic features distinguishing metastatic from nonmetastatic chRCC are unknown. An integrated approach, including whole-genome sequencing (WGS), targeted ultradeep cancer gene sequencing, and chromosome analyses (FACETS, OncoScan, and FISH), was performed on 79 chRCC patients including 38 metastatic (M-chRCC) cases. We demonstrate that TP53 mutations (58%), PTEN mutations (24%), and imbalanced chromosome duplication (ICD, duplication of ≥ 3 chromosomes) (25%) were enriched in M-chRCC. Reconstruction of the subclonal composition of paired primary-metastatic chRCC tumors supports the role of TP53, PTEN, and ICD in metastatic evolution. Finally, the presence of these 3 genomic features in primary tumors of both The Cancer Genome Atlas kidney chromophobe (KICH) (n = 64) and M-chRCC (n = 35) cohorts was associated with worse survival. In summary, our study provides genomic insights into the metastatic progression of chRCC and identifies TP53 mutations, PTEN mutations, and ICD as high-risk features.
Collapse
Affiliation(s)
- Jozefina Casuscelli
- Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| | | | | | | | | | | | - Patricia I. Wang
- Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Almedina Redzematovic
- Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, and
| | - Amrita M. Nargund
- Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brandon J. Manley
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | - Paul Russo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Emily H. Cheng
- Department of Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Pathology
| | | | | | | | - Chad J. Creighton
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - A. Ari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - James J. Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
463
|
Bailey ST, Smith AM, Kardos J, Wobker SE, Wilson HL, Krishnan B, Saito R, Lee HJ, Zhang J, Eaton SC, Williams LA, Manocha U, Peters DJ, Pan X, Carroll TJ, Felsher DW, Walter V, Zhang Q, Parker JS, Yeh JJ, Moffitt RA, Leung JY, Kim WY. MYC activation cooperates with Vhl and Ink4a/Arf loss to induce clear cell renal cell carcinoma. Nat Commun 2017; 8:15770. [PMID: 28593993 PMCID: PMC5472759 DOI: 10.1038/ncomms15770] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 04/26/2017] [Indexed: 11/17/2022] Open
Abstract
Renal carcinoma is a common and aggressive malignancy whose histopathogenesis is incompletely understood and that is largely resistant to cytotoxic chemotherapy. We present two mouse models of kidney cancer that recapitulate the genomic alterations found in human papillary (pRCC) and clear cell RCC (ccRCC), the most common RCC subtypes. MYC activation results in highly penetrant pRCC tumours (MYC), while MYC activation, when combined with Vhl and Cdkn2a (Ink4a/Arf) deletion (VIM), produce kidney tumours that approximate human ccRCC. RNAseq of the mouse tumours demonstrate that MYC tumours resemble Type 2 pRCC, which are known to harbour MYC activation. Furthermore, VIM tumours more closely simulate human ccRCC. Based on their high penetrance, short latency, and histologic fidelity, these models of papillary and clear cell RCC should be significant contributions to the field of kidney cancer research. Renal cell carcinoma (RCC) is a common and aggressive malignancy. Here, the authors generate two mouse models of the most common RCC subtypes: the human papillary RCC through MYC activation and clear cell RCC through MYC activation combined with Vhl and Cdkn2a deletion.
Collapse
Affiliation(s)
- Sean T Bailey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Aleisha M Smith
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jordan Kardos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Sara E Wobker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Harper L Wilson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Bhavani Krishnan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ryoichi Saito
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Hyo Jin Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
| | - Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Samuel C Eaton
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Lindsay A Williams
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ujjawal Manocha
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Dorien J Peters
- Department of Pathology, Leiden University Medical Center, Leiden 2333, The Netherlands
| | - Xinchao Pan
- Departments of Internal Medicine and Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Thomas J Carroll
- Departments of Internal Medicine and Molecular Biology, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Dean W Felsher
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California 94305-5151, USA
| | - Vonn Walter
- Department of Biochemistry and Molecular Biology, Penn State Milton S. Hershey College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Jen Jen Yeh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard A Moffitt
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Janet Y Leung
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
464
|
Farber NJ, Kim CJ, Modi PK, Hon JD, Sadimin ET, Singer EA. Renal cell carcinoma: the search for a reliable biomarker. Transl Cancer Res 2017; 6:620-632. [PMID: 28775935 PMCID: PMC5538266 DOI: 10.21037/tcr.2017.05.19] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
One particular challenge in the treatment of kidney tumors is the range of histologies and tumor phenotypes a renal mass can represent. A kidney tumor can range from benign (e.g., oncocytoma) to a clinically indolent malignancy (e.g., papillary type I, chromophobe) to aggressive disease [e.g., papillary type II or high-grade clear cell renal cell carcinoma (ccRCC)]. Even among various subtypes, kidney cancers are genetically diverse with variable prognoses and treatment response rates. Therefore, the key to proper treatment is the differentiation of these subtypes. Currently, a wide array of diagnostic, prognostic, and predictive biomarkers exist that may help guide the individualized care of kidney cancer patients. This review will discuss the various serum, urine, imaging, and immunohistological biomarkers available in practice.
Collapse
Affiliation(s)
- Nicholas J. Farber
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christopher J. Kim
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Parth K. Modi
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jane D. Hon
- Section of Urologic Pathology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Evita T. Sadimin
- Section of Urologic Pathology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
465
|
Harlander S, Schönenberger D, Toussaint NC, Prummer M, Catalano A, Brandt L, Moch H, Wild PJ, Frew IJ. Combined mutation in Vhl, Trp53 and Rb1 causes clear cell renal cell carcinoma in mice. Nat Med 2017; 23:869-877. [PMID: 28553932 PMCID: PMC5509015 DOI: 10.1038/nm.4343] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 04/15/2017] [Indexed: 12/17/2022]
Abstract
Clear cell renal cell carcinomas (ccRCC) frequently exhibit inactivation of the VHL tumour suppressor gene and often harbour multiple copy number alterations in genes that regulate cell cycle progression. We show here that modelling these genetic alterations by combined renal epithelium-specific deletion of Vhl, Trp53 and Rb1 in mice caused ccRCC. These tumours arose from proximal tubule epithelial cells and shared molecular markers and mRNA expression profiles with human ccRCC. Exome sequencing revealed that mouse and human ccRCCs exhibit recurrent mutations in genes associated with the primary cilium, uncovering a mutational convergence on this organelle and implicating a subset of ccRCCs as genetic ciliopathies. Different mouse tumours responded differently to standard therapies for advanced human ccRCC, mimicking the range of clinical behaviours in the human disease. Inhibition of HIF-α transcription factors with Acriflavine as third line therapy had therapeutic effects in some tumours, providing pre-clinical evidence for further investigation of HIF-α inhibition as a ccRCC treatment. This autochthonous mouse ccRCC model represents a tool to investigate the biology of ccRCC and to identify new treatment strategies.
Collapse
Affiliation(s)
- Sabine Harlander
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | | | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Michael Prummer
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Antonella Catalano
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Center for Translational Cell Research, Clinic of Internal Medicine I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Brandt
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Peter J Wild
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Ian J Frew
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Center for Translational Cell Research, Clinic of Internal Medicine I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
466
|
Beksac AT, Paulucci DJ, Blum KA, Yadav SS, Sfakianos JP, Badani KK. Heterogeneity in renal cell carcinoma. Urol Oncol 2017; 35:507-515. [PMID: 28551412 DOI: 10.1016/j.urolonc.2017.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/20/2017] [Accepted: 05/08/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION In recent years, molecular characterization of renal cell carcinoma has facilitated the identification of driver genes, specific molecular pathways, and characterization of the tumor microenvironment, which has led to a better understanding of the disease. This comprehension has revolutionized the treatment for patients with metastatic disease, but despite these advancements many patients will develop resistance leading to treatment failure. A primary cause of this resistance and subsequent treatment failure is tumor heterogeneity. We reviewed the literature on the mechanisms of tumor heterogeneity and its clinical implications. METHODS A comprehensive literature search was performed using the MEDLINE/PubMed Index. RESULTS Intertumor and intratumor heterogeneity is possibly a reason for treatment failure and development of resistance. Specifically, the genetic profile of a renal tumor differs spatially within a tumor as well as among patients. Genomic mutations can change temporally with resistant subclones becoming dominant over time. CONCLUSIONS Accounting for intratumor and intertumor heterogeneity with better sampling of cancer tissue is needed. This will hopefully lead to improved identification of driver mutations and actionable targets. Only then, we can move past the one-size-fits-all approach toward personalized treatment based on each individual׳s molecular profile.
Collapse
Affiliation(s)
- Alp Tuna Beksac
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David J Paulucci
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kyle A Blum
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shalini Singh Yadav
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John P Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ketan K Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
467
|
Abstract
Technical advances in the development of organoid systems enable cell lines, primary adult cells, or stem or progenitor cells to develop into diverse, multicellular entities, which can self-renew, self-organize, and differentiate. These 3D organoid cultures have proven to be of value in increasing our understanding of the biology of disease and offer the potential of regenerative and genetic therapies. The successful application of 3D organoids derived from adult tissue into urological cancer research can further our understanding of these diseases and could also provide preclinical cancer models to realize the precision medicine paradigm by therapeutic screening of individual patient samples ex vivo. Kidney organoids derived from induced pluripotent stem cells provide personalized biomarkers, which can be correlated with genetic and clinical information. Organoid models can also improve our comprehension of aspects of particular diseases; for example, in prostate cancer, 3D organoids can aid in the identification of tumour-initiating cells from an epithelial cell lineage. Furthermore, kidney organoid differentiation from human pluripotent stem cells enables gene editing to model disease in kidney tubular epithelial cells. State-of-the-art human organoid cultures have potential as tools in basic and clinical research in renal, bladder, and prostatic diseases.
Collapse
Affiliation(s)
- Shangqian Wang
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Urology Department, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dong Gao
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Key Laboratory of Systems Biology,CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.,Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| |
Collapse
|
468
|
Analysis of renal cancer cell lines from two major resources enables genomics-guided cell line selection. Nat Commun 2017; 8:15165. [PMID: 28489074 PMCID: PMC5436135 DOI: 10.1038/ncomms15165] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/06/2017] [Indexed: 12/19/2022] Open
Abstract
The utility of cancer cell lines is affected by the similarity to endogenous tumour cells. Here we compare genomic data from 65 kidney-derived cell lines from the Cancer Cell Line Encyclopedia and the COSMIC Cell Lines Project to three renal cancer subtypes from The Cancer Genome Atlas: clear cell renal cell carcinoma (ccRCC, also known as kidney renal clear cell carcinoma), papillary (pRCC, also known as kidney papillary) and chromophobe (chRCC, also known as kidney chromophobe) renal cell carcinoma. Clustering copy number alterations shows that most cell lines resemble ccRCC, a few (including some often used as models of ccRCC) resemble pRCC, and none resemble chRCC. Human ccRCC tumours clustering with cell lines display clinical and genomic features of more aggressive disease, suggesting that cell lines best represent aggressive tumours. We stratify mutations and copy number alterations for important kidney cancer genes by the consistency between databases, and classify cell lines into established gene expression-based indolent and aggressive subtypes. Our results could aid investigators in analysing appropriate renal cancer cell lines.
Collapse
|
469
|
Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017. [PMID: 28481359 DOI: 10.1038/nm.4333] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Tumor molecular profiling is a fundamental component of precision oncology, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. The existence of recurrent targetable alterations across distinct histologically defined tumor types, coupled with an expanding portfolio of molecularly targeted therapies, demands flexible and comprehensive approaches to profile clinically relevant genes across the full spectrum of cancers. We established a large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which we have compiled tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer and available pathological and clinical annotations. Using these data, we identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types. Patients were enrolled on genomically matched clinical trials at a rate of 11%. To enable discovery of novel biomarkers and deeper investigation into rare alterations and tumor types, all results are publicly accessible.
Collapse
|
470
|
Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, Srinivasan P, Gao J, Chakravarty D, Devlin SM, Hellmann MD, Barron DA, Schram AM, Hameed M, Dogan S, Ross DS, Hechtman JF, DeLair DF, Yao J, Mandelker DL, Cheng DT, Chandramohan R, Mohanty AS, Ptashkin RN, Jayakumaran G, Prasad M, Syed MH, Rema AB, Liu ZY, Nafa K, Borsu L, Sadowska J, Casanova J, Bacares R, Kiecka IJ, Razumova A, Son JB, Stewart L, Baldi T, Mullaney KA, Al-Ahmadie H, Vakiani E, Abeshouse AA, Penson AV, Jonsson P, Camacho N, Chang MT, Won HH, Gross BE, Kundra R, Heins ZJ, Chen HW, Phillips S, Zhang H, Wang J, Ochoa A, Wills J, Eubank M, Thomas SB, Gardos SM, Reales DN, Galle J, Durany R, Cambria R, Abida W, Cercek A, Feldman DR, Gounder MM, Hakimi AA, Harding JJ, Iyer G, Janjigian YY, Jordan EJ, Kelly CM, Lowery MA, Morris LGT, Omuro AM, Raj N, Razavi P, Shoushtari AN, Shukla N, Soumerai TE, Varghese AM, Yaeger R, Coleman J, Bochner B, Riely GJ, Saltz LB, Scher HI, Sabbatini PJ, Robson ME, Klimstra DS, Taylor BS, Baselga J, Schultz N, Hyman DM, Arcila ME, Solit DB, Ladanyi M, Berger MF. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017; 23:703-713. [PMID: 28481359 PMCID: PMC5461196 DOI: 10.1038/nm.4333] [Citation(s) in RCA: 2275] [Impact Index Per Article: 325.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/04/2017] [Indexed: 02/07/2023]
Abstract
Tumor molecular profiling is a fundamental component of precision oncology, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. The existence of recurrent targetable alterations across distinct histologically defined tumor types, coupled with an expanding portfolio of molecularly targeted therapies, demands flexible and comprehensive approaches to profile clinically relevant genes across the full spectrum of cancers. We established a large-scale, prospective clinical sequencing initiative using a comprehensive assay, MSK-IMPACT, through which we have compiled tumor and matched normal sequence data from a unique cohort of more than 10,000 patients with advanced cancer and available pathological and clinical annotations. Using these data, we identified clinically relevant somatic mutations, novel noncoding alterations, and mutational signatures that were shared by common and rare tumor types. Patients were enrolled on genomically matched clinical trials at a rate of 11%. To enable discovery of novel biomarkers and deeper investigation into rare alterations and tumor types, all results are publicly accessible.
Collapse
Affiliation(s)
- Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ronak H Shah
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Aijazuddin Syed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sumit Middha
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hyunjae R Kim
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Preethi Srinivasan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jianjiong Gao
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Debyani Chakravarty
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David A Barron
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Meera Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Deborah F DeLair
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - JinJuan Yao
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Diana L Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Donavan T Cheng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Raghu Chandramohan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Abhinita S Mohanty
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ryan N Ptashkin
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gowtham Jayakumaran
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Meera Prasad
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mustafa H Syed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Zhen Y Liu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Khedoudja Nafa
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Justyna Sadowska
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jacklyn Casanova
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ruben Bacares
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Iwona J Kiecka
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anna Razumova
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Julie B Son
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lisa Stewart
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tessara Baldi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kerry A Mullaney
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Adam A Abeshouse
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Alexander V Penson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Philip Jonsson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Niedzica Camacho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matthew T Chang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Helen H Won
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Benjamin E Gross
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ritika Kundra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zachary J Heins
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hsiao-Wei Chen
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sarah Phillips
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hongxin Zhang
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jiaojiao Wang
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Angelica Ochoa
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonathan Wills
- Information Systems, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael Eubank
- Information Systems, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stacy B Thomas
- Information Systems, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stuart M Gardos
- Information Systems, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dalicia N Reales
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jesse Galle
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert Durany
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Roy Cambria
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mrinal M Gounder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - A Ari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - James J Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gopa Iyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Emmet J Jordan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ciara M Kelly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maeve A Lowery
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Antonio M Omuro
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nitya Raj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tara E Soumerai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jonathan Coleman
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Bernard Bochner
- Clinical Research Administration, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gregory J Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Leonard B Saltz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Paul J Sabbatini
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David S Klimstra
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Barry S Taylor
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jose Baselga
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David B Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
471
|
Abstract
PURPOSE This AUA Guideline focuses on evaluation/counseling and management of adult patients with clinically localized renal masses suspicious for cancer, including solid-enhancing tumors and Bosniak 3/4 complex-cystic lesions. MATERIALS AND METHODS Systematic review utilized research from the Agency for Healthcare Research and Quality and additional supplementation by the authors and consultant methodologists. Evidence-based statements were based on body of evidence strength Grade A/B/C (Strong/Moderate/Conditional Recommendations, respectively) with additional statements presented as Clinical Principles or Expert Opinions. RESULTS Great progress has been made since the previous guidelines on management of localized renal masses were released (2009). The current guidelines provide updated, evidence-based recommendations regarding evaluation/counseling of patients with clinically localized renal masses, including the evolving role of renal mass biopsy. Given great variability of clinical, oncologic and functional characteristics, index patients are not utilized and the panel advocates individualized counseling/management. Management options (partial nephrectomy/radical nephrectomy/thermal ablation/active surveillance) are reviewed including recent data about comparative effectiveness and potential morbidities. Oncologic issues are prioritized while recognizing that functional outcomes are of great importance for survivorship for most patients with localized kidney cancer. A more restricted role for radical nephrectomy is recommended following well-defined selection criteria. Priority for partial nephrectomy is recommended for clinical T1a lesions, along with selective use of thermal ablation, particularly for tumors ≤3.0 cm. Important considerations for shared decision-making about active surveillance are explicitly defined. CONCLUSIONS Several factors should be considered during counseling/management of patients with clinically localized renal masses, including general health/comorbidities, oncologic potential of the mass, pertinent functional issues and relative efficacy/potential morbidities of various management strategies.
Collapse
|
472
|
Gu YF, Cohn S, Christie A, McKenzie T, Wolff N, Do QN, Madhuranthakam AJ, Pedrosa I, Wang T, Dey A, Busslinger M, Xie XJ, Hammer RE, McKay RM, Kapur P, Brugarolas J. Modeling Renal Cell Carcinoma in Mice: Bap1 and Pbrm1 Inactivation Drive Tumor Grade. Cancer Discov 2017; 7:900-917. [PMID: 28473526 DOI: 10.1158/2159-8290.cd-17-0292] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/29/2017] [Accepted: 05/02/2017] [Indexed: 12/22/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by BAP1 and PBRM1 mutations, which are associated with tumors of different grade and prognosis. However, whether BAP1 and PBRM1 loss causes ccRCC and determines tumor grade is unclear. We conditionally targeted Bap1 and Pbrm1 (with Vhl) in the mouse using several Cre drivers. Sglt2 and Villin proximal convoluted tubule drivers failed to cause tumorigenesis, challenging the conventional notion of ccRCC origins. In contrast, targeting with PAX8, a transcription factor frequently overexpressed in ccRCC, led to ccRCC of different grades. Bap1-deficient tumors were of high grade and showed greater mTORC1 activation than Pbrm1-deficient tumors, which exhibited longer latency. Disrupting one allele of the mTORC1 negative regulator, Tsc1, in Pbrm1-deficient kidneys triggered higher grade ccRCC. This study establishes Bap1 and Pbrm1 as lineage-specific drivers of ccRCC and histologic grade, implicates mTORC1 as a tumor grade rheostat, and suggests that ccRCCs arise from Bowman capsule cells.Significance: Determinants of tumor grade and aggressiveness across cancer types are poorly understood. Using ccRCC as a model, we show that Bap1 and Pbrm1 loss drives tumor grade. Furthermore, we show that the conversion from low grade to high grade can be promoted by activation of mTORC1. Cancer Discov; 7(8); 900-17. ©2017 AACR.See related commentary by Leung and Kim, p. 802This article is highlighted in the In This Issue feature, p. 783.
Collapse
Affiliation(s)
- Yi-Feng Gu
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shannon Cohn
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tiffani McKenzie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nicholas Wolff
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quyen N Do
- Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ananth J Madhuranthakam
- Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ivan Pedrosa
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Radiology and the Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tao Wang
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Quantitative Biomedical Research Center, Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anwesha Dey
- Department of Molecular Oncology, Genentech, South San Francisco, California
| | | | - Xian-Jin Xie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Robert E Hammer
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Renée M McKay
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas.,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas. .,Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, The University of Texas Southwestern Medical Center, Dallas, Texas. .,Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
473
|
Lee S, Borah S, Bahrami A. Detection of Aberrant TERT Promoter Methylation by Combined Bisulfite Restriction Enzyme Analysis for Cancer Diagnosis. J Mol Diagn 2017; 19:378-386. [PMID: 28284778 PMCID: PMC5417004 DOI: 10.1016/j.jmoldx.2017.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/13/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Aberrant CpG dinucleotide methylation in a specific region of the telomerase reverse transcriptase (TERT) promoter is associated with increased TERT mRNA levels and malignancy in several cancer types. However, routine screening of this region to aid cancer diagnosis can be challenging because i) several established methylation assays may inaccurately report on hypermethylation of this particular region, ii) interpreting the results of methylation assays can sometimes be difficult for clinical laboratories, and iii) use of high-throughput methylation assays for a few patient samples can be cost prohibitive. Herein, we describe the use of combined bisulfite restriction enzyme analysis (COBRA) as a diagnostic tool for detecting the hypermethylated TERT promoter using in vitro methylated and unmethylated genomic DNA as well as genomic DNA from four melanomas and two benign melanocytic lesions. We compare COBRA with MassARRAY, a more commonly used high-throughput approach, in screening for promoter hypermethylation in 28 formalin-fixed, paraffin-embedded neuroblastoma samples. COBRA sensitively and specifically detected samples with hypermethylated TERT promoter and was as effective as MassARRAY at differentiating high-risk from benign or low-risk tumors. This study demonstrates the utility of this low-cost, technically straightforward, and easily interpretable assay for cancer diagnosis in tumors of an ambiguous nature.
Collapse
Affiliation(s)
- Seungjae Lee
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sumit Borah
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
474
|
Diagnostic criteria for oncocytic renal neoplasms: a survey of urologic pathologists. Hum Pathol 2017; 63:149-156. [DOI: 10.1016/j.humpath.2017.03.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/05/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022]
|
475
|
Ball MW, Singer EA, Srinivasan R. Renal cell carcinoma: molecular characterization and evolving treatment paradigms. Curr Opin Oncol 2017; 29:201-209. [PMID: 28252459 PMCID: PMC5581274 DOI: 10.1097/cco.0000000000000364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW The treatment landscape of advanced renal cell carcinoma (RCC) continues to shift as both new targeted therapies and immunotherapies show efficacy in treating the disease. Contemporary insights into the molecular characterization of RCC are likely to fuel the development of additional therapies. This review summarizes recent advancements in the biologic characterization of RCC and discusses newly approved therapies and ongoing studies in the treatment of advanced RCC. RECENT FINDINGS The Cancer Genome Atlas has now completed comprehensive molecular characterization of clear cell, papillary, and chromophobe RCC, providing insights into the biology of these entities. Two new 'targeted' therapies, cabozantinib and lenvatinib, as well as a novel immune checkpoint inhibitor, the programed death 1 inhibitor nivolumab, have recently been approved for the treatment of metastatic RCC. Although some of these newer therapies are associated with prolongation of survival, there are few long-term responders and the quest for more durable treatment strategies continues. SUMMARY The addition of several new agents effective in metastatic RCC has resulted in improvements in overall survival; however, there are few avenues to durable responses or cure. Ongoing studies as well advances in our understanding of the molecular alterations underlying distinct forms of RCC promise further therapeutic advances and have the potential to alter the current treatment paradigm.
Collapse
Affiliation(s)
- Mark W. Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
476
|
Lee CH, Gundem G, Lee W, Chen YB, Cross JR, Dong Y, Redzematovic A, Mano R, Wei EY, Cheng EH, Srinivasan R, Oschwald D, Hakimi AA, Dunphy MP, Linehan WM, Papaemmanuil E, Hsieh JJ. Persistent Severe Hyperlactatemia and Metabolic Derangement in Lethal SDHB-Mutated Metastatic Kidney Cancer: Clinical Challenges and Examples of Extreme Warburg Effect. JCO Precis Oncol 2017; 1:PO.16.00007. [PMID: 35172488 PMCID: PMC9797236 DOI: 10.1200/po.16.00007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
To describe the unique clinical features, determine the genomics, and investigate the metabolic derangement of an extremely rare form of a hereditary lethal kidney cancer syndrome. Patients and Methods Three patients with lethal kidney cancer (age 19, 20, and 37 years) exhibiting persistent (1 to 3 months) extremely high levels of blood lactate (> 5 mM) despite normal oxygen perfusion, highly avid tumors on [18F]fluorodeoxyglucose positron emission tomography (PET), and pleomorphic histopathologic features were identified and treated in a single institute. Integrated studies including whole-genome sequencing (WGS), targeted sequencing, immunohistochemistry, cell-based assays, and 18F-glutamine PET imaging were performed to investigate this rare kidney cancer syndrome. Results All three patients with kidney cancer were initially given various diagnoses as a result of diverse tumor histopathology and atypical clinical presentations. The correct diagnoses of these SDHB-mutated renal cell carcinomas were first made based on cancer genomics. Genomic studies of the blood and tumors of these patients identified three different kinds of germline loss-of-function mutations in the SDHB gene and the common loss of heterozygosity in the remaining SDHB allele thorough somatic chromosome 1p deletion. In one patient, WGS revealed that a germline mutation of SDHB coupled with loss of heterozygosity was the sole genetic event. Cancer evolution analysis of SDHB tumors based on WGS demonstrated that SDHB in kidney epithelium fulfills the Knudson two-hit criteria as a major tumor suppressor gene. SDHB -/- tumor cells displayed increase in glucose uptake and lactate production, alteration in mitochondrial architecture, and defect in oxidative respiration. 18F-Glutamine PET imaging studies demonstrated increased glutamine metabolism. Conclusion SDHB-deficient metastatic renal cell carcinoma is a rare, aggressive form of kidney cancer that manifests with clinical evidence of a severe Warburg effect, and genomic studies demonstrated two genetic hits at SDHB genes during kidney tumorigenesis.
Collapse
Affiliation(s)
- Chung-Han Lee
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Gunes Gundem
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - William Lee
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Ying-Bei Chen
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Justin R. Cross
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Yiyu Dong
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Almedina Redzematovic
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Roy Mano
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Elizabeth Y. Wei
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Emily H. Cheng
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Ramaprasad Srinivasan
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Dayna Oschwald
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - A. Ari Hakimi
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Mark P. Dunphy
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - W. Marston Linehan
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - Elli Papaemmanuil
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| | - James J. Hsieh
- Chung-Han Lee, Gunes Gundem, William
Lee, Ying-Bei Chen, Justin R. Cross,
Yiyu Dong, Almedina Redzematovic, Roy
Mano, Elizabeth Y. Wei, Emily H. Cheng,
A. Ari Hakimi, Mark P. Dunphy, and Elli
Papaemmanuil, Memorial Sloan Kettering Cancer Center; Dayna
Oschwald, New York Genome Center, New York, NY; James J.
Hsieh, Washington University School of Medicine, St Louis, MO; and
Ramaprasad Srinivasan and W. Marston Linehan,
National Cancer Institute, Bethesda, MD
| |
Collapse
|
477
|
Hsieh JJ, Manley BJ, Khan N, Gao J, Carlo MI, Cheng EH. Overcome tumor heterogeneity-imposed therapeutic barriers through convergent genomic biomarker discovery: A braided cancer river model of kidney cancer. Semin Cell Dev Biol 2017; 64:98-106. [PMID: 27615548 PMCID: PMC5522717 DOI: 10.1016/j.semcdb.2016.09.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
Tumor heterogeneity, encompassing genetic, epigenetic, and microenvironmental variables, is extremely complex and presents challenges to cancer diagnosis and therapy. Genomic efforts on genetic intratumor heterogeneity (G-ITH) confirm branched evolution, support the trunk-branch cancer model, and present a seemingly insurmountable obstacle to conquering cancers. G-ITH is conspicuous in clear cell renal cell carcinoma (ccRCC), where its presence complicates identification and validation of biomarkers and thwarts efforts in advancing precision cancer therapeutics. However, long-term clinical benefits on targeted therapy are not uncommon in metastatic ccRCC patients, implicating that there are underlying constraints during ccRCC evolution, which in turn force a nonrandom sequence of parallel gene/pathway/function/phenotype convergence within individual tumors. Accordingly, we proposed a "braided cancer river model" depicting ccRCC evolution, which deduces cancer development based on multiregion tumor genomics of exceptional mTOR inhibitor (mTORi) responders. Furthermore, we employ an outlier case to explore the river model and highlight the importance of "Five NGS Matters: Number, Frequency, Position, Site and Time" in assessing cancer genomics for precision medicine. This mutable cancer river model may capture clinically significant phenotype-convergent events, predict vulnerability/resistance mechanisms, and guide effective therapeutic strategies. Our model originates from studying exceptional responders in ccRCC, which warrants further refinement and future validation concerning its applicability to other cancer types. The goal of this review is employing kidney cancer as an example to illustrate critical issues concerning tumor heterogeneity.
Collapse
Affiliation(s)
- James J Hsieh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Brandon J Manley
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Nabeela Khan
- Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY11203, United States
| | - JianJiong Gao
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
478
|
Effect of Metformin Use on Survival Outcomes in Patients With Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2017; 15:221-229. [DOI: 10.1016/j.clgc.2016.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/17/2016] [Accepted: 06/19/2016] [Indexed: 01/05/2023]
|
479
|
de Velasco G, McKay RR, Lin X, Moreira RB, Simantov R, Choueiri TK. Comprehensive Analysis of Survival Outcomes in Non-Clear Cell Renal Cell Carcinoma Patients Treated in Clinical Trials. Clin Genitourin Cancer 2017; 15:652-660.e1. [PMID: 28410911 DOI: 10.1016/j.clgc.2017.03.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND Clinical data from patients with non-clear cell renal cell carcinoma (nccRCC) receiving targeted therapy are limited, and many clinical trials have excluded these patients from study entry. We sought to investigate the outcomes of patients with nccRCC treated in clinical trials in the modern era compared with the outcomes of patients with clear cell RCC (ccRCC). PATIENTS AND METHODS We conducted a retrospective study of patients with metastatic RCC who had received targeted therapy in Pfizer-sponsored phase II and III clinical trials from 2003 to 2013. Associations between the histologic type and treatment outcome (overall survival [OS] and progression-free survival [PFS]) were assessed using the log-rank test on univariate analysis or the Wald χ2 test from Cox regression on multivariable analysis, adjusted for baseline characteristics, including age, sex, Eastern Cooperative Oncology Group performance status, body mass index, International Metastatic RCC Database Consortium risk factors, previous nephrectomy, previous therapy, metastatic sites, angiotensin system inhibitor use, and statin use. RESULTS We identified 4527 patients with metastatic RCC: 4235 with ccRCC and 337 with nccRCC. Overall, the median OS was shorter for those with nccRCC than for those with ccRCC (15.7 vs. 20.2 months; hazard ratio [HR], 1.41; 95% confidence interval 1.22-1.63; P < .001). When stratified by the International Metastatic RCC Database Consortium risk group, the median OS was inferior for the intermediate- and poor-risk patients with nccRCC than for those with ccRCC. However, no differences were found in the favorable risk group for nccRCC versus ccRCC. The patients with nccRCC who had received vascular endothelial growth factor-targeted therapy had shorter PFS compared with that of ccRCC patients (median, 6.1 vs. 8.5 months; HR, 1.49; P < .001) but similar PFS when treated with mammalian target of rapamycin inhibitors (median, 4.3 vs. 4.4 months; HR, 0.92; P = .63). CONCLUSION Our findings have confirmed that patients with nccRCC are underrepresented in clinical trials and highlight the need for further prospective studies exploring current and novel agents for this patient population.
Collapse
Affiliation(s)
- Guillermo de Velasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Rana R McKay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Xun Lin
- Pfizer Oncology, Pfizer Inc, La Jolla, CA
| | - Raphel B Moreira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
| |
Collapse
|
480
|
Farshidfar F, Zheng S, Gingras MC, Newton Y, Shih J, Robertson AG, Hinoue T, Hoadley KA, Gibb EA, Roszik J, Covington KR, Wu CC, Shinbrot E, Stransky N, Hegde A, Yang JD, Reznik E, Sadeghi S, Pedamallu CS, Ojesina AI, Hess JM, Auman JT, Rhie SK, Bowlby R, Borad MJ, Zhu AX, Stuart JM, Sander C, Akbani R, Cherniack AD, Deshpande V, Mounajjed T, Foo WC, Torbenson MS, Kleiner DE, Laird PW, Wheeler DA, McRee AJ, Bathe OF, Andersen JB, Bardeesy N, Roberts LR, Kwong LN. Integrative Genomic Analysis of Cholangiocarcinoma Identifies Distinct IDH-Mutant Molecular Profiles. Cell Rep 2017; 18:2780-2794. [PMID: 28297679 PMCID: PMC5493145 DOI: 10.1016/j.celrep.2017.02.033] [Citation(s) in RCA: 351] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 01/04/2017] [Accepted: 02/09/2017] [Indexed: 02/07/2023] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy of the bile ducts, with poor prognosis and limited treatment options. Here, we describe the integrated analysis of somatic mutations, RNA expression, copy number, and DNA methylation by The Cancer Genome Atlas of a set of predominantly intrahepatic CCA cases and propose a molecular classification scheme. We identified an IDH mutant-enriched subtype with distinct molecular features including low expression of chromatin modifiers, elevated expression of mitochondrial genes, and increased mitochondrial DNA copy number. Leveraging the multi-platform data, we observed that ARID1A exhibited DNA hypermethylation and decreased expression in the IDH mutant subtype. More broadly, we found that IDH mutations are associated with an expanded histological spectrum of liver tumors with molecular features that stratify with CCA. Our studies reveal insights into the molecular pathogenesis and heterogeneity of cholangiocarcinoma and provide classification information of potential therapeutic significance.
Collapse
Affiliation(s)
- Farshad Farshidfar
- Departments of Surgery and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Siyuan Zheng
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marie-Claude Gingras
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yulia Newton
- University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Juliann Shih
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Toshinori Hinoue
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Katherine A Hoadley
- Departments of Genetics and Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ewan A Gibb
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Jason Roszik
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyle R Covington
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chia-Chin Wu
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eve Shinbrot
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Apurva Hegde
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ju Dong Yang
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Ed Reznik
- Memorial Sloan Kettering Cancer Center, New York, NY 10005, USA
| | - Sara Sadeghi
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Chandra Sekhar Pedamallu
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Akinyemi I Ojesina
- University of Alabama at Birmingham, Birmingham, AL 35294, USA; HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Julian M Hess
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - J Todd Auman
- Departments of Genetics and Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Suhn K Rhie
- USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Reanne Bowlby
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC V5Z 4S6, Canada
| | - Mitesh J Borad
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ 85054, USA
| | - Andrew X Zhu
- Departments of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Josh M Stuart
- University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Chris Sander
- Memorial Sloan Kettering Cancer Center, New York, NY 10005, USA
| | - Rehan Akbani
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew D Cherniack
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Vikram Deshpande
- Departments of Pathology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Taofic Mounajjed
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Wai Chin Foo
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S Torbenson
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Autumn J McRee
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Oliver F Bathe
- Departments of Surgery and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, Department of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| | - Nabeel Bardeesy
- Departments of Pathology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Lewis R Roberts
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | - Lawrence N Kwong
- Departments of Genomic Medicine, Melanoma Medical Oncology, Bioinformatics and Computational Biology, Pathology, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
481
|
Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers 2017; 3:17009. [PMID: 28276433 PMCID: PMC5936048 DOI: 10.1038/nrdp.2017.9] [Citation(s) in RCA: 1614] [Impact Index Per Article: 230.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC) denotes cancer originated from the renal epithelium and accounts for >90% of cancers in the kidney. The disease encompasses >10 histological and molecular subtypes, of which clear cell RCC (ccRCC) is most common and accounts for most cancer-related deaths. Although somatic VHL mutations have been described for some time, more-recent cancer genomic studies have identified mutations in epigenetic regulatory genes and demonstrated marked intra-tumour heterogeneity, which could have prognostic, predictive and therapeutic relevance. Localized RCC can be successfully managed with surgery, whereas metastatic RCC is refractory to conventional chemotherapy. However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Since 2015, agents with additional targets aside from VEGFR have been approved, such as cabozantinib and lenvatinib; immunotherapies, such as nivolumab, have also been added to the armamentarium for metastatic RCC. Here, we provide an overview of the biology of RCC, with a focus on ccRCC, as well as updates to complement the current clinical guidelines and an outline of potential future directions for RCC research and therapy.
Collapse
Affiliation(s)
- James J. Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8069, St. Louis, Missouri, USA
| | - Mark P. Purdue
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Charles Swanton
- Francis Crick Institute, UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, UK
| | - Laurence Albiges
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Manuela Schmidinger
- Department of Medicine I, Clinical Division of Oncology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daniel Y. Heng
- Department of Medical Oncolgy, Tom Baker Cancer Center, Calgary, Alberta, Canada
| | - James Larkin
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Vincenzo Ficarra
- Department of Experimental and Clinical Medical Sciences - Urologic Clinic, University of Udine, Italy
| |
Collapse
|
482
|
Kasoji SK, Chang EH, Mullin LB, Chong WK, Rathmell WK, Dayton PA. A Pilot Clinical Study in Characterization of Malignant Renal-cell Carcinoma Subtype with Contrast-enhanced Ultrasound. ULTRASONIC IMAGING 2017; 39:126-136. [PMID: 27659687 PMCID: PMC5599099 DOI: 10.1177/0161734616666383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Malignant renal cell carcinoma (RCC) is a diverse set of diseases, which are independently difficult to characterize using conventional MRI and CT protocols due to low temporal resolution to study perfusion characteristics. Because different disease subtypes have different prognoses and involve varying treatment regimens, the ability to determine RCC subtype non-invasively is a clinical need. Contrast-enhanced ultrasound (CEUS) has been assessed as a tool to characterize kidney lesions based on qualitative and quantitative assessment of perfusion patterns, and we hypothesize that this technique might help differentiate disease subtypes. Twelve patients with RCC confirmed pathologically were imaged using contrast-enhanced ultrasound. Time intensity curves were generated and analyzed quantitatively using 10 characteristic metrics. Results showed that peak intensity ( p = 0.001) and time-to-80% on wash-out ( p = 0.004) provided significant differences between clear cell, papillary, and chromophobe RCC subtypes. These results suggest that CEUS may be a feasible test for characterizing RCC subtypes.
Collapse
Affiliation(s)
- Sandeep K. Kasoji
- Joint Dept. of Biomedical Engineering, UNC Chapel Hill/North Carolina State University, 333 S. Columbia St, Chapel Hill, NC 27517 USA/911 Oval Dr, Raleigh, NC 27606 USA
| | - Emily H. Chang
- UNC Kidney Center, 7024 Burnett-Womack CB # 7155, Chapel Hill, NC 27599 USA
| | - Lee B. Mullin
- Joint Dept. of Biomedical Engineering, UNC Chapel Hill/North Carolina State University, 333 S. Columbia St, Chapel Hill, NC 27517 USA/911 Oval Dr, Raleigh, NC 27606 USA
| | - Wui K. Chong
- UNC Hospitals Dept. of Radiology, 101 Manning Dr #2, Chapel Hill, NC 27514 USA
| | - W. Kimryn Rathmell
- UNC Hospitals Dept. of Radiology, 101 Manning Dr #2, Chapel Hill, NC 27514 USA
- Vanderbilt University Hospitals Dept. of Medicine/Hematology & Oncology, 2200 Pierce Ave, Nashville, TN 37232 USA
| | - Paul A. Dayton
- Joint Dept. of Biomedical Engineering, UNC Chapel Hill/North Carolina State University, 333 S. Columbia St, Chapel Hill, NC 27517 USA/911 Oval Dr, Raleigh, NC 27606 USA
- UNC Hospitals Dept. of Radiology, 101 Manning Dr #2, Chapel Hill, NC 27514 USA
- Biomedical Research Imaging Center, 125 Mason Farm Road, Chapel Hill, NC 27599 USA
| |
Collapse
|
483
|
Li S, Shuch BM, Gerstein MB. Whole-genome analysis of papillary kidney cancer finds significant noncoding alterations. PLoS Genet 2017; 13:e1006685. [PMID: 28358873 PMCID: PMC5391127 DOI: 10.1371/journal.pgen.1006685] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/13/2017] [Accepted: 03/13/2017] [Indexed: 01/30/2023] Open
Abstract
To date, studies on papillary renal-cell carcinoma (pRCC) have largely focused on coding alterations in traditional drivers, particularly the tyrosine-kinase, Met. However, for a significant fraction of tumors, researchers have been unable to determine a clear molecular etiology. To address this, we perform the first whole-genome analysis of pRCC. Elaborating on previous results on MET, we find a germline SNP (rs11762213) in this gene predicting prognosis. Surprisingly, we detect no enrichment for small structural variants disrupting MET. Next, we scrutinize noncoding mutations, discovering potentially impactful ones associated with MET. Many of these are in an intron connected to a known, oncogenic alternative-splicing event; moreover, we find methylation dysregulation nearby, leading to a cryptic promoter activation. We also notice an elevation of mutations in the long noncoding RNA NEAT1, and these mutations are associated with increased expression and unfavorable outcome. Finally, to address the origin of pRCC heterogeneity, we carry out whole-genome analyses of mutational processes. First, we investigate genome-wide mutational patterns, finding they are governed mostly by methylation-associated C-to-T transitions. We also observe significantly more mutations in open chromatin and early-replicating regions in tumors with chromatin-modifier alterations. Finally, we reconstruct cancer-evolutionary trees, which have markedly different topologies and suggested evolutionary trajectories for the different subtypes of pRCC.
Collapse
Affiliation(s)
- Shantao Li
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Brian M. Shuch
- Department of Urology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Mark B. Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, United States of America
- Department of Computer Science, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
484
|
Fernández-Pello S, Hofmann F, Tahbaz R, Marconi L, Lam TB, Albiges L, Bensalah K, Canfield SE, Dabestani S, Giles RH, Hora M, Kuczyk MA, Merseburger AS, Powles T, Staehler M, Volpe A, Ljungberg B, Bex A. A Systematic Review and Meta-analysis Comparing the Effectiveness and Adverse Effects of Different Systemic Treatments for Non-clear Cell Renal Cell Carcinoma. Eur Urol 2017; 71:426-436. [DOI: 10.1016/j.eururo.2016.11.020] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 11/24/2022]
|
485
|
Udager AM, Mehra R. Morphologic, Molecular, and Taxonomic Evolution of Renal Cell Carcinoma: A Conceptual Perspective With Emphasis on Updates to the 2016 World Health Organization Classification. Arch Pathol Lab Med 2017; 140:1026-37. [PMID: 27684973 DOI: 10.5858/arpa.2016-0218-ra] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Molecular and morphologic interrogation has driven a much-needed reexamination of renal cell carcinoma (RCC). Indeed, the recently released 2016 World Health Organization classification now recognizes 12 distinct RCC subtypes, as well as several other emerging/provisional RCC entities. From a clinical perspective, accurate RCC classification may have important implications for patients and their families, including prognostic risk stratification, targeted therapeutics selection, and identification for genetic testing. In this review, we provide a conceptual framework for approaching RCC diagnosis and classification by categorizing RCCs as tumors with clear cytoplasm, papillary architecture, and eosinophilic (oncocytic) cytoplasm. The currently recognized 2016 World Health Organization classification for RCC subtypes is briefly discussed, including new diagnostic entities (clear cell papillary RCC, hereditary leiomyomatosis and RCC-associated RCC, succinate dehydrogenase-deficient RCC, tubulocystic RCC, and acquired cystic disease-associated RCC) and areas of evolving RCC classification, such as transcription elongation factor B subunit 1 (TCEB1)-mutated RCC/RCC with angioleiomyoma-like stroma/RCC with leiomyomatous stroma, RCC associated with anaplastic lymphoma receptor tyrosine kinase (ALK) gene rearrangement, thyroidlike follicular RCC, and RCC in neuroblastoma survivors. For each RCC subtype, relevant clinical, molecular, gross, and microscopic findings are reviewed, and ancillary studies helpful for its differential diagnosis are presented, providing a practical approach to modern RCC classification.
Collapse
Affiliation(s)
- Aaron M Udager
- From the Department of Pathology (Drs Udager and Mehra) and the Comprehensive Cancer Center (Dr Mehra), University of Michigan Health System, Ann Arbor; and the Michigan Center for Translational Pathology, Ann Arbor (Dr Mehra)
| | | |
Collapse
|
486
|
Long C, Jian J, Li X, Wang G, Wang J. A comprehensive analysis of cancer-driving mutations and genes in kidney cancer. Oncol Lett 2017; 13:2151-2160. [PMID: 28454375 PMCID: PMC5403472 DOI: 10.3892/ol.2017.5689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/09/2016] [Indexed: 02/06/2023] Open
Abstract
An accumulation of driver mutations is important for cancer formation and progression, and leads to the disruption of genes and signaling pathways. The identification of driver mutations and genes has been the subject of numerous previous studies. The present study was performed to identify cancer-driving mutations and genes in renal cell carcinoma (RCC), prioritizing noncoding variants with a high functional impact, in order to analyze the most informative features. Sorting Intolerant From Tolerant (SIFT), Polymorphism Phenotyping version 2 (Polyphen2) and MutationAssessor were applied to predict deleterious mutations in the coding genome. OncodriveFM and OncodriveCLUST were used to detect potential driver genes and signaling pathways. The functional impact of noncoding variants was evaluated using Combined Annotation Dependent Depletion, FunSeq2 and Genome-Wide Annotation of Variants. Noncoding features were analyzed with respect to their enrichment of high-scoring variants. A total of 1,327 coding mutations in clear cell RCC, 258 in chromophobe RCC and 1,186 in papillary RCC were predicted to be deleterious by all three of MutationAssessor, Polyphen2 and SIFT. In total, 77 genes were positively selected by OncodriveFM and 1 by OncodriveCLUST, 45 of which were recurrently mutated genes. In addition, 10 signaling pathways were recurrently mutated and had a high functional impact bias (FM bias), and 31 novel signaling pathways with high FM bias were identified. Furthermore, noncoding regulatory features and conserved regions contained numerous high-scoring variants, and expression, replication time, GC content and recombination rate were positively correlated with the densities of high-scoring variants. In conclusion, the present study identified a list of cancer-driving genes and signaling pathways, features like regulatory elements, conserved regions, replication time, expression, GC content and recombination rate are major factors that affect the distribution of high-scoring non-coding mutations in kidney cancer.
Collapse
Affiliation(s)
- Chengmei Long
- Department of Organ Transplantation, Jiangxi Provincial People's Hospital, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jinbo Jian
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Xinchang Li
- Department of Organ Transplantation, Jiangxi Provincial People's Hospital, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jingen Wang
- Department of Urology, Jiangxi Provincial People's Hospital, School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
487
|
Kamps R, Brandão RD, Bosch BJVD, Paulussen ADC, Xanthoulea S, Blok MJ, Romano A. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int J Mol Sci 2017; 18:ijms18020308. [PMID: 28146134 PMCID: PMC5343844 DOI: 10.3390/ijms18020308] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022] Open
Abstract
Next-generation sequencing (NGS) technology has expanded in the last decades with significant improvements in the reliability, sequencing chemistry, pipeline analyses, data interpretation and costs. Such advances make the use of NGS feasible in clinical practice today. This review describes the recent technological developments in NGS applied to the field of oncology. A number of clinical applications are reviewed, i.e., mutation detection in inherited cancer syndromes based on DNA-sequencing, detection of spliceogenic variants based on RNA-sequencing, DNA-sequencing to identify risk modifiers and application for pre-implantation genetic diagnosis, cancer somatic mutation analysis, pharmacogenetics and liquid biopsy. Conclusive remarks, clinical limitations, implications and ethical considerations that relate to the different applications are provided.
Collapse
Affiliation(s)
- Rick Kamps
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Rita D Brandão
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Bianca J van den Bosch
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Aimee D C Paulussen
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Sofia Xanthoulea
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Marinus J Blok
- Department of Clinical Genetics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| | - Andrea Romano
- Department of Gynaecology and Obstetrics: GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, 6229HX Maastricht, The Netherlands.
| |
Collapse
|
488
|
Barthel FP, Wei W, Tang M, Martinez-Ledesma E, Hu X, Amin SB, Akdemir KC, Seth S, Song X, Wang Q, Lichtenberg T, Hu J, Zhang J, Zheng S, Verhaak RGW. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat Genet 2017; 49:349-357. [PMID: 28135248 DOI: 10.1038/ng.3781] [Citation(s) in RCA: 419] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 01/04/2017] [Indexed: 12/13/2022]
Abstract
Cancer cells survive cellular crisis through telomere maintenance mechanisms. We report telomere lengths in 18,430 samples, including tumors and non-neoplastic samples, across 31 cancer types. Telomeres were shorter in tumors than in normal tissues and longer in sarcomas and gliomas than in other cancers. Among 6,835 cancers, 73% expressed telomerase reverse transcriptase (TERT), which was associated with TERT point mutations, rearrangements, DNA amplifications and transcript fusions and predictive of telomerase activity. TERT promoter methylation provided an additional deregulatory TERT expression mechanism. Five percent of cases, characterized by undetectable TERT expression and alterations in ATRX or DAXX, demonstrated elongated telomeres and increased telomeric repeat-containing RNA (TERRA). The remaining 22% of tumors neither expressed TERT nor harbored alterations in ATRX or DAXX. In this group, telomere length positively correlated with TP53 and RB1 mutations. Our analysis integrates TERT abnormalities, telomerase activity and genomic alterations with telomere length in cancer.
Collapse
Affiliation(s)
- Floris P Barthel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,Oncology Graduate School Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.,Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Wei
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ming Tang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emmanuel Martinez-Ledesma
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Neuro-Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xin Hu
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Program in Biostatistics, Bioinformatics, and Systems Biology, the University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| | - Samirkumar B Amin
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Graduate Program in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Kadir C Akdemir
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sahil Seth
- Institute for Applied Cancer Science, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xingzhi Song
- Institute for Applied Cancer Science, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qianghu Wang
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tara Lichtenberg
- Biopathology Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jian Hu
- Department of Cancer Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jianhua Zhang
- Institute for Applied Cancer Science, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Siyuan Zheng
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Neuro-Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roel G W Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
489
|
Affiliation(s)
- Toni K Choueiri
- From the Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston (T.K.C.); and the Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York (R.J.M.)
| | - Robert J Motzer
- From the Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston (T.K.C.); and the Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York (R.J.M.)
| |
Collapse
|
490
|
Abbosh P, Sundararajan S, Millis SZ, Hauben A, Reddy S, Geynisman DM, Uzzo R. Molecular and Genomic Profiling to Identify Actionable Targets in Chromophobe Renal Cell Cancer. Eur Urol Focus 2017; 4:969-971. [PMID: 28753842 DOI: 10.1016/j.euf.2017.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 01/09/2017] [Indexed: 10/20/2022]
Abstract
Metastatic chromophobe renal cell cancer (chRCC) is a rare subtype of RCC with no standard treatment. We performed molecular profiling of 12 chRCC cases to identify alterations predictive of response to therapy. Tests included immunohistochemistry assays, fluorescence in situ hybridization, and next-generation sequencing. Analysis identified c-KIT overexpression in 6/9 (67%) samples analyzed, and loss of protein expression of RRM1 and MGMT in 11/12 (92%) and of PTEN in 7/12 samples (58%). Mutations of TP53, PTEN, APC, and VHL genes were identified. In summary, molecular profiling of chRCC identified alterations in genes and protein expression that might provide a mechanistic rationale for off-label use of approved therapies in advanced chRCC, and could guide the design of molecularly targeted clinical trials. PATIENT SUMMARY: In this study, we evaluated samples of a rare type of kidney cancer (chromophobe type) and identified potential genetic markers that could be used to individualize treatment and possibly improve treatment outcomes.
Collapse
Affiliation(s)
- Philip Abbosh
- Division of Urological Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Srinath Sundararajan
- Division of Hematology and Oncology, Department of Medicine, University of Arizona, Tuscon, AZ, USA.
| | | | | | | | - Daniel M Geynisman
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Robert Uzzo
- Division of Urological Oncology, Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
491
|
Reznik E, Wang Q, La K, Schultz N, Sander C. Mitochondrial respiratory gene expression is suppressed in many cancers. eLife 2017; 6:e21592. [PMID: 28099114 PMCID: PMC5243113 DOI: 10.7554/elife.21592] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/09/2016] [Indexed: 12/18/2022] Open
Abstract
The fundamental metabolic decision of a cell, the balance between respiration and fermentation, rests in part on expression of the mitochondrial genome (mtDNA) and coordination with expression of the nuclear genome (nuDNA). Previously we described mtDNA copy number depletion across many solid tumor types (Reznik et al., 2016). Here, we use orthogonal RNA-sequencing data to quantify mtDNA expression (mtRNA), and report analogously lower expression of mtRNA in tumors (relative to normal tissue) across a majority of cancer types. Several cancers exhibit a trio of mutually consistent evidence suggesting a drop in respiratory activity: depletion of mtDNA copy number, decreases in mtRNA levels, and decreases in expression of nuDNA-encoded respiratory proteins. Intriguingly, a minority of cancer types exhibit a drop in mtDNA expression but an increase in nuDNA expression of respiratory proteins, with unknown implications for respiratory activity. Our results indicate suppression of respiratory gene expression across many cancer types.
Collapse
Affiliation(s)
- Ed Reznik
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Qingguo Wang
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Konnor La
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Nikolaus Schultz
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Chris Sander
- Department of Cell Biology, Harvard Medical School, Boston, United States
- cBio Center, Dana-Farber Cancer Institute, Boston, United States
| |
Collapse
|
492
|
Weyandt JD, Thompson CB, Giaccia AJ, Rathmell WK. Metabolic Alterations in Cancer and Their Potential as Therapeutic Targets. Am Soc Clin Oncol Educ Book 2017; 37:825-832. [PMID: 28561705 PMCID: PMC5954416 DOI: 10.1200/edbk_175561] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Otto Warburg's discovery in the 1920s that tumor cells took up more glucose and produced more lactate than normal cells provided the first clues that cancer cells reprogrammed their metabolism. For many years, however, it was unclear as to whether these metabolic alterations were a consequence of tumor growth or an adaptation that provided a survival advantage to these cells. In more recent years, interest in the metabolic differences in cancer cells has surged, as tumor proliferation and survival have been shown to be dependent upon these metabolic changes. In this educational review, we discuss some of the mechanisms that tumor cells use for reprogramming their metabolism to provide the energy and nutrients that they need for quick or sustained proliferation and discuss the potential for therapeutic targeting of these pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Jamie D. Weyandt
- Department of Medicine, Division of Hemtology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Craig B. Thompson
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, 10065
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305
| | - W. Kimryn Rathmell
- Department of Medicine, Division of Hemtology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
493
|
Epidemiology of Renal Cell Carcinoma and Its Predisposing Risk Factors. Urol Oncol 2017. [DOI: 10.1007/978-3-319-42603-7_55-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
494
|
|
495
|
Abstract
The majority of kidney cancers are associated with mutations in the von Hippel-Lindau gene and a small proportion are associated with infrequent mutations in other well characterized tumour-suppressor genes. In the past 15 years, efforts to uncover other key genes involved in renal cancer have identified many genes that are dysregulated or silenced via epigenetic mechanisms, mainly through methylation of promoter CpG islands or dysregulation of specific microRNAs. In addition, the advent of next-generation sequencing has led to the identification of several novel genes that are mutated in renal cancer, such as PBRM1, BAP1 and SETD2, which are all involved in histone modification and nucleosome and chromatin remodelling. In this Review, we discuss how altered DNA methylation, microRNA dysregulation and mutations in histone-modifying enzymes disrupt cellular pathways in renal cancers.
Collapse
Affiliation(s)
- Mark R Morris
- Brain Tumour Research Centre, Wolverhampton School of Sciences, University of Wolverhampton, Wulfruna Street, Wolverhampton WV1 1LY, UK
| | - Farida Latif
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
496
|
Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nat Genet 2016; 49:65-74. [PMID: 27869826 DOI: 10.1038/ng.3722] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
Abstract
Extensive prior research focused on somatic copy-number alterations (SCNAs) affecting cancer genes, yet the extent to which recurrent SCNAs exert their influence through rearrangement of cis-regulatory elements (CREs) remains unclear. Here we present a framework for inferring cancer-related gene overexpression resulting from CRE reorganization (e.g., enhancer hijacking) by integrating SCNAs, gene expression data and information on topologically associating domains (TADs). Analysis of 7,416 cancer genomes uncovered several pan-cancer candidate genes, including IRS4, SMARCA1 and TERT. We demonstrate that IRS4 overexpression in lung cancer is associated with recurrent deletions in cis, and we present evidence supporting a tumor-promoting role. We additionally pursued cancer-type-specific analyses and uncovered IGF2 as a target for enhancer hijacking in colorectal cancer. Recurrent tandem duplications intersecting with a TAD boundary mediate de novo formation of a 3D contact domain comprising IGF2 and a lineage-specific super-enhancer, resulting in high-level gene activation. Our framework enables systematic inference of CRE rearrangements mediating dysregulation in cancer.
Collapse
|
497
|
Global analysis of somatic structural genomic alterations and their impact on gene expression in diverse human cancers. Proc Natl Acad Sci U S A 2016; 113:13768-13773. [PMID: 27856756 DOI: 10.1073/pnas.1606220113] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tumor genomes are mosaics of somatic structural variants (SVs) that may contribute to the activation of oncogenes or inactivation of tumor suppressors, for example, by altering gene copy number amplitude. However, there are multiple other ways in which SVs can modulate transcription, but the general impact of such events on tumor transcriptional output has not been systematically determined. Here we use whole-genome sequencing data to map SVs across 600 tumors and 18 cancers, and investigate the relationship between SVs, copy number alterations (CNAs), and mRNA expression. We find that 34% of CNA breakpoints can be clarified structurally and that most amplifications are due to tandem duplications. We observe frequent swapping of strong and weak promoters in the context of gene fusions, and find that this has a measurable global impact on mRNA levels. Interestingly, several long noncoding RNAs were strongly activated by this mechanism. Additionally, SVs were confirmed in telomere reverse transcriptase (TERT) upstream regions in several cancers, associated with elevated TERT mRNA levels. We also highlight high-confidence gene fusions supported by both genomic and transcriptomic evidence, including a previously undescribed paired box 8 (PAX8)-nuclear factor, erythroid 2 like 2 (NFE2L2) fusion in thyroid carcinoma. In summary, we combine SV, CNA, and expression data to provide insights into the structural basis of CNAs as well as the impact of SVs on gene expression in tumors.
Collapse
|
498
|
Haake SM, Rathmell WK. Renal cancer subtypes: Should we be lumping or splitting for therapeutic decision making? Cancer 2016; 123:200-209. [PMID: 27861752 DOI: 10.1002/cncr.30314] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 01/05/2023]
Abstract
The treatment of advanced renal cell carcinoma has posed a challenge for decades, in part because of common themes related to intrinsic resistance to cytotoxic chemotherapy and the obscure biology of these cancer types. Forward movement in the treatment of the renal cell carcinomas thus can be approached in 2 ways: by splitting the tumor types along histologic and molecular features, in the hopes of coupling highly precision-focused therapy on a subset of patients who have disease with the most potential for benefit; or by lumping the various biologies and histologies together, to include the rarer renal cell carcinoma types with the more common types. The former strategy satisfies the desire for customized precision in treatment delivery, whereas the latter strategy allows clinicians to offer a wider therapeutic menu in a set of diseases we are continuing to learn about on a physiologic and molecular level. Cancer 2017;123:200-209. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Scott M Haake
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - W Kimryn Rathmell
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
499
|
Fendler A, Stephan C, Yousef GM, Kristiansen G, Jung K. The translational potential of microRNAs as biofluid markers of urological tumours. Nat Rev Urol 2016; 13:734-752. [DOI: 10.1038/nrurol.2016.193] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
500
|
Grampp S, Platt JL, Lauer V, Salama R, Kranz F, Neumann VK, Wach S, Stöhr C, Hartmann A, Eckardt KU, Ratcliffe PJ, Mole DR, Schödel J. Genetic variation at the 8q24.21 renal cancer susceptibility locus affects HIF binding to a MYC enhancer. Nat Commun 2016; 7:13183. [PMID: 27774982 PMCID: PMC5079059 DOI: 10.1038/ncomms13183] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 09/08/2016] [Indexed: 12/26/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by loss of function of the von Hippel-Lindau tumour suppressor (VHL) and unrestrained activation of hypoxia-inducible transcription factors (HIFs). Genetic and epigenetic determinants have an impact on HIF pathways. A recent genome-wide association study on renal cancer susceptibility identified single-nucleotide polymorphisms (SNPs) in an intergenic region located between the oncogenes MYC and PVT1. Here using assays of chromatin conformation, allele-specific chromatin immunoprecipitation and genome editing, we show that HIF binding to this regulatory element is necessary to trans-activate MYC and PVT1 expression specifically in cells of renal tubular origins. Moreover, we demonstrate that the risk-associated polymorphisms increase chromatin accessibility and activity as well as HIF binding to the enhancer. These findings provide further evidence that genetic variation at HIF-binding sites modulates the oncogenic transcriptional output of the VHL-HIF axis and provide a functional explanation for the disease-associated effects of SNPs in ccRCC.
Collapse
MESH Headings
- Aryl Hydrocarbon Receptor Nuclear Translocator/genetics
- Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism
- Binding Sites
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Chromatin/chemistry
- Chromatin/metabolism
- Chromatin Immunoprecipitation
- Chromosomes, Human, Pair 8
- DNA, Intergenic/genetics
- DNA, Intergenic/metabolism
- Enhancer Elements, Genetic
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Genome-Wide Association Study
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Polymorphism, Single Nucleotide
- Protein Binding
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Von Hippel-Lindau Tumor Suppressor Protein/metabolism
Collapse
Affiliation(s)
- Steffen Grampp
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| | - James L. Platt
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Victoria Lauer
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| | - Rafik Salama
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Franziska Kranz
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
- Department of Computer Science 9, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Cauerstraße 11, 91058 Erlangen, Germany
| | - Viviana K. Neumann
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| | - Sven Wach
- Department of Urology, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Christine Stöhr
- Institute of Pathology, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Krankenhausstraße 8-10, 91054 Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Krankenhausstraße 8-10, 91054 Erlangen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| | - Peter J. Ratcliffe
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - David R. Mole
- Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| |
Collapse
|