632
Views
27
CrossRef citations to date
0
Altmetric
Review Article

Nucleic acid-based tissue biomarkers of urologic malignancies

, , , , , , & show all
Pages 173-199 | Received 08 Nov 2013, Accepted 14 Mar 2013, Published online: 30 May 2014

References

  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11–30
  • Shen H, Laird PW. Interplay between the cancer genome and epigenome. Cell 2013;153:38–55
  • Aguilera A, Gomez-Gonzalez B. Genome instability: a mechanistic view of its causes and consequences. Nat Rev Genet 2008;9:204–17
  • Bristow RG, Hill RP. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer 2008;8:180–92
  • McGranahan N, Burrell RA, Endesfelder D, et al. Cancer chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep 2012;13:528–38
  • Bakhoum SF, Compton DA. Chromosomal instability and cancer: a complex relationship with therapeutic potential. J Clin Invest 2012;122:1138–43
  • Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–6
  • Wyatt AW, Collins CC. In Brief: chromothripsis and cancer. J Pathol 2013;231:1–3
  • Maher CA, Wilson RK. Chromothripsis and human disease: piecing together the shattering process. Cell 2012;148:29–32
  • Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of prostate cancer genomes. Cell 2013;153:666–77
  • Bouvard V, Baan R, Straif K, et al. A review of human carcinogens – Part B: biological agents. Lancet Oncol 2009;10:321–2
  • Moore PS, Chang Y. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer 2010;10:878–89
  • Bouvard V, Baan R, Straif K, et al. A review of human carcinogens – Part B: biological agents. Lancet Oncol 2009;10:321–2
  • Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 2008;319:1096–100
  • Dalianis T, Hirsch HH. Human polyomaviruses in disease and cancer. Virology 2013;437:63–72
  • Hindorff LA, Sethupathy P, Junkins HA, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci USA 2009;106:9362–7
  • Burns MB, Temiz NA, Harris RS. Evidence for APOBEC3B mutagenesis in multiple human cancers. Nat Genet 2013;45:977–83
  • Roberts SA, Lawrence MS, Klimczak LJ, et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat Genet 2013;45:970–6
  • Nik-Zainal S, Alexandrov LB, Wedge DC, et al. Mutational processes molding the genomes of 21 breast cancers. Cell 2012;149:979–93
  • Kuong KJ, Loeb LA. APOBEC3B mutagenesis in cancer. Nat Genet 2013;45:964–5
  • Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature 2013;500:415–21
  • Narod SA. BRCA mutations in the management of breast cancer: the state of the art. Nat Rev Clin Oncol 2010;7:702–7
  • Lynch HT, Lynch PM, Lanspa SJ, et al. Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet 2009;76:1–18
  • Tuppen HA, Blakely EL, Turnbull DM, Taylor RW. Mitochondrial DNA mutations and human disease. Biochim Biophys Acta 2010;1797:113–28
  • Santos C, Martinez M, Lima M, et al. Mitochondrial DNA mutations in cancer: a review. Curr Top Med Chem 2008;8:1351–66
  • Ellinger J, Muller DC, Muller SC, et al. Circulating mitochondrial DNA in serum: a universal diagnostic biomarker for patients with urological malignancies. Urol Oncol 2012;30:509–15
  • Ellinger J, Albers P, Muller SC, et al. Circulating mitochondrial DNA in the serum of patients with testicular germ cell cancer as a novel noninvasive diagnostic biomarker. BJU Int 2009;104:48–52
  • Fliss MS, Usadel H, Caballero OL, et al. Facile detection of mitochondrial DNA mutations in tumors and bodily fluids. Science 2000;287:2017–9
  • Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 2012;13:484–92
  • Tahiliani M, Koh KP, Shen Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009;324:930–5
  • Ito S, D'Alessio AC, Taranova OV, et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 2010;466:1129–33
  • Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 2011;333:1300–3
  • Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 2009;324:929–30
  • Ono R, Taki T, Taketani T, et al. LCX, leukemia-associated protein with a CXXC domain, is fused to MLL in acute myeloid leukemia with trilineage dysplasia having t(10;11)(q22;q23). Cancer Res 2002;62:4075–80
  • Haffner MC, Chaux A, Meeker AK, et al. Global 5-hydroxymethylcytosine content is significantly reduced in tissue stem/progenitor cell compartments and in human cancers. Oncotarget 2011;2:627–37
  • Li W, Liu M. Distribution of 5-hydroxymethylcytosine in different human tissues. J Nucleic Acids 2011;2011:870726
  • Shock LS, Thakkar PV, Peterson EJ, et al. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proc Natl Acad Sci USA 2011;108:3630–5
  • Pirola CJ, Gianotti TF, Burgueno AL, et al. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut 2013;62:1356–63
  • Infantino V, Castegna A, Iacobazzi F, et al. Impairment of methyl cycle affects mitochondrial methyl availability and glutathione level in Down's syndrome. Mol Genet Metab 2011;102:378–82
  • Schatz P, Dietrich D, Koenig T, et al. Development of a diagnostic microarray assay to assess the risk of recurrence of prostate cancer based on PITX2 DNA methylation. J Mol Diagn 2010;12:345–53
  • Dietrich D, Kneip C, Raji O, et al. Performance evaluation of the DNA methylation biomarker SHOX2 for the aid in diagnosis of lung cancer based on the analysis of bronchial aspirates. Int J Oncol 2012;40:825–32
  • Kneip C, Schmidt B, Seegebarth A, et al. SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. J Thorac Oncol 2011;6:1632–8
  • Esteller M, Garcia-Foncillas J, Andion E, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med 2000;343:1350–4
  • Church TR, Wandell M, Lofton-Day C, et al. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut 2014;63:317–25
  • Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 2004;351:2817–26
  • van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002;347:1999–2009
  • Cheetham SW, Gruhl F, Mattick JS, Dinger ME. Long noncoding RNAs and the genetics of cancer. Br J Cancer 2013;108:2419–25
  • Shi X, Sun M, Liu H, et al. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett 2013;339:159–66
  • Van RK, Pollet J, Calin GA. miRNAs and long noncoding RNAs as biomarkers in human diseases. Expert Rev Mol Diagn 2013;13:183–204
  • Esteller M. Non-coding RNAs in human disease. Nat Rev Genet 2011;12:861–74
  • Nana-Sinkam SP, Croce CM. Clinical applications for microRNAs in cancer. Clin Pharmacol Ther 2013;93:98–104
  • Griffiths-Jones S, Grocock RJ, van DS, et al. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res 2006;34:D140–4
  • Nair VS, Maeda LS, Ioannidis JP. Clinical outcome prediction by microRNAs in human cancer: a systematic review. J Natl Cancer Inst 2012;104:528–40
  • Schaefer A, Jung M, Kristiansen G, et al. MicroRNAs and cancer: current state and future perspectives in urologic oncology. Urol Oncol 2010;28:4–13
  • Bamezai S, Rawat VP, Buske C. Concise review: the Piwi-piRNA axis: pivotal beyond transposon silencing. Stem Cells 2012;30:2603–11
  • Peng JC, Lin H. Beyond transposons: the epigenetic and somatic functions of the Piwi-piRNA mechanism. Curr Opin Cell Biol 2013;25:190–4
  • Mei Y, Clark D, Mao L. Novel dimensions of piRNAs in cancer. Cancer Lett 2013;336:46–52
  • Cheng J, Guo JM, Xiao BX, et al. piRNA, the new non-coding RNA, is aberrantly expressed in human cancer cells. Clin Chim Acta 2011;412:1621–5
  • Huang G, Hu H, Xue X, et al. Altered expression of piRNAs and their relation with clinicopathologic features of breast cancer. Clin Transl Oncol 2013;15:563–8
  • Matera AG, Terns RM, Terns MP. Non-coding RNAs: lessons from the small nuclear and small nucleolar RNAs. Nat Rev Mol Cell Biol 2007;8:209–20
  • Mannoor K, Liao J, Jiang F. Small nucleolar RNAs in cancer. Biochim Biophys Acta 2012;1826:121–8
  • Su H, Xu T, Ganapathy S, et al. Elevated snoRNA biogenesis is essential in breast cancer. Oncogene 2014;33:1348–58
  • Xhemalce B, Robson SC, Kouzarides T. Human RNA methyltransferase BCDIN3D regulates microRNA processing. Cell 2012;151:278–88
  • Tuorto F, Liebers R, Musch T, et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat Struct Mol Biol 2012;19:900–5
  • Amort T, Souliere MF, Wille A, et al. Long non-coding RNAs as targets for cytosine methylation. RNA Biol 2013;10:1003–9
  • Meyer KD, Saletore Y, Zumbo P, et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 2012;149:1635–46
  • Schroder FH. PSA screening – a review of recent studies. Eur J Cancer 2009;45:402–4
  • Bangma CH, Bul M, van der Kwast TH, et al. Active surveillance for low-risk prostate cancer. Crit Rev Oncol Hematol 2013;85:295–302
  • Moyer VA. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2012;157:120–34
  • Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 2009;360:1320–8
  • Andriole GL, Crawford ED, Grubb RL III, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med 2009;360:1310–9
  • Kattan MW, Eastham JA, Stapleton AM, et al. A preoperative nomogram for disease recurrence following radical prostatectomy for prostate cancer. J Natl Cancer Inst 1998;90:766–71
  • Stewart GD, Van NL, Delvenne P, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol 2013;189:1110–6
  • Van NL, Bigley J, Toll A, et al. A tissue biopsy-based epigenetic multiplex PCR assay for prostate cancer detection. BMC Urol 2012;12:16
  • Weiss G, Cottrell S, Distler J, et al. DNA methylation of the PITX2 gene promoter region is a strong independent prognostic marker of biochemical recurrence in patients with prostate cancer after radical prostatectomy. J Urol 2009;181:1678–85
  • Banez LL, Sun L, van Leenders GJ, et al. Multicenter clinical validation of PITX2 methylation as a prostate specific antigen recurrence predictor in patients with post-radical prostatectomy prostate cancer. J Urol 2010;184:149–56
  • Dietrich D, Hasinger O, Banez LL, et al. Development and clinical validation of a real-time PCR assay for PITX2 DNA methylation to predict prostate-specific antigen recurrence in prostate cancer patients following radical prostatectomy. J Mol Diagn 2013;15:270–9
  • Vinarskaja A, Schulz WA, Ingenwerth M, et al. Association of PITX2 mRNA down-regulation in prostate cancer with promoter hypermethylation and poor prognosis. Urol Oncol 2013;31:622–7
  • Schayek H, Bentov I, Jacob-Hirsch J, et al. Global methylation analysis identifies PITX2 as an upstream regulator of the androgen receptor and IGF-I receptor genes in prostate cancer. Horm Metab Res 2012;44:511–9
  • Chao C, Chi M, Preciado M, Black MH. Methylation markers for prostate cancer prognosis: a systematic review. Cancer Causes Control 2013;24:1615–41
  • Heichman KA, Warren JD. DNA methylation biomarkers and their utility for solid cancer diagnostics. Clin Chem Lab Med 2012;50:1707–21
  • Knezevic D, Goddard AD, Natraj N, et al. Analytical validation of the Oncotype DX prostate cancer assay - a clinical RT-PCR assay optimized for prostate needle biopsies. BMC Genomics 2013;14:690
  • Karnes RJ, Bergstralh EJ, Davicioni E, et al. Validation of a genomic classifier that predicts metastasis following radical prostatectomy in an at risk patient population. J Urol 2013;190:2047–53
  • Cuzick J, Swanson GP, Fisher G, et al. Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol 2011;12:245–55
  • Cooperberg MR, Simko JP, Cowan JE, et al. Validation of a cell-cycle progression gene panel to improve risk stratification in a contemporary prostatectomy cohort. J Clin Oncol 2013;31:1428–34
  • Hessels D, Klein Gunnewiek JM, van OI, et al. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol 2003;44:8–15
  • Hansen J, Auprich M, Ahyai SA, et al. Initial prostate biopsy: development and internal validation of a biopsy-specific nomogram based on the prostate cancer antigen 3 assay. Eur Urol 2013;63:201–9
  • Auprich M, Bjartell A, Chun FK, et al. Contemporary role of prostate cancer antigen 3 in the management of prostate cancer. Eur Urol 2011;60:1045–54
  • Powledge TM. Virus linked with prostate cancer. Lancet Oncol 2006;7:291
  • Hampton T. New virus linked to prostate cancer. JAMA 2006;295. doi:10.1001/jama.295.13.1503
  • Aloia AL, Sfanos KS, Isaacs WB, et al. XMRV: a new virus in prostate cancer? Cancer Res 2010;70:10028–33
  • Shen MM. Chromoplexy: a new category of complex rearrangements in the cancer genome. Cancer Cell 2013;23:567–9
  • Beltran H, Rubin MA. New strategies in prostate cancer: translating genomics into the clinic. Clin Cancer Res 2013;19:517–23
  • Barbieri CE, Demichelis F, Rubin MA. Molecular genetics of prostate cancer: emerging appreciation of genetic complexity. Histopathology 2012;60:187–98
  • Barbieri CE, Bangma CH, Bjartell A, et al. The mutational landscape of prostate cancer. Eur Urol 2013;64:567–76
  • Barbieri CE, Baca SC, Lawrence MS, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 2012;44:685–9
  • Stoehr R, Taubert H, Gaisa NT, et al. Lack of evidence for frequent MED12 p.L1224F mutation in prostate tumours from Caucasian patients. J Pathol 2013;230:453–6
  • Barbieri CE, Sboner A, Rubin MA, et al. Lack of evidence for frequent MED12 p.L1224F mutation in prostate tumours from Caucasian patients. J Pathol 2013;230:453–6
  • Eeles RA, Olama AA, Benlloch S, et al. Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat Genet 2013;45:385–2
  • Gudmundsson J, Sulem P, Gudbjartsson DF, et al. A study based on whole-genome sequencing yields a rare variant at 8q24 associated with prostate cancer. Nat Genet 2012;44:1326–9
  • Kote-Jarai Z, Saunders EJ, Leongamornlert DA, et al. Fine-mapping identifies multiple prostate cancer risk loci at 5p15, one of which associates with TERT expression. Hum Mol Genet 2013;22:2520–8
  • Kote-Jarai Z, Olama AA, Giles GG, et al. Seven prostate cancer susceptibility loci identified by a multi-stage genome-wide association study. Nat Genet 2011;43:785–91
  • Schumacher FR, Berndt SI, Siddiq A, et al. Genome-wide association study identifies new prostate cancer susceptibility loci. Hum Mol Genet 2011;20:3867–75
  • Takata R, Akamatsu S, Kubo M, et al. Genome-wide association study identifies five new susceptibility loci for prostate cancer in the Japanese population. Nat Genet 2010;42:751–4
  • Haiman CA, Chen GK, Blot WJ, et al. Genome-wide association study of prostate cancer in men of African ancestry identifies a susceptibility locus at 17q21. Nat Genet 2011;43:570–3
  • Haiman CA, Han Y, Feng Y, et al. Genome-wide testing of putative functional exonic variants in relationship with breast and prostate cancer risk in a multiethnic population. PLoS Genet 2013;9:e1003419
  • Jin G, Sun J, Liu W, et al. Genome-wide copy-number variation analysis identifies common genetic variants at 20p13 associated with aggressiveness of prostate cancer. Carcinogenesis 2011;32:1057–62
  • Tindall EA, Severi G, Hoang HN, et al. Comprehensive analysis of the cytokine-rich chromosome 5q31.1 region suggests a role for IL-4 gene variants in prostate cancer risk. Carcinogenesis 2010;31:1748–54
  • Yeager M, Deng Z, Boland J, et al. Comprehensive resequence analysis of a 97 kb region of chromosome 10q11.2 containing the MSMB gene associated with prostate cancer. Hum Genet 2009;126:743–50
  • Chang BL, Cramer SD, Wiklund F, et al. Fine mapping association study and functional analysis implicate a SNP in MSMB at 10q11 as a causal variant for prostate cancer risk. Hum Mol Genet 2009;18:1368–75
  • Gudmundsson J, Sulem P, Rafnar T, et al. Common sequence variants on 2p15 and Xp11.22 confer susceptibility to prostate cancer. Nat Genet 2008;40:281–3
  • Gudmundsson J, Sulem P, Steinthorsdottir V, et al. Two variants on chromosome 17 confer prostate cancer risk, and the one in TCF2 protects against type 2 diabetes. Nat Genet 2007;39:977–83
  • Tindall EA, Severi G, Hoang HN, et al. Interleukin-6 promoter variants, prostate cancer risk, and survival. Prostate 2012;72:1701–7
  • Bailey-Wilson JE, Childs EJ, Cropp CD, et al. Analysis of Xq27-28 linkage in the international consortium for prostate cancer genetics (ICPCG) families. BMC Med Genet 2012;13:46
  • Akamatsu S, Takata R, Haiman CA, et al. Common variants at 11q12, 10q26 and 3p11.2 are associated with prostate cancer susceptibility in Japanese. Nat Genet 2012;44:426–9
  • Rafnar T, Sulem P, Stacey SN, et al. Sequence variants at the TERT-CLPTM1L locus associate with many cancer types. Nat Genet 2009;41:221–7
  • Baird DM. Variation at the TERT locus and predisposition for cancer. Expert Rev Mol Med 2010;12. doi: 10.1017/S146239941000147X
  • Gunes C, Rudolph KL. The role of telomeres in stem cells and cancer. Cell 2013;152:390–3
  • Amundadottir LT, Sulem P, Gudmundsson J, et al. A common variant associated with prostate cancer in European and African populations. Nat Genet 2006;38:652–8
  • Yeager M, Orr N, Hayes RB, et al. Genome-wide association study of prostate cancer identifies a second risk locus at 8q24. Nat Genet 2007;39:645–9
  • Yeager M, Xiao N, Hayes RB, et al. Comprehensive resequence analysis of a 136 kb region of human chromosome 8q24 associated with prostate and colon cancers. Hum Genet 2008;124:161–70
  • Haiman CA, Patterson N, Freedman ML, et al. Multiple regions within 8q24 independently affect risk for prostate cancer. Nat Genet 2007;39:638–44
  • Freedman ML, Haiman CA, Patterson N, et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc Natl Acad Sci USA 2006;103:14068–73
  • Wang C, Lisanti MP, Liao DJ. Reviewing once more the c-myc and Ras collaboration: converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle 2011;10:57–67
  • Prensner JR, Iyer MK, Balbin OA, et al. Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 2011;29:742–9
  • Chung S, Nakagawa H, Uemura M, et al. Association of a novel long non-coding RNA in 8q24 with prostate cancer susceptibility. Cancer Sci 2011;102:245–52
  • Yang L, Lin C, Jin C, et al. lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature 2013;500:598–602
  • Takayama K, Horie-Inoue K, Katayama S, et al. Androgen-responsive long noncoding RNA CTBP1-AS promotes prostate cancer. EMBO J 2013;32:1665–80
  • Pflueger D, Rickman DS, Sboner A, et al. N-myc downstream regulated gene 1 (NDRG1) is fused to ERG in prostate cancer. Neoplasia 2009;11:804–11
  • Laurent C, Valet F, Planque N, et al. High PTP4A3 phosphatase expression correlates with metastatic risk in uveal melanoma patients. Cancer Res 2011;71:666–74
  • Lu Y, Lemon W, Liu PY, et al. A gene expression signature predicts survival of patients with stage I non-small cell lung cancer. PLoS Med 2006;3:e467
  • Gu Z, Thomas G, Yamashiro J, et al. Prostate stem cell antigen (PSCA) expression increases with high gleason score, advanced stage and bone metastasis in prostate cancer. Oncogene 2000;19:1288–96
  • Reiter RE, Sato I, Thomas G, et al. Coamplification of prostate stem cell antigen (PSCA) and MYC in locally advanced prostate cancer. Genes Chromosomes Cancer 2000;27:95–103
  • Su Y, Meador JA, Calaf GM, et al. Human RecQL4 helicase plays critical roles in prostate carcinogenesis. Cancer Res 2010;70:9207–17
  • Pearson HB, Perez-Mancera PA, Dow LE, et al. SCRIB expression is deregulated in human prostate cancer, and its deficiency in mice promotes prostate neoplasia. J Clin Invest 2011;121:4257–67
  • Yoo NJ, Hur SY, Kim MS, et al. Immunohistochemical analysis of RNA-induced silencing complex-related proteins AGO2 and TNRC6A in prostate and esophageal cancers. APMIS 2010;118:271–6
  • Visakorpi T, Kallioniemi AH, Syvanen AC, et al. Genetic changes in primary and recurrent prostate cancer by comparative genomic hybridization. Cancer Res 1995;55:342–7
  • Nupponen NN, Kakkola L, Koivisto P, Visakorpi T. Genetic alterations in hormone-refractory recurrent prostate carcinomas. Am J Pathol 1998;153:141–8
  • El GA, Bubendorf L, Willi N, et al. Discovery of new DNA amplification loci in prostate cancer by comparative genomic hybridization. Prostate 2001;46:184–90
  • Paris PL, Albertson DG, Alers JC, et al. High-resolution analysis of paraffin-embedded and formalin-fixed prostate tumors using comparative genomic hybridization to genomic microarrays. Am J Pathol 2003;162:763–70
  • Kumar-Sinha C, Tomlins SA, Chinnaiyan AM. Recurrent gene fusions in prostate cancer. Nat Rev Cancer 2008;8:497–511
  • Kristiansen G. Diagnostic and prognostic molecular biomarkers for prostate cancer. Histopathology 2012;60:125–41
  • Scher HI, Morris MJ, Larson S, Heller G. Validation and clinical utility of prostate cancer biomarkers. Nat Rev Clin Oncol 2013;10:225–34
  • Grasso CS, Wu YM, Robinson DR, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239–43
  • de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011;364:1995–2005
  • Tran C, Ouk S, Clegg NJ, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 2009;324:787–90
  • Visakorpi T, Hyytinen E, Koivisto P, et al. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat Genet 1995;9:401–6
  • Linja MJ, Savinainen KJ, Saramaki OR, et al. Amplification and overexpression of androgen receptor gene in hormone-refractory prostate cancer. Cancer Res 2001;61:3550–5
  • Taylor BS, Schultz N, Hieronymus H, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 2010;18:11–22
  • Bubendorf L, Kononen J, Koivisto P, et al. Survey of gene amplifications during prostate cancer progression by high-throughout fluorescence in situ hybridization on tissue microarrays. Cancer Res 1999;59:803–6
  • Palmberg C, Koivisto P, Hyytinen E, et al. Androgen receptor gene amplification in a recurrent prostate cancer after monotherapy with the nonsteroidal potent antiandrogen Casodex (bicalutamide) with a subsequent favorable response to maximal androgen blockade. Eur Urol 1997;31:216–9
  • Guo Z, Yang X, Sun F, et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res 2009;69:2305–13
  • Hu R, Dunn TA, Wei S, et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res 2009;69:16–22
  • Sun S, Sprenger CC, Vessella RL, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clin Invest 2010;120:2715–30
  • Hara T, Miyazaki J, Araki H, et al. Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome. Cancer Res 2003;63:149–53
  • Taplin ME, Bubley GJ, Ko YJ, et al. Selection for androgen receptor mutations in prostate cancers treated with androgen antagonist. Cancer Res 1999;59:2511–5
  • Zhao XY, Malloy PJ, Krishnan AV, et al. Glucocorticoids can promote androgen-independent growth of prostate cancer cells through a mutated androgen receptor. Nat Med 2000;6:703–6
  • Gottlieb B, Beitel LK, Nadarajah A, et al. The androgen receptor gene mutations database: 2012 update. Hum Mutat 2012;33:887–94
  • Signoretti S, Montironi R, Manola J, et al. Her-2-neu expression and progression toward androgen independence in human prostate cancer. J Natl Cancer Inst 2000;92:1918–25
  • Gregory CW, Whang YE, McCall W, et al. Heregulin-induced activation of HER2 and HER3 increases androgen receptor transactivation and CWR-R1 human recurrent prostate cancer cell growth. Clin Cancer Res 2005;11:1704–12
  • Neto AS, Tobias-Machado M, Wroclawski ML, et al. Her-2/neu expression in prostate adenocarcinoma: a systematic review and meta-analysis. J Urol 2010;184:842–50
  • Verhagen PC, van Duijn PW, Hermans KG, et al. The PTEN gene in locally progressive prostate cancer is preferentially inactivated by bi-allelic gene deletion. J Pathol 2006;208:699–707
  • Yoshimoto M, Cutz JC, Nuin PA, et al. Interphase FISH analysis of PTEN in histologic sections shows genomic deletions in 68% of primary prostate cancer and 23% of high-grade prostatic intra-epithelial neoplasias. Cancer Genet Cytogenet 2006;169:128–37
  • Yoshimoto M, Cunha IW, Coudry RA, et al. FISH analysis of 107 prostate cancers shows that PTEN genomic deletion is associated with poor clinical outcome. Br J Cancer 2007;97:678–85
  • King JC, Xu J, Wongvipat J, et al. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nat Genet 2009;41:524–6
  • Carver BS, Tran J, Gopalan A, et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat Genet 2009;41:619–24
  • Yoshimoto M, Joshua AM, Cunha IW, et al. Absence of TMPRSS2:ERG fusions and PTEN losses in prostate cancer is associated with a favorable outcome. Mod Pathol 2008;21:1451–60
  • Reid AH, Attard G, Ambroisine L, et al. Molecular characterisation of ERG, ETV1 and PTEN gene loci identifies patients at low and high risk of death from prostate cancer. Br J Cancer 2010;102:678–84
  • Edwards SM, Evans DG, Hope Q, et al. Prostate cancer in BRCA2 germline mutation carriers is associated with poorer prognosis. Br J Cancer 2010;103:918–24
  • Agalliu I, Gern R, Leanza S, Burk RD. Associations of high-grade prostate cancer with BRCA1 and BRCA2 founder mutations. Clin Cancer Res 2009;15:1112–20
  • Agalliu I, Karlins E, Kwon EM, et al. Rare germline mutations in the BRCA2 gene are associated with early-onset prostate cancer. Br J Cancer 2007;97:826–31
  • Narod SA, Neuhausen S, Vichodez G, et al. Rapid progression of prostate cancer in men with a BRCA2 mutation. Br J Cancer 2008;99:371–4
  • Castro E, Goh C, Olmos D, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol 2013;31:1748–57
  • Cooney KA, Wetzel JC, Merajver SD, et al. Distinct regions of allelic loss on 13q in prostate cancer. Cancer Res 1996;56:1142–5
  • Edwards SM, Dunsmuir WD, Gillett CE, et al. Immunohistochemical expression of BRCA2 protein and allelic loss at the BRCA2 locus in prostate cancer. CRC/BPG UK Familial Prostate Cancer Study Collaborators. Int J Cancer 1998;78:1–7
  • Willems AJ, Dawson SJ, Samaratunga H, et al. Loss of heterozygosity at the BRCA2 locus detected by multiplex ligation-dependent probe amplification is common in prostate cancers from men with a germline BRCA2 mutation. Clin Cancer Res 2008;14:2953–61
  • Bancroft EK, Page EC, Castro E, et al. Targeted prostate cancer screening in BRCA1 and BRCA2 mutation carriers: results from the initial screening round of the IMPACT study. Eur Urol 2014. doi: 10.1016/j.eururo.2014.01.003
  • Burger M, Catto JW, Dalbagni G, et al. Epidemiology and risk factors of urothelial bladder cancer. Eur Urol 2013;63:234–41
  • Abol-Enein H, Kava BR, Carmack AJ. Nonurothelial cancer of the bladder. Urology 2007;69:93–104
  • Raghavan D, Shipley WU, Garnick MB, et al. Biology and management of bladder cancer. N Engl J Med 1990;322:1129–38
  • Cappellen D, De OC, Ricol D, et al. Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas. Nat Genet 1999;23:18–20
  • Gibas Z, Prout GR Jr, Connolly JG, et al. Nonrandom chromosomal changes in transitional cell carcinoma of the bladder. Cancer Res 1984;44:1257–64
  • Al Hussain TO, Akhtar M. Molecular basis of urinary bladder cancer. Adv Anat Pathol 2013;20:53–60
  • Wagner U, Bubendorf L, Gasser TC, et al. Chromosome 8p deletions are associated with invasive tumor growth in urinary bladder cancer. Am J Pathol 1997;151:753–9
  • Gui Y, Guo G, Huang Y, et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet 2011;43:875–8
  • Kiemeney LA, Thorlacius S, Sulem P, et al. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat Genet 2008;40:1307–12
  • Wu X, Ye Y, Kiemeney LA, et al. Genetic variation in the prostate stem cell antigen gene PSCA confers susceptibility to urinary bladder cancer. Nat Genet 2009;41:991–5
  • Kiemeney LA, Sulem P, Besenbacher S, et al. A sequence variant at 4p16.3 confers susceptibility to urinary bladder cancer. Nat Genet 2010;42:415–9
  • Rothman N, Garcia-Closas M, Chatterjee N, et al. A multi-stage genome-wide association study of bladder cancer identifies multiple susceptibility loci. Nat Genet 2010;42:978–84
  • Black PC, Dinney CP. Growth factors and receptors as prognostic markers in urothelial carcinoma. Curr Urol Rep 2008;9:55–61
  • Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions in bladder cancer. Hum Mol Genet 2013;22:795–803
  • Kandimalla R, van Tilborg AA, Zwarthoff EC. DNA methylation-based biomarkers in bladder cancer. Nat Rev Urol 2013;10:327–35
  • Guney AI, Ergec DS, Tavukcu HH, et al. Detection of mitochondrial DNA mutations in nonmuscle invasive bladder cancer. Genet Test Mol Biomarkers 2012;16:672–8
  • Lam JS, Shvarts O, Leppert JT, et al. Renal cell carcinoma 2005: new frontiers in staging, prognostication and targeted molecular therapy. J Urol 2005;173:1853–62
  • Montie JE. Follow-up after partial or total nephrectomy for renal cell carcinoma. Urol Clin North Am 1994;21:589–92
  • Furge KA, Lucas KA, Takahashi M, et al. Robust classification of renal cell carcinoma based on gene expression data and predicted cytogenetic profiles. Cancer Res 2004;64:4117–21
  • Crumley SM, Divatia M, Truong L, et al. Renal cell carcinoma: evolving and emerging subtypes. World J Clin Cases 2013;1:262–75
  • Girgis AH, Iakovlev VV, Beheshti B, et al. Multilevel whole-genome analysis reveals candidate biomarkers in clear cell renal cell carcinoma. Cancer Res 2012;72:5273–84
  • Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013;499:43–9
  • van den Berg E, Dijkhuizen T. Classification of renal cell cancer based on (cyto)genetic analysis. Contrib Nephrol 1999;128:51–61
  • van HG, Dalgliesh GL, Davies H, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 2009;41:521–3
  • Dalgliesh GL, Furge K, Greenman C, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 2010;463:360–3
  • Varela I, Tarpey P, Raine K, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 2011;469:539–42
  • Pena-Llopis S, Vega-Rubin-de-Celis S, Liao A, et al. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 2012;44:751–9
  • Kapur P, Pena-Llopis S, Christie A, et al. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol 2013;14:159–67
  • Junker K, Weirich G, Amin MB, et al. Genetic subtyping of renal cell carcinoma by comparative genomic hybridization. Recent Results Cancer Res 2003;162:169–75
  • Boja E, Hiltke T, Rivers R, et al. Evolution of clinical proteomics and its role in medicine. J Proteome Res 2011;10:66–84
  • Kovacs G, Fuzesi L, Emanual A, Kung HF. Cytogenetics of papillary renal cell tumors. Genes Chromosomes Cancer 1991;3:249–55
  • Toro JR, Glenn G, Duray P, et al. Birt-Hogg-Dube syndrome: a novel marker of kidney neoplasia. Arch Dermatol 1999;135:1195–202
  • Nagy A, Zoubakov D, Stupar Z, Kovacs G. Lack of mutation of the folliculin gene in sporadic chromophobe renal cell carcinoma and renal oncocytoma. Int J Cancer 2004;109:472–5
  • Davidoff AM. Wilms tumor. Adv Pediatr 2012;59:247–67
  • Bamford S, Dawson E, Forbes S, et al. The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br J Cancer 2004;91:355–8
  • Purdue MP, Johansson M, Zelenika D, et al. Genome-wide association study of renal cell carcinoma identifies two susceptibility loci on 2p21 and 11q13.3. Nat Genet 2011;43:60–5
  • Xia G, Kageyama Y, Hayashi T, et al. Regulation of vascular endothelial growth factor transcription by endothelial PAS domain protein 1 (EPAS1) and possible involvement of EPAS1 in the angiogenesis of renal cell carcinoma. Cancer 2001;91:1429–36
  • Gudmundsson J, Sulem P, Gudbjartsson DF, et al. A common variant at 8q24.21 is associated with renal cell cancer. Nat Commun 2013;4. doi: 10.1038/ncomms3776
  • Bertolotto C, Lesueur F, Giuliano S, et al. A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 2011;480:94–8
  • van der Veldt AA, Eechoute K, Gelderblom H, et al. Genetic polymorphisms associated with a prolonged progression-free survival in patients with metastatic renal cell cancer treated with sunitinib. Clin Cancer Res 2011;17:620–9
  • Hoffman AM, Cairns P. Epigenetics of kidney cancer and bladder cancer. Epigenomics 2011;3:19–34
  • Young AC, Craven RA, Cohen D, et al. Analysis of VHL gene alterations and their relationship to clinical parameters in sporadic conventional renal cell carcinoma. Clin Cancer Res 2009;15:7582–92
  • Dahl E, Wiesmann F, Woenckhaus M, et al. Frequent loss of SFRP1 expression in multiple human solid tumours: association with aberrant promoter methylation in renal cell carcinoma. Oncogene 2007;26:5680–91
  • Dulaimi E, Ibanez dCI, Uzzo RG, et al. Promoter hypermethylation profile of kidney cancer. Clin Cancer Res 2004;10:3972–9
  • Ricketts CJ, Morris MR, Gentle D, et al. Genome-wide CpG island methylation analysis implicates novel genes in the pathogenesis of renal cell carcinoma. Epigenetics 2012;7:278–90
  • Youssef YM, White NM, Grigull J, et al. Accurate molecular classification of kidney cancer subtypes using microRNA signature. Eur Urol 2011;59:721–30
  • Jung M, Mollenkopf HJ, Grimm C, et al. MicroRNA profiling of clear cell renal cell cancer identifies a robust signature to define renal malignancy. J Cell Mol Med 2009;13:3918–28
  • Petillo D, Kort EJ, Anema J, et al. MicroRNA profiling of human kidney cancer subtypes. Int J Oncol 2009;35:109–14
  • Catto JW, Alcaraz A, Bjartell AS, et al. MicroRNA in prostate, bladder, and kidney cancer: a systematic review. Eur Urol 2011;59:671–81
  • Chen Z, Li Y, Zhang H, et al. Hypoxia-regulated microRNA-210 modulates mitochondrial function and decreases ISCU and COX10 expression. Oncogene 2010;29:4362–8
  • Nakada C, Matsuura K, Tsukamoto Y, et al. Genome-wide microRNA expression profiling in renal cell carcinoma: significant down-regulation of miR-141 and miR-200c. J Pathol 2008;216:418–27
  • Mahotka C, Krieg T, Krieg A, et al. Distinct in vivo expression patterns of survivin splice variants in renal cell carcinomas. Int J Cancer 2002;100:30–6
  • Parker AS, Kosari F, Lohse CM, et al. High expression levels of survivin protein independently predict a poor outcome for patients who undergo surgery for clear cell renal cell carcinoma. Cancer 2006;107:37–45
  • Higgins JP, Shinghal R, Gill H, et al. Gene expression patterns in renal cell carcinoma assessed by complementary DNA microarray. Am J Pathol 2003;162:925–32
  • Takahashi M, Rhodes DR, Furge KA, et al. Gene expression profiling of clear cell renal cell carcinoma: gene identification and prognostic classification. Proc Natl Acad Sci USA 2001;98:9754–9
  • Brannon AR, Reddy A, Seiler M, et al. Molecular stratification of clear cell renal cell carcinoma by consensus clustering reveals distinct subtypes and survival patterns. Genes Cancer 2010;1:152–63
  • Brannon AR, Haake SM, Hacker KE, et al. Meta-analysis of clear cell renal cell carcinoma gene expression defines a variant subgroup and identifies gender influences on tumor biology. Eur Urol 2012;61:258–68
  • Hayes-Lattin B, Nichols CR. Testicular cancer: a prototypic tumor of young adults. Semin Oncol 2009;36:432–8
  • Hemminki K, Li X. Familial risk in testicular cancer as a clue to a heritable and environmental aetiology. Br J Cancer 2004;90:1765–70
  • Rashid HH, Cos LR, Weinberg E, Messing EM. Testicular microlithiasis: a review and its association with testicular cancer. Urol Oncol 2004;22:285–9
  • Horwich A, Shipley J, Huddart R. Testicular germ-cell cancer. Lancet 2006;367:754–65
  • Chaganti RS, Houldsworth J. Genetics and biology of adult human male germ cell tumors. Cancer Res 2000;60:1475–82
  • Reuter VE. Origins and molecular biology of testicular germ cell tumors. Mod Pathol 2005;18(Suppl 2):S51–60
  • Mostert MC, Verkerk AJ, van de Pol M, et al. Identification of the critical region of 12p over-representation in testicular germ cell tumors of adolescents and adults. Oncogene 1998;16:2617–27
  • Korkola JE, Houldsworth J, Chadalavada RS, et al. Down-regulation of stem cell genes, including those in a 200-kb gene cluster at 12p13.31, is associated with in vivo differentiation of human male germ cell tumors. Cancer Res 2006;66:820–7
  • Nathanson KL, Kanetsky PA, Hawes R, et al. The Y deletion gr/gr and susceptibility to testicular germ cell tumor. Am J Hum Genet 2005;77:1034–43
  • Turnbull C, Rapley EA, Seal S, et al. Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer. Nat Genet 2010;42:604–7
  • Rapley EA, Turnbull C, Al Olama AA, et al. A genome-wide association study of testicular germ cell tumor. Nat Genet 2009;41:807–10
  • Kanetsky PA, Mitra N, Vardhanabhuti S, et al. Common variation in KITLG and at 5q31.3 predisposes to testicular germ cell cancer. Nat Genet 2009;41:811–5
  • Kanetsky PA, Mitra N, Vardhanabhuti S, et al. A second independent locus within DMRT1 is associated with testicular germ cell tumor susceptibility. Hum Mol Genet 2011;20:3109–17
  • Ruark E, Seal S, McDonald H, et al. Identification of nine new susceptibility loci for testicular cancer, including variants near DAZL and PRDM14. Nat Genet 2013;45:686–9
  • Wermann H, Stoop H, Gillis AJ, et al. Global DNA methylation in fetal human germ cells and germ cell tumours: association with differentiation and cisplatin resistance. J Pathol 2010;221:433–42
  • Koul S, McKiernan JM, Narayan G, et al. Role of promoter hypermethylation in Cisplatin treatment response of male germ cell tumors. Mol Cancer 2004;3. doi:10.1186/1476-4598-3-16
  • Velasco A, Corvalan A, Wistuba II, et al. Mismatch repair expression in testicular cancer predicts recurrence and survival. Int J Cancer 2008;122:1774–7
  • Palmer RD, Murray MJ, Saini HK, et al. Malignant germ cell tumors display common microRNA profiles resulting in global changes in expression of messenger RNA targets. Cancer Res 2010;70:2911–23
  • Dieckmann KP, Spiekermann M, Balks T, et al. MicroRNAs miR-371-3 in serum as diagnostic tools in the management of testicular germ cell tumours. Br J Cancer 2012;107:1754–60
  • Giuliano AR, Lee JH, Fulp W, et al. Incidence and clearance of genital human papillomavirus infection in men (HIM): a cohort study. Lancet 2011;377:932–40
  • Miralles-Guri C, Bruni L, Cubilla AL, et al. Human papillomavirus prevalence and type distribution in penile carcinoma. J Clin Pathol 2009;62:870–8
  • Rubin MA, Kleter B, Zhou M, et al. Detection and typing of human papillomavirus DNA in penile carcinoma: evidence for multiple independent pathways of penile carcinogenesis. Am J Pathol 2001;159:1211–8
  • Guerrero-Setas D, Perez-Janices N, Ojer A, et al. Differential gene hypermethylation in genital lichen sclerosus and cancer: a comparative study. Histopathology 2013;63:659–69
  • Guerrero D, Guarch R, Ojer A, et al. Hypermethylation of the thrombospondin-1 gene is associated with poor prognosis in penile squamous cell carcinoma. BJU Int 2008;102:747–55
  • Kuasne H, Marchi FA, Rogatto SR, de Syllos Colus IM. Epigenetic mechanisms in penile carcinoma. Int J Mol Sci 2013;14:10791–808
  • Maier S, Wilbertz T, Braun M, et al. SOX2 amplification is a common event in squamous cell carcinomas of different organ sites. Hum Pathol 2011;42:1078–88
  • Bass AJ, Watanabe H, Mermel CH, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet 2009;41:1238–42
  • Andersson P, Kolaric A, Windahl T, et al. PIK3CA, HRAS and KRAS gene mutations in human penile cancer. J Urol 2008;179:2030–4
  • Gou HF, Li X, Qiu M, et al. Epidermal growth factor receptor (EGFR)-RAS signaling pathway in penile squamous cell carcinoma. PLoS One 2013;8:e62175
  • Hall MC, Sanders JS, Vuitch F, et al. Deoxyribonucleic acid flow cytometry and traditional pathologic variables in invasive penile carcinoma: assessment of prognostic significance. Urology 1998;52:111–6
  • Berman BP, Weisenberger DJ, Aman JF, et al. Regions of focal DNA hypermethylation and long-range hypomethylation in colorectal cancer coincide with nuclear lamina-associated domains. Nat Genet 2012;44:40–6
  • Toyota M, Ahuja N, Ohe-Toyota M, et al. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA 1999;96:8681–6
  • Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 2006;38:787–93
  • Hughes LA, Melotte V, de SJ, et al. The CpG island methylator phenotype: what’s in a name? Cancer Res 2013;73:5858–68
  • Ibragimova I, Dulaimi E, Slifker MJ, et al. A global profile of gene promoter methylation in treatment-naive urothelial cancer. Epigenetics 2014;9. doi: 10.4161/epi.28078
  • Nonn L, Ananthanarayanan V, Gann PH. Evidence for field cancerization of the prostate. Prostate 2009;69:1470–9
  • Schneider KU, Dietrich D, Fleischhacker M, et al. Correlation of SHOX2 gene amplification and DNA methylation in lung cancer tumors. BMC Cancer 2011;11:102
  • Robinson MD, Stirzaker C, Statham AL, et al. Evaluation of affinity-based genome-wide DNA methylation data: effects of CpG density, amplification bias, and copy number variation. Genome Res 2010;20:1719–29
  • Ozen M, Creighton CJ, Ozdemir M, Ittmann M. Widespread deregulation of microRNA expression in human prostate cancer. Oncogene 2008;27:1788–93
  • Porkka KP, Pfeiffer MJ, Waltering KK, et al. MicroRNA expression profiling in prostate cancer. Cancer Res 2007;67:6130–5
  • Zaman MS, Chen Y, Deng G, et al. The functional significance of microRNA-145 in prostate cancer. Br J Cancer 2010;103:256–64
  • Szczyrba J, Loprich E, Wach S, et al. The microRNA profile of prostate carcinoma obtained by deep sequencing. Mol Cancer Res 2010;8:529–38
  • Wang L, Tang H, Thayanithy V, et al. Gene networks and microRNAs implicated in aggressive prostate cancer. Cancer Res 2009;69:9490–7
  • Wach S, Nolte E, Theil A, et al. MicroRNA profiles classify papillary renal cell carcinoma subtypes. Br J Cancer 2013;109:714–22
  • Hirata H, Hinoda Y, Ueno K, et al. MicroRNA-1826 directly targets beta-catenin (CTNNB1) and MEK1 (MAP2K1) in VHL-inactivated renal cancer. Carcinogenesis 2012;33:501–8
  • Gandellini P, Folini M, Longoni N, et al. miR-205 Exerts tumor-suppressive functions in human prostate through down-regulation of protein kinase Cepsilon. Cancer Res 2009;69:2287–95
  • Majid S, Dar AA, Saini S, et al. MicroRNA-205-directed transcriptional activation of tumor suppressor genes in prostate cancer. Cancer 2010;116:5637–49
  • Gottardo F, Liu CG, Ferracin M, et al. Micro-RNA profiling in kidney and bladder cancers. Urol Oncol 2007;25:387–92
  • Wiklund ED, Bramsen JB, Hulf T, et al. Coordinated epigenetic repression of the miR-200 family and miR-205 in invasive bladder cancer. Int J Cancer 2011;128:1327–34
  • Hagman Z, Haflidadottir BS, Ceder JA, et al. miR-205 negatively regulates the androgen receptor and is associated with adverse outcome of prostate cancer patients. Br J Cancer 2013;108:1668–76
  • Ribas J, Ni X, Haffner M, et al. miR-21: an androgen receptor-regulated microRNA that promotes hormone-dependent and hormone-independent prostate cancer growth. Cancer Res 2009;69:7165–9
  • Lv L, Huang F, Mao H, et al. MicroRNA-21 is overexpressed in renal cell carcinoma. Int J Biol Markers 2013;28:201–7
  • Faragalla H, Youssef YM, Scorilas A, et al. The clinical utility of miR-21 as a diagnostic and prognostic marker for renal cell carcinoma. J Mol Diagn 2012;14:385–92
  • Dong Q, Meng P, Wang T, et al. MicroRNA let-7a inhibits proliferation of human prostate cancer cells in vitro and in vivo by targeting E2F2 and CCND2. PLoS One 2010;5:e10147
  • Ambs S, Prueitt RL, Yi M, et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res 2008;68:6162–70
  • Hidaka H, Seki N, Yoshino H, et al. Tumor suppressive microRNA-1285 regulates novel molecular targets: aberrant expression and functional significance in renal cell carcinoma. Oncotarget 2012;3:44–57
  • Yoshino H, Chiyomaru T, Enokida H, et al. The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer 2011;104:808–18
  • Shi XB, Xue L, Yang J, et al. An androgen-regulated miRNA suppresses Bak1 expression and induces androgen-independent growth of prostate cancer cells. Proc Natl Acad Sci USA 2007;104:19983–8
  • Prueitt RL, Yi M, Hudson RS, et al. Expression of microRNAs and protein-coding genes associated with perineural invasion in prostate cancer. Prostate 2008;68:1152–64
  • Huang L, Luo J, Cai Q, et al. MicroRNA-125b suppresses the development of bladder cancer by targeting E2F3. Int J Cancer 2011;128:1758–69
  • Epis MR, Giles KM, Barker A, et al. miR-331-3p regulates ERBB-2 expression and androgen receptor signaling in prostate cancer. J Biol Chem 2009;284:24696–704
  • Kong D, Li Y, Wang Z, et al. miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells 2009;27:1712–21
  • Novotny GW, Belling KC, Bramsen JB, et al. MicroRNA expression profiling of carcinoma in situ cells of the testis. Endocr Relat Cancer 2012;19:365–79
  • Yu XY, Zhang Z, Liu J, et al. MicroRNA-141 is downregulated in human renal cell carcinoma and regulates cell survival by targeting CDC25B. Onco Targets Ther 2013;6:349–54
  • Kort EJ, Farber L, Tretiakova M, et al. The E2F3-Oncomir-1 axis is activated in Wilms' tumor. Cancer Res 2008;68:4034–8
  • Novotny GW, Nielsen JE, Sonne SB, et al. Analysis of gene expression in normal and neoplastic human testis: new roles of RNA. Int J Androl 2007;30:316–26
  • Sylvestre Y, De GV, Querido E, et al. An E2F/miR-20a autoregulatory feedback loop. J Biol Chem 2007;282:2135–43
  • Wu D, Huang P, Wang L, et al. MicroRNA-143 inhibits cell migration and invasion by targeting matrix metalloproteinase 13 in prostate cancer. Mol Med Rep 2013;8:626–30
  • Xu B, Niu X, Zhang X, et al. miR-143 decreases prostate cancer cells proliferation and migration and enhances their sensitivity to docetaxel through suppression of KRAS. Mol Cell Biochem 2011;350:207–13
  • Powers MP, Alvarez K, Kim HJ, Monzon FA. Molecular classification of adult renal epithelial neoplasms using microRNA expression and virtual karyotyping. Diagn Mol Pathol 2011;20:63–70
  • Noguchi S, Mori T, Hoshino Y, et al. MicroRNA-143 functions as a tumor suppressor in human bladder cancer T24 cells. Cancer Lett 2011;307:211–20
  • Lin T, Dong W, Huang J, et al. MicroRNA-143 as a tumor suppressor for bladder cancer. J Urol 2009;181:1372–80
  • Noguchi S, Yasui Y, Iwasaki J, et al. Replacement treatment with microRNA-143 and -145 induces synergistic inhibition of the growth of human bladder cancer cells by regulating PI3K/Akt and MAPK signaling pathways. Cancer Lett 2013;328:353–61
  • Galardi S, Mercatelli N, Giorda E, et al. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem 2007;282:23716–24
  • Leucci E, Patella F, Waage J, et al. microRNA-9 targets the long non-coding RNA MALAT1 for degradation in the nucleus. Sci Rep 2013;3. doi:10.1038/srep02535
  • Friedman JM, Liang G, Liu CC, et al. The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2. Cancer Res 2009;69:2623–9
  • Qiang XF, Zhang ZW, Liu Q, et al. miR-20a promotes Prostate cancer invasion and migration through targeting ABL2. J Cell Biochem 2014. doi: 10.1002/jcb.24778
  • Lichner Z, Fendler A, Saleh C, et al. MicroRNA signature helps distinguish early from late biochemical failure in prostate cancer. Clin Chem 2013;59:1595–603
  • Kotake Y, Nakagawa T, Kitagawa K, et al. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 2011;30:1956–62
  • Pasmant E, Sabbagh A, Vidaud M, Bieche I. ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J 2011;25:444–8
  • Poliseno L, Salmena L, Zhang J, et al. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 2010;465:1033–8
  • Berteaux N, Lottin S, Adriaenssens E, et al. Hormonal regulation of H19 gene expression in prostate epithelial cells. J Endocrinol 2004;183:69–78
  • Luo M, Li Z, Wang W, et al. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett 2013;333:213–21
  • Zhu Y, Yu M, Li Z, et al. ncRAN, a newly identified long noncoding RNA, enhances human bladder tumor growth, invasion, and survival. Urology 2011;77:510–5
  • Ferreira LB, Palumbo A, de Mello KD, et al. PCA3 noncoding RNA is involved in the control of prostate-cancer cell survival and modulates androgen receptor signaling. BMC Cancer 2012;12:507
  • Lin R, Maeda S, Liu C, et al. A large noncoding RNA is a marker for murine hepatocellular carcinomas and a spectrum of human carcinomas. Oncogene 2007;26:851–8
  • Wang F, Li X, Xie X, et al. UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Lett 2008;582:1919–27
  • Wang XS, Zhang Z, Wang HC, et al. Rapid identification of UCA1 as a very sensitive and specific unique marker for human bladder carcinoma. Clin Cancer Res 2006;12:4851–8
  • Dong XY, Rodriguez C, Guo P, et al. SnoRNA U50 is a candidate tumor-suppressor gene at 6q14.3 with a mutation associated with clinically significant prostate cancer. Hum Mol Genet 2008;17:1031–42

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.