Heterogeneity in the inter-tumor transcriptome of high risk prostate cancer

Alexander W Wyatt, Fan Mo, Kendric Wang, Brian McConeghy, Sonal Brahmbhatt, Lina Jong, Devon M Mitchell, Rebecca L Johnston, Anne Haegert, Estelle Li, Janet Liew, Jake Yeung, Raunak Shrestha, Anna V Lapuk, Andrew McPherson, Robert Shukin, Robert H Bell, Shawn Anderson, Jennifer Bishop, Antonio Hurtado-Coll, Hong Xiao, Arul M Chinnaiyan, Rohit Mehra, Dong Lin, Yuzhuo Wang, Ladan Fazli, Martin E Gleave, Stanislav V Volik, Colin C Collins, Alexander W Wyatt, Fan Mo, Kendric Wang, Brian McConeghy, Sonal Brahmbhatt, Lina Jong, Devon M Mitchell, Rebecca L Johnston, Anne Haegert, Estelle Li, Janet Liew, Jake Yeung, Raunak Shrestha, Anna V Lapuk, Andrew McPherson, Robert Shukin, Robert H Bell, Shawn Anderson, Jennifer Bishop, Antonio Hurtado-Coll, Hong Xiao, Arul M Chinnaiyan, Rohit Mehra, Dong Lin, Yuzhuo Wang, Ladan Fazli, Martin E Gleave, Stanislav V Volik, Colin C Collins

Abstract

Background: Genomic analyses of hundreds of prostate tumors have defined a diverse landscape of mutations and genome rearrangements, but the transcriptomic effect of this complexity is less well understood, particularly at the individual tumor level. We selected a cohort of 25 high-risk prostate tumors, representing the lethal phenotype, and applied deep RNA-sequencing and matched whole genome sequencing, followed by detailed molecular characterization.

Results: Ten tumors were exposed to neo-adjuvant hormone therapy and expressed marked evidence of therapy response in all except one extreme case, which demonstrated early resistance via apparent neuroendocrine transdifferentiation. We observe high inter-tumor heterogeneity, including unique sets of outlier transcripts in each tumor. Interestingly, outlier expression converged on druggable cellular pathways associated with cell cycle progression, translational control or immune regulation, suggesting distinct contemporary pathway affinity and a mechanism of tumor stratification. We characterize hundreds of novel fusion transcripts, including a high frequency of ETS fusions associated with complex genome rearrangements and the disruption of tumor suppressors. Remarkably, several tumors express unique but potentially-oncogenic non-ETS fusions, which may contribute to the phenotype of individual tumors, and have significance for disease progression. Finally, one ETS-negative tumor has a striking tandem duplication genotype which appears to be highly aggressive and present at low recurrence in ETS-negative prostate cancer, suggestive of a novel molecular subtype.

Conclusions: The multitude of rare genomic and transcriptomic events detected in a high-risk tumor cohort offer novel opportunities for personalized oncology and their convergence on key pathways and functions has broad implications for precision medicine.

Figures

Figure 1
Figure 1
Therapy response and resistance in neo-adjuvant hormone treated tumors. (A) Breakdown of the patient cohort. (B) Principal component analysis using RNA-seq derived transcript expression (after removal of three outliers; see Additional file 2: Figure S1) demonstrating the global split between hormone-naive tumors and tumors treated with neo-adjuvant hormone therapy (NHT). (C) Representative genes that were significantly differentially expressed between hormone-naive and NHT tumors (DESeq comparison; Benjamini-Hochberg corrected P values). (D) Expression levels of the neuroendocrine prostate cancer marker CHGA in the cohort, showing elevated levels in NHT tumors. † indicates the unique hybrid adenocarcinoma-neuroendocrine tumor described in-depth in a separate study (ref 45). (E-G) CHGA protein staining showing diffuse positivity (<10% of cells overall) in selected NHT tumors with elevated CHGA mRNA expression levels. (H) Dual AR (brown) and CHGA (pink) staining in tumor T20 (with the highest mRNA expression of CHGA in (D)) showing evidence of neuroendocrine transdifferentiation, potentially in response to 8 months of NHT. The top panel shows the diagnostic biopsy, prior to treatment, showing few (pink) CHGA positive cells but predominant (brown) AR staining, while the bottom panel shows tumor T20 (at radical prostatectomy (RP)), demonstrating small cell morphology, widespread (pink) CHGA positivity and scant remaining AR positive foci.
Figure 2
Figure 2
Significant pathway enrichment of transcripts with outlier gene expression. The heatmap in the top panel provides the pathway enrichment scores for representative canonical pathways in each tumor sample and benign tissue showing that outlier transcript sets from different tumors converge on distinct cellular functions (pathway score = -log10(Benjamini–Hochberg adjusted P value); only pathway scores >1 (that is, B-H <0.1) are shown). Note pathway names truncated from 1Role of CHK Proteins in Cell Cycle Checkpoint Control, 2Cell Cycle: G2/M DNA Damage Checkpoint Regulation, 3Role of Pattern Recognition Receptors in Recognition of Bacteria and Viruses, 4Altered T Cell and B Cell Signaling in Rheumatoid Arthritis. The bottom panel provides explorations of several key pathways, highlighting the expression distribution of representative genes within those pathways across the entire tumor cohort (full list of outlier gene sets within enriched pathways in Additional file 1: Table S8). Tumors with outlier gene enrichment within a given pathway are represented by colored circles. Immunohistochemistry images show high Ki67 indices in three tumors reflecting the probable high proliferation rate of tumors with outlier gene enrichment within cell cycle-related pathways (for additional images with larger fields see Additional file 2: Figure S3).
Figure 3
Figure 3
The landscape of non-ETS fusion genes and complex genome rearrangements in high-risk prostate cancer. (A) Depicts the number of expressed fusion genes (‘Fusion load’) and complex genome rearrangements (‘Complex’) detected in each tumor sample, and indicates selected genes involved in fusion events. Only genes with putative links to cancer are shown (full list of involved genes in Additional file 1: Table S9). Genes marked with * indicates a putative gain-of-function. † highly complex chromothripsis-driven rearrangements; ‡ involvement in complex rearrangement but not expressed; § DNA sequence coverage too low to elucidate complex rearrangements. (B) Schematic of the complex genome rearrangement (also known as chromoplexy [6]) in tumor T20 which lead to the disruption of TP53. Breakpoints in the six genes involved are indicated on the left, together with the final configuration on the right. This particular rearrangement led to the expression of just one fusion transcript (i.e. MATR3-HMG3P22). (C-F) Further schematics of complex genome rearrangements (C) shows the same rearrangement as (B)]. Green nodes indicate a gene is disrupted by rearrangement while red and gray indicate potential activating or neutral effects, respectively. Full edges represent a DNA rearrangement, and dotted lines indicate a rearrangement that was also detected in the RNA sequence data (that is, a fusion transcript was expressed). (G) Schematics of selected fusion genes with putative gain-of-function. Tumor ID is provided in each box, and major protein domains are annotated.
Figure 4
Figure 4
A tandem duplication genotype in high-risk prostate cancer. (A) Copy number profile of hormone-naïve tumor T4, showing focal gains across the entire genome, distinctive of tandem duplications. Key events discussed in the text are annotated. (B) Schematic illustrating how individual tandem duplications present at the genomic level. (C) Serial tandem duplications across the MDM2 loci in T4. The copy number plot shows a focal high gain, with the colored lines representing segments that have been duplicated. Tandem duplication ID is indicated next to each colored line and in brackets is the estimated number of copies of each tandem duplication. For example, the genetic breakpoint of the most focal tandem duplication (green line) was detectable at a high frequency indicating multiple copies, suggesting that the breakpoint itself had been subsequently duplicated, potentially by the broader tandem duplications (pink and blue lines). Note that one breakpoint (purple line) was predicted at <1 suggestive of sub-clonality and highlighting the potential for continual evolution. (D) Transcript expression of MDM2 across the high-risk cohort showing elevated expression in tumor T4 relative to the other tumors and benign samples. (E, F) Chromogenic in situ hybridization of MDM2 in benign tissue (E) and tumor (F) from T4 confirming MDM2 amplification. Note the ‘clumps’ of staining in the tumor cells suggesting MDM2 amplifications are proximally located (consistent with tandem duplication). (G) Copy number profiles from two tumors in a public dataset, which appear to harbor the distinctive pattern of focal gains across the entire genome (see Additional file 2: Figure S9 for further examples). Oncogenes within focal amplifications are annotated.

References

    1. Wyatt AW, Mo F, Wang Y, Collins CC. The diverse heterogeneity of molecular alterations in prostate cancer identified through next-generation sequencing. Asian J Androl. 2013;15:301–308. doi: 10.1038/aja.2013.13.
    1. Spans L, Clinckemalie L, Helsen C, Vanderschueren D, Boonen S, Lerut E, Joniau S, Claessens F. The genomic landscape of prostate cancer. Int J Mol Sci. 2013;14:10822–10851. doi: 10.3390/ijms140610822.
    1. Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS, Menon A, Jing X, Cao Q, Han B, Yu J, Wang L, Montie JE, Rubin MA, Pienta KJ, Roulston D, Shah RB, Varambally S, Mehra R, Chinnaiyan AM. Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature. 2007;448:595–599. doi: 10.1038/nature06024.
    1. Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, White TA, Stojanov P, Van Allen E, Stransky N, Nickerson E, Chae SS, Boysen G, Auclair D, Onofrio RC, Park K, Kitabayashi N, MacDonald TY, Sheikh K, Vuong T, Guiducci C, Cibulskis K, Sivachenko A, Carter SL, Saksena G, Voet D, Hussain WM, Ramos AH, Winckler W, Redman MC, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 2012;44:685–689. doi: 10.1038/ng.2279.
    1. Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ, Brenner JC, Asangani IA, Ateeq B, Chun SY, Siddiqui J, Sam L, Anstett M, Mehra R, Prensner JR, Palanisamy N, Ryslik GA, Vandin F, Raphael BJ, Kunju LP, Rhodes DR, Pienta KJ, Chinnaiyan AM, Tomlins SA. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487:239–243. doi: 10.1038/nature11125.
    1. Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, Drier Y, Park K, Kitabayashi N, MacDonald TY, Ghandi M, Van Allen E, Kryukov GV, Sboner A, Theurillat JP, Soong TD, Nickerson E, Auclair D, Tewari A, Beltran H, Onofrio RC, Boysen G, Guiducci C, Barbieri CE, Cibulskis K, Sivachenko A, Carter SL, Saksena G, Voet D, Ramos AH, Winckler W, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–677. doi: 10.1016/j.cell.2013.03.021.
    1. Meng MV, Elkin EP, Latini DM, Duchane J, Carroll PR. Treatment of patients with high risk localized prostate cancer: results from cancer of the prostate strategic urological research endeavor (CaPSURE) J Urol. 2005;173:1557–1561. doi: 10.1097/01.ju.0000154610.81916.81.
    1. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, Antipin Y, Mitsiades N, Landers T, Dolgalev I, Major JE, Wilson M, Socci ND, Lash AE, Heguy A, Eastham JA, Scher HI, Reuter VE, Scardino PT, Sander C, Sawyers CL, Gerald WL. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. doi: 10.1016/j.ccr.2010.05.026.
    1. Attard G, Cooper CS, de Bono JS. Steroid hormone receptors in prostate cancer: a hard habit to break? Cancer Cell. 2009;16:458–462. doi: 10.1016/j.ccr.2009.11.006.
    1. Sorrentino C, Musiani P, Pompa P, Cipollone G, Di Carlo E. Androgen deprivation boosts prostatic infiltration of cytotoxic and regulatory T lymphocytes and has no effect on disease-free survival in prostate cancer patients. Clin Cancer Res. 2011;17:1571–1581. doi: 10.1158/1078-0432.CCR-10-2804.
    1. Tang S, Moore ML, Grayson JM, Dubey P. Increased CD8+ T-cell function following castration and immunization is countered by parallel expansion of regulatory T cells. Cancer Res. 2012;72:1975–1985. doi: 10.1158/0008-5472.CAN-11-2499.
    1. Beltran H, Tagawa ST, Park K, MacDonald T, Milowsky MI, Mosquera JM, Rubin MA, Nanus DM. Challenges in recognizing treatment-related neuroendocrine prostate cancer. J Clin Oncol. 2012;30:e386–e389. doi: 10.1200/JCO.2011.41.5166.
    1. Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, Wu R, Brahmbhatt S, Mo F, Jong L, Bell RH, Anderson S, Hurtado-Cull A, Fazli L, Sharma M, Beltran H, Rubin MA, Cox ME, Gout PW, Morris J, Goldenberg L, Volik SV, Gleave ME, Collins CC. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res. 2014;74:1272–1283. doi: 10.1158/0008-5472.CAN-13-2921-T.
    1. Williamson SR, Zhang S, Yao JL, Huang J, Lopez-Beltran A, Shen S, Osunkoya AO, MacLennan GT, Montironi R, Cheng L. ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin. Mod Pathol. 2011;24:1120–1127. doi: 10.1038/modpathol.2011.56.
    1. Lotan TL, Gupta NS, Wang W, Toubaji A, Haffner MC, Chaux A, Hicks JL, Meeker AK, Bieberich CJ, De Marzo AM, Epstein JI, Netto GJ. ERG gene rearrangements are common in prostatic small cell carcinomas. Mod Pathol. 2011;24:820–828. doi: 10.1038/modpathol.2011.7.
    1. Guo CC, Dancer JY, Wang Y, Aparicio A, Navone NM, Troncoso P, Czerniak BA. TMPRSS2-ERG gene fusion in small cell carcinoma of the prostate. Hum Pathol. 2011;42:11–17. doi: 10.1016/j.humpath.2010.05.026.
    1. Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, Wang Y, Sheikh KL, Terry S, Tagawa ST, Dhir R, Nelson JB, de la Taille A, Allory Y, Gerstein MB, Perner S, Pienta KJ, Chinnaiyan AM, Wang Y, Collins CC, Gleave ME, Demichelis F, Nanus DM, Rubin MA. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discovery. 2011;1:487–495. doi: 10.1158/-11-0130.
    1. Wissing MD, Mendonca J, Kortenhorst MS, Kaelber NS, Gonzalez M, Kim E, Hammers H, van Diest PJ, Carducci MA, Kachhap SK. Targeting prostate cancer cell lines with polo-like kinase 1 inhibitors as a single agent and in combination with histone deacetylase inhibitors. FASEB J. 2013;27:4279–4293. doi: 10.1096/fj.12-222893.
    1. Chiu YT, Han HY, Leung SC, Yuen HF, Chau CW, Guo Z, Qiu Y, Chan KW, Wang X, Wong YC, Ling MT. CDC25A functions as a novel Ar corepressor in prostate cancer cells. J Mol Biol. 2009;385:446–456. doi: 10.1016/j.jmb.2008.10.070.
    1. Chen S, Xu Y, Yuan X, Bubley GJ, Balk SP. Androgen receptor phosphorylation and stabilization in prostate cancer by cyclin-dependent kinase 1. Proc Natl Acad Sci U S A. 2006;103:15969–15974. doi: 10.1073/pnas.0604193103.
    1. Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, Shi EY, Stumpf CR, Christensen C, Bonham MJ, Wang S, Ren P, Martin M, Jessen K, Feldman ME, Weissman JS, Shokat KM, Rommel C, Ruggero D. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature. 2012;485:55–61. doi: 10.1038/nature10912.
    1. Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat Rev Cancer. 2003;3:179–192. doi: 10.1038/nrc1015.
    1. Pelicano H, Xu RH, Du M, Feng L, Sasaki R, Carew JS, Hu Y, Ramdas L, Hu L, Keating MJ, Zhang W, Plunkett W, Huang P. Mitochondrial respiration defects in cancer cells cause activation of Akt survival pathway through a redox-mediated mechanism. J Cell Biol. 2006;175:913–923. doi: 10.1083/jcb.200512100.
    1. Rubin MA, Maher CA, Chinnaiyan AM. Common gene rearrangements in prostate cancer. J Clin Oncol. 2011;29:3659–3668. doi: 10.1200/JCO.2011.35.1916.
    1. McPherson A, Hormozdiari F, Zayed A, Giuliany R, Ha G, Sun MG, Griffith M, Heravi Moussavi A, Senz J, Melnyk N, Pacheco M, Marra MA, Hirst M, Nielsen TO, Sahinalp SC, Huntsman D, Shah SP. DeFuse: an algorithm for gene fusion discovery in tumor RNA-Seq data. PLoS Comput Biol. 2011;7:e1001138. doi: 10.1371/journal.pcbi.1001138.
    1. Krohn A, Seidel A, Burkhardt L, Bachmann F, Mader M, Grupp K, Eichenauer T, Becker A, Adam M, Graefen M, Huland H, Kurtz S, Steurer S, Tsourlakis MC, Minner S, Michl U, Schlomm T, Sauter G, Simon R, Sirma H. Recurrent deletion of 3p13 targets multiple tumour suppressor genes and defines a distinct subgroup of aggressive ERG fusion-positive prostate cancers. J Pathol. 2013;231:130–141. doi: 10.1002/path.4223.
    1. Osborne MJ, Volpon L, Kornblatt JA, Culjkovic-Kraljacic B, Baguet A, Borden KL. eIF4E3 acts as a tumor suppressor by utilizing an atypical mode of methyl-7-guanosine cap recognition. Proc Natl Acad Sci U S A. 2013;110:3877–3882. doi: 10.1073/pnas.1216862110.
    1. Feng S, Dakhova O, Creighton CJ, Ittmann M. Endocrine fibroblast growth factor FGF19 promotes prostate cancer progression. Cancer Res. 2013;73:2551–2562. doi: 10.1158/0008-5472.CAN-12-4108.
    1. Bookout AL, de Groot MH, Owen BM, Lee S, Gautron L, Lawrence HL, Ding X, Elmquist JK, Takahashi JS, Mangelsdorf DJ, Kliewer SA. FGF21 regulates metabolism and circadian behavior by acting on the nervous system. Nat Med. 2013;19:1147–1152. doi: 10.1038/nm.3249.
    1. Darby S, Cross SS, Brown NJ, Hamdy FC, Robson CN. BMP-6 over-expression in prostate cancer is associated with increased Id-1 protein and a more invasive phenotype. J Pathol. 2008;214:394–404. doi: 10.1002/path.2292.
    1. Nakayama KI, Nakayama K. Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer. 2006;6:369–381. doi: 10.1038/nrc1881.
    1. Sun Y. E3 ubiquitin ligases as cancer targets and biomarkers. Neoplasia. 2006;8:645–654. doi: 10.1593/neo.06376.
    1. Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ, Pleasance ED, Lau KW, Beare D, Stebbings LA, McLaren S, Lin ML, McBride DJ, Varela I, Nik-Zainal S, Leroy C, Jia M, Menzies A, Butler AP, Teague JW, Quail MA, Burton J, Swerdlow H, Carter NP, Morsberger LA, Iacobuzio-Donahue C, Follows GA, Green AR, Flanagan AM, Stratton MR, et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell. 2011;144:27–40. doi: 10.1016/j.cell.2010.11.055.
    1. McBride DJ, Etemadmoghadam D, Cooke SL, Alsop K, George J, Butler A, Cho J, Galappaththige D, Greenman C, Howarth KD, Lau KW, Ng CK, Raine K, Teague J, Wedge DC, Cancer Study Group AO. Caubit X, Stratton MR, Brenton JD, Campbell PJ, Futreal PA, Bowtell DD. Tandem duplication of chromosomal segments is common in ovarian and breast cancer genomes. J Pathol. 2012;227:446–455. doi: 10.1002/path.4042.
    1. Ng CK, Cooke SL, Howe K, Newman S, Xian J, Temple J, Batty EM, Pole JC, Langdon SP, Edwards PA, Brenton JD. The role of tandem duplicator phenotype in tumour evolution in high-grade serous ovarian cancer. J Pathol. 2012;226:703–712. doi: 10.1002/path.3980.
    1. Goel HL, Chang C, Pursell B, Leav I, Lyle S, Xi HS, Hsieh CC, Adisetiyo H, Roy-Burman P, Coleman IM, Nelson PS, Vessella RL, Davis RJ, Plymate SR, Mercurio AM. VEGF/neuropilin-2 regulation of Bmi-1 and consequent repression of IGF-IR define a novel mechanism of aggressive prostate cancer. Cancer Discovery. 2012;2:906–921. doi: 10.1158/-12-0085.
    1. Wade M, Li YC, Wahl GM. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nat Rev Cancer. 2013;13:83–96. doi: 10.1038/nrc3430.
    1. Stephens PJ, McBride DJ, Lin ML, Varela I, Pleasance ED, Simpson JT, Stebbings LA, Leroy C, Edkins S, Mudie LJ, Greenman CD, Jia M, Latimer C, Teague JW, Lau KW, Burton J, Quail MA, Swerdlow H, Churcher C, Natrajan R, Sieuwerts AM, Martens JW, Silver DP, Langerod A, Russnes HE, Foekens JA, Reis-Filho JS, Vant Veer L, Richardson AL, Borresen-Dale AL, et al. Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature. 2009;462:1005–1010. doi: 10.1038/nature08645.
    1. Pflueger D, Terry S, Sboner A, Habegger L, Esgueva R, Lin PC, Svensson MA, Kitabayashi N, Moss BJ, MacDonald TY, Cao X, Barrette T, Tewari AK, Chee MS, Chinnaiyan AM, Rickman DS, Demichelis F, Gerstein MB, Rubin MA. Discovery of non-ETS gene fusions in human prostate cancer using next-generation RNA sequencing. Genome Res. 2011;21:56–67. doi: 10.1101/gr.110684.110.
    1. Weischenfeldt J, Simon R, Feuerbach L, Schlangen K, Weichenhan D, Minner S, Wuttig D, Warnatz HJ, Stehr H, Rausch T, Jager N, Gu L, Bogatyrova O, Stutz AM, Claus R, Eils J, Eils R, Gerhauser C, Huang PH, Hutter B, Kabbe R, Lawerenz C, Radomski S, Bartholomae CC, Falth M, Gade S, Schmidt M, Amschler N, Hass T, Galal R, et al. Integrative genomic analyses reveal an androgen-driven somatic alteration landscape in early-onset prostate cancer. Cancer Cell. 2013;23:159–170. doi: 10.1016/j.ccr.2013.01.002.
    1. Palanisamy N, Ateeq B, Kalyana-Sundaram S, Pflueger D, Ramnarayanan K, Shankar S, Han B, Cao Q, Cao X, Suleman K, Kumar-Sinha C, Dhanasekaran SM, Chen YB, Esgueva R, Banerjee S, LaFargue CJ, Siddiqui J, Demichelis F, Moeller P, Bismar TA, Kuefer R, Fullen DR, Johnson TM, Greenson JK, Giordano TJ, Tan P, Tomlins SA, Varambally S, Rubin MA, Maher CA, et al. Rearrangements of the RAF kinase pathway in prostate cancer, gastric cancer and melanoma. Nat Med. 2010;16:793–798. doi: 10.1038/nm.2166.
    1. Tomlins SA, Rhodes DR, Yu J, Varambally S, Mehra R, Perner S, Demichelis F, Helgeson BE, Laxman B, Morris DS, Cao Q, Cao X, Andren O, Fall K, Johnson L, Wei JT, Shah RB, Al-Ahmadie H, Eastham JA, Eggener SE, Fine SW, Hotakainen K, Stenman UH, Tsodikov A, Gerald WL, Lilja H, Reuter VE, Kantoff PW, Scardino PT, Rubin MA, et al. The role of SPINK1 in ETS rearrangement-negative prostate cancers. Cancer Cell. 2008;13:519–528. doi: 10.1016/j.ccr.2008.04.016.
    1. Matsumoto H, Yamamoto Y, Shiota M, Kuruma H, Beraldi E, Matsuyama H, Zoubeidi A, Gleave M. Cotargeting androgen receptor and clusterin delays castrate-resistant prostate cancer progression by inhibiting adaptive stress response and AR stability. Cancer Res. 2013;73:5206–5217. doi: 10.1158/0008-5472.CAN-13-0359.
    1. Lapuk AV, Wu C, Wyatt AW, McPherson A, McConeghy BJ, Brahmbhatt S, Mo F, Zoubeidi A, Anderson S, Bell RH, Haegert A, Shukin R, Wang Y, Fazli L, Hurtado-Coll A, Jones EC, Hach F, Hormozdiari F, Hajiresouliha I, Boutros PC, Bristow RG, Zhao Y, Marra MA, Fanjul A, Maher CA, Chinnaiyan AM, Rubin MA, Beltran H, Sahinalp SC, Gleave ME, et al. From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J Pathol. 2012;227:286–297. doi: 10.1002/path.4047.
    1. Wu C, Wyatt AW, Lapuk AV, McPherson A, McConeghy BJ, Bell RH, Anderson S, Haegert A, Brahmbhatt S, Shukin R, Mo F, Li E, Fazli L, Hurtado-Coll A, Jones EC, Butterfield YS, Hach F, Hormozdiari F, Hajirasouliha I, Boutros PC, Bristow RG, Jones SJ, Hirst M, Marra MA, Maher CA, Chinnaiyan AM, Sahinalp SC, Gleave ME, Volik SV, Collins CC. Integrated genome and transcriptome sequencing identifies a novel form of hybrid and aggressive prostate cancer. J Pathol. 2012;227:53–61. doi: 10.1002/path.3987.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics. 2010;26:589–595. doi: 10.1093/bioinformatics/btp698.
    1. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics. 2009;25:1105–1111. doi: 10.1093/bioinformatics/btp120.
    1. Barnett DW, Garrison EK, Quinlan AR, Strömberg MP, Marth GT. BamTools: a C++ API and toolkit for analyzing and managing BAM files. Bioinformatics. 2011;27:1691–1692. doi: 10.1093/bioinformatics/btr174.
    1. Picard. []
    1. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, Philippakis AA, Del Angel G, Rivas MA, Hanna M. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43:491–498. doi: 10.1038/ng.806.
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110.
    1. Li H. Improving SNP discovery by base alignment quality. Bioinformatics. 2011;27:1157–1158. doi: 10.1093/bioinformatics/btr076.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603.
    1. González-Pérez A, López-Bigas N. Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel. Am J Hum Genet. 2011;88:440–449. doi: 10.1016/j.ajhg.2011.03.004.
    1. Futreal PA, Coin L, Marshall M, Down T, Hubbard T, Wooster R, Rahman N, Stratton MR. A census of human cancer genes. Nat Rev Cancer. 2004;4:177–183. doi: 10.1038/nrc1299.
    1. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546–1558. doi: 10.1126/science.1235122.
    1. Anders S, Huber W. Differential expression analysis for sequence count data. Genome Biol. 2010;11:R106. doi: 10.1186/gb-2010-11-10-r106.
    1. Rosner B. Percentage points for a generalized Esd many-outlier procedure. Technometrics. 1983;25:165–172. doi: 10.1080/00401706.1983.10487848.
    1. Ingenuity Pathway Analysis. []
    1. McPherson A, Wu C, Wyatt AW, Shah S, Collins C, Sahinalp SC. nFuse: Discovery of complex genomic rearrangements in cancer using high-throughput sequencing. Genome Res. 2012;22:2250–2261. doi: 10.1101/gr.136572.111.
    1. Mehra R, Tomlins SA, Yu J, Cao X, Wang L, Menon A, Rubin MA, Pienta KJ, Shah RB, Chinnaiyan AM. Characterization of TMPRSS2-ETS gene aberrations in androgen-independent metastatic prostate cancer. Cancer Res. 2008;68:3584–3590. doi: 10.1158/0008-5472.CAN-07-6154.

Source: PubMed

3
Předplatit