Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution

Rosalie Fisher, Stuart Horswell, Andrew Rowan, Maximilian P Salm, Elza C de Bruin, Sakshi Gulati, Nicholas McGranahan, Mark Stares, Marco Gerlinger, Ignacio Varela, Andrew Crockford, Francesco Favero, Virginie Quidville, Fabrice André, Carolina Navas, Eva Grönroos, David Nicol, Steve Hazell, David Hrouda, Tim O'Brien, Nik Matthews, Ben Phillimore, Sharmin Begum, Adam Rabinowitz, Jennifer Biggs, Paul A Bates, Neil Q McDonald, Gordon Stamp, Bradley Spencer-Dene, James J Hsieh, Jianing Xu, Lisa Pickering, Martin Gore, James Larkin, Charles Swanton, Rosalie Fisher, Stuart Horswell, Andrew Rowan, Maximilian P Salm, Elza C de Bruin, Sakshi Gulati, Nicholas McGranahan, Mark Stares, Marco Gerlinger, Ignacio Varela, Andrew Crockford, Francesco Favero, Virginie Quidville, Fabrice André, Carolina Navas, Eva Grönroos, David Nicol, Steve Hazell, David Hrouda, Tim O'Brien, Nik Matthews, Ben Phillimore, Sharmin Begum, Adam Rabinowitz, Jennifer Biggs, Paul A Bates, Neil Q McDonald, Gordon Stamp, Bradley Spencer-Dene, James J Hsieh, Jianing Xu, Lisa Pickering, Martin Gore, James Larkin, Charles Swanton

Abstract

Background: Genomic analysis of multi-focal renal cell carcinomas from an individual with a germline VHL mutation offers a unique opportunity to study tumor evolution.

Results: We perform whole exome sequencing on four clear cell renal cell carcinomas removed from both kidneys of a patient with a germline VHL mutation. We report that tumors arising in this context are clonally independent and harbour distinct secondary events exemplified by loss of chromosome 3p, despite an identical genetic background and tissue microenvironment. We propose that divergent mutational and copy number anomalies are contingent upon the nature of 3p loss of heterozygosity occurring early in tumorigenesis. However, despite distinct 3p events, genomic, proteomic and immunohistochemical analyses reveal evidence for convergence upon the PI3K-AKT-mTOR signaling pathway. Four germline tumors in this young patient, and in a second, older patient with VHL syndrome demonstrate minimal intra-tumor heterogeneity and mutational burden, and evaluable tumors appear to follow a linear evolutionary route, compared to tumors from patients with sporadic clear cell renal cell carcinoma.

Conclusions: In tumors developing from a germline VHL mutation, the evolutionary principles of contingency and convergence in tumor development are complementary. In this small set of patients with early stage VHL-associated tumors, there is reduced mutation burden and limited evidence of intra-tumor heterogeneity.

Figures

Figure 1
Figure 1
Multi-focal renal cell carcinoma tumors in a patient with VHL disease. Coronal sections from computed tomography scans show the spatial orientation of the tumors within the kidneys. (A) A macroscopically single tumor in the right kidney, but defined by exome sequencing as two tumors (one and two). (B) Two separate tumors in the left kidney, tumors three and four. Cysts typical of VHL disease are also present in both kidneys.
Figure 2
Figure 2
Intra-tumor heterogeneity in four germline tumors. (A,B) Heatmaps show the regional distribution of all non-silent mutations; presence (blue) or absence (grey) of each mutation is indicated for every tumor region in tumors from the right (A) and left kidneys (B). Whole exome sequencing defines four distinct tumors. In each, tumor evolution is linear rather than branched.
Figure 3
Figure 3
Copy number analysis in four germline tumors. (A) Copy number profiles of representative samples from each of the four tumors, with integer copy number on the x-axis. Loss of chromosome 3p is ubiquitous. (B) The chromosome 3p breakpoint locations for the four tumors. Each y-axis is logR, and the x-axis represents position along chromosome 3 in hg19. An ‘X’ near the x-axis marks the approximate position of the centromere.
Figure 4
Figure 4
Two distinct activating mutations in mTOR. Left: a schematic of the mTOR protein structure (cyan) in complex with mLST8 (dark blue) (PDB code 4JSP). Key residues and structural features are highlighted close to mTOR mutations relevant to the current study. The FRB and FAT domains are also labeled together with the FATC sub-domain. Right: a close-up of regions in proximity to the mTOR mutation site L2427 abutting the nucleotide-binding cleft and activation loop (upper right panel). Lower right panel shows the environment surrounding residue T1652 in the FAT domain.
Figure 5
Figure 5
Convergence upon the PI3K-AKT-mTOR pathway. Graphs showing the ratio of phosphorylated to total protein for intra-cellular kinases of the PI3-AKT-mTOR pathway. For tumors one, two and three, more than one region from each tumor was analyzed.

References

    1. Seizinger BR, Rouleau GA, Ozelius LJ, Lane AH, Farmer GE, Lamiell JM, Haines J, Yuen JW, Collins D, Majoor-Krakauer D, Bonner T, Mathew C, Rubenstein A, Halperin J, McConkie-Rosell A, Green JS, Trofatter JA, Ponder BA, Eierman L, Bowmer MI, Schimke R, Oostra B, Aronin N, Smith DI, Drabkin H, Waziri MH, Hobbs WJ, Martuza RL, Conneally PM, Hsia YE, et al. Von Hippel-Lindau disease maps to the region of chromosome 3 associated with renal cell carcinoma. Nature. 1988;332:268–269. doi: 10.1038/332268a0.
    1. Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, Stackhouse T, Kuzmin I, Modi W, Geil L. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science. 1993;260:1317–1320. doi: 10.1126/science.8493574.
    1. Knudson AG., Jr Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A. 1971;68:820–823. doi: 10.1073/pnas.68.4.820.
    1. Nickerson ML, Jaeger E, Shi Y, Durocher JA, Mahurkar S, Zaridze D, Matveev V, Janout V, Kollarova H, Bencko V, Navratilova M, Szeszenia-Dabrowska N, Mates D, Mukeria A, Holcatova I, Schmidt LS, Toro JR, Karami S, Hung R, Gerard GF, Linehan WM, Merino M, Zbar B, Boffetta P, Brennan P, Rothman N, Chow WH, Waldman FM, Moore LE. Improved identification of von Hippel-Lindau gene alterations in clear cell renal tumors. Clin Cancer Res. 2008;14:4726–4734. doi: 10.1158/1078-0432.CCR-07-4921.
    1. Moore LE, Nickerson ML, Brennan P, Toro JR, Jaeger E, Rinsky J, Han SS, Zaridze D, Matveev V, Janout V, Kollarova H, Bencko V, Navratilova M, Szeszenia-Dabrowska N, Mates D, Schmidt LS, Lenz P, Karami S, Linehan WM, Merino M, Chanock S, Boffetta P, Chow WH, Waldman FM, Rothman N. Von Hippel-Lindau (VHL) inactivation in sporadic clear cell renal cancer: associations with germline VHL polymorphisms and etiologic risk factors. PLoS Genet. 2011;7:e1002312. doi: 10.1371/journal.pgen.1002312.
    1. Cancer Genome Atlas Research Network Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499:43–49. doi: 10.1038/nature12222.
    1. Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, Shimamura T, Sato-Otsubo A, Nagae G, Suzuki H, Nagata Y, Yoshida K, Kon A, Suzuki Y, Chiba K, Tanaka H, Niida A, Fujimoto A, Tsunoda T, Morikawa T, Maeda D, Kume H, Sugano S, Fukayama M, Aburatani H, Sanada M, Miyano S, Homma Y, Ogawa S. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013;45:860–867. doi: 10.1038/ng.2699.
    1. Martinez P, Birkbak NJ, Gerlinger M, McGranahan N, Burrell RA, Rowan AJ, Joshi T, Fisher R, Larkin J, Szallasi Z, Swanton C. Parallel evolution of tumour subclones mimics diversity between tumours. J Pathol. 2013;230:356–364. doi: 10.1002/path.4214.
    1. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46:225–233. doi: 10.1038/ng.2891.
    1. Zbar B, Brauch H, Talmadge C, Linehan M. Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma. Nature. 1987;327:721–724. doi: 10.1038/327721a0.
    1. Gerlinger M, Rowan A, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Mcdonald N, Butler A, Jones D, Raine K, Santos C, Varela I, Nohadani M, Eklund A, Spencer–Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–893. doi: 10.1056/NEJMoa1113205.
    1. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, Davies H, Jones D, Lin ML, Teague J, Bignell G, Butler A, Cho J, Dalgliesh GL, Galappaththige D, Greenman C, Hardy C, Jia M, Latimer C, Lau KW, Marshall J, McLaren S, Menzies A, Mudie L, Stebbings L, Largaespada DA, Wessels LF, Richard S, Kahnoski RJ, Anema J, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469:539–542. doi: 10.1038/nature09639.
    1. Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H, Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck G, Campbell PJ, Forbes S, Jia M, Jones D, Knott H, Kok CY, Lau KW, Leroy C, Lin ML, McBride DJ, Maddison M, Maguire S, McLay K, Menzies A, Mironenko T, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. 2010;463:360–363. doi: 10.1038/nature08672.
    1. Guo G, Gui Y, Gao S, Tang A, Hu X, Huang Y, Jia W, Li Z, He M, Sun L, Song P, Sun X, Zhao X, Yang S, Liang C, Wan S, Zhou F, Chen C, Zhu J, Li X, Jian M, Zhou L, Ye R, Huang P, Chen J, Jiang T, Liu X, Wang Y, Zou J, Jiang Z, et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nat Genet. 2012;44:17–19. doi: 10.1038/ng.1014.
    1. Larkin J, Goh XY, Vetter M, Pickering L, Swanton C. Epigenetic regulation in RCC: opportunities for therapeutic intervention? Nat Rev Urol. 2012;9:147–155. doi: 10.1038/nrurol.2011.236.
    1. Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–28. doi: 10.1126/science.959840.
    1. Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501:346–354. doi: 10.1038/nature12626.
    1. Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–313. doi: 10.1038/nature10762.
    1. Gould SJ. Wonderful Life: The Burgess Shale and the Nature of History. New York: Norton; 1989.
    1. Conway Morris S. Life’s Solution: Inevitable Humans in a Lonely Universe. New York: Cambridge University Press; 2003.
    1. Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack T, Laird PW, Onofrio RC, Winckler W, Weir BA, Beroukhim R, Pellman D, Levine DA, Lander ES, Meyerson M, Getz G. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 2012;30:413–421. doi: 10.1038/nbt.2203.
    1. Gordon MS, Hussey M, Nagle RB, Lara PN, Jr, Mack PC, Dutcher J, Samlowski W, Clark JI, Quinn DI, Pan CX, Crawford D. Phase II study of erlotinib in patients with locally advanced or metastatic papillary histology renal cell cancer: SWOG S0317. J Clin Oncol. 2009;27:5788–5793. doi: 10.1200/JCO.2008.18.8821.
    1. Maher ER, Yates JR, Harries R, Benjamin C, Harris R, Moore AT, Ferguson-Smith MA. Clinical features and natural history of von Hippel-Lindau disease. Q J Med. 1990;77:1151–1163. doi: 10.1093/qjmed/77.2.1151.
    1. Urano J, Sato T, Matsuo T, Otsubo Y, Yamamoto M, Tamanoi F. Point mutations in TOR confer Rheb-independent growth in fission yeast and nutrient-independent mammalian TOR signaling in mammalian cells. Proc Natl Acad Sci U S A. 2007;104:3514–3519. doi: 10.1073/pnas.0608510104.
    1. Ohne Y, Takahara T, Hatakeyama R, Matsuzaki T, Noda M, Mizushima N, Maeda T. Isolation of hyperactive mutants of mammalian target of rapamycin. J Biol Chem. 2008;283:31861–31870. doi: 10.1074/jbc.M801546200.
    1. Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, Jordan A, Beck AH, Sabatini DM. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov. 2014;4:554–563. doi: 10.1158/-13-0929.
    1. Murugan AK, Alzahrani A, Xing M. Mutations in critical domains confer the human mTOR gene strong tumorigenicity. J Biol Chem. 2013;288:6511–6521. doi: 10.1074/jbc.M112.399485.
    1. Voss MH, Hakimi AA, Pham CG, Brannon AR, Chen YB, Cunha LF, Akin O, Liu H, Takeda S, Scott SN, Socci ND, Viale A, Schultz N, Sander C, Reuter VE, Russo P, Cheng EH, Motzer RJ, Berger MF, Hsieh JJ. Tumor genetic analyses of patients with metastatic renal cell carcinoma and extended benefit from mTOR inhibitor therapy. Clin Cancer Res. 2014;20:1955–1964. doi: 10.1158/1078-0432.CCR-13-2345.
    1. Wagle N, Grabiner BC, Van Allen EM, Hodis E, Jacobus S, Supko JG, Stewart M, Choueiri TK, Gandhi L, Cleary JM, Elfiky AA, Taplin ME, Stack EC, Signoretti S, Loda M, Shapiro GI, Sabatini DM, Lander ES, Gabriel SB, Kantoff PW, Garraway LA, Rosenberg JE. Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. Cancer Discov. 2014;4:546–553. doi: 10.1158/-13-0353.
    1. Nawroth R, Stellwagen F, Schulz WA, Stoehr R, Hartmann A, Krause BJ, Gschwend JE, Retz M. S6K1 and 4E-BP1 are independent regulated and control cellular growth in bladder cancer. PLoS One. 2011;6:e27509. doi: 10.1371/journal.pone.0027509.
    1. She QB, Halilovic E, Ye Q, Zhen W, Shirasawa S, Sasazuki T, Solit DB, Rosen N. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell. 2010;18:39–51. doi: 10.1016/j.ccr.2010.05.023.
    1. Liang H, Cheung LW, Li J, Ju Z, Yu S, Stemke-Hale K, Dogruluk T, Lu Y, Liu X, Gu C, Guo W, Scherer SE, Carter H, Westin SN, Dyer MD, Verhaak RG, Zhang F, Karchin R, Liu CG, Lu KH, Broaddus RR, Scott KL, Hennessy BT, Mills GB. Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer. Genome Res. 2012;22:2120–2129. doi: 10.1101/gr.137596.112.
    1. Li L, Shen C, Nakamura E, Ando K, Signoretti S, Beroukhim R, Cowley GS, Lizotte P, Liberzon E, Bair S, Root DE, Tamayo P, Tsherniak A, Cheng SC, Tabak B, Jacobsen A, Hakimi AA, Schultz N, Ciriello G, Sander C, Hsieh JJ, Kaelin WG., Jr SQSTM1 is a pathogenic target of 5q copy number gains in kidney cancer. Cancer Cell. 2013;24:738–750. doi: 10.1016/j.ccr.2013.10.025.
    1. He X, Wang J, Messing EM, Wu G. Regulation of receptor for activated C kinase 1 protein by the von Hippel-Lindau tumor suppressor in IGF-I-induced renal carcinoma cell invasiveness. Oncogene. 2011;30:535–547. doi: 10.1038/onc.2010.427.
    1. Duran A, Amanchy R, Linares JF, Joshi J, Abu-Baker S, Porollo A, Hansen M, Moscat J, Diaz-Meco MT. p62 is a key regulator of nutrient sensing in the mTORC1 pathway. Mol Cell. 2011;44:134–146. doi: 10.1016/j.molcel.2011.06.038.
    1. Mazza S, Maffucci T. Class II phosphoinositide 3-kinase C2alpha: what we learned so far. Int J Biochem Mol Biol. 2011;2:168–182.
    1. Tibolla G, Pineiro R, Chiozzotto D, Mavrommati I, Wheeler AP, Norata GD, Catapano AL, Maffucci T, Falasca M. Class II phosphoinositide 3-kinases contribute to endothelial cells morphogenesis. PLoS One. 2013;8:e53808. doi: 10.1371/journal.pone.0053808.
    1. Beroukhim R, Brunet JP, Di Napoli A, Mertz KD, Seeley A, Pires MM, Linhart D, Worrell RA, Moch H, Rubin MA, Sellers WR, Meyerson M, Linehan WM, Kaelin WG, Jr, Signoretti S. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer Res. 2009;69:4674–4681. doi: 10.1158/0008-5472.CAN-09-0146.
    1. Whyte LL. Internal factors in evolution. Acta Biotheor. 1965;17:33–48. doi: 10.1007/BF01602054.
    1. Waddington CH. Canalization of development and the inheritance of acquired characters. Nature. 1942;3811:563–565. doi: 10.1038/150563a0.
    1. Beatty J. Replaying life’s tape. J Philosophy. 2006;CIII:336–362.
    1. Travisano M, Mongold JA, Bennett AF, Lenski RE. Experimental tests of the roles of adaptation, chance, and history in evolution. Science. 1995;267:87–90. doi: 10.1126/science.7809610.
    1. Greaves MF, Maia AT, Wiemels JL, Ford AM. Leukemia in twins: lessons in natural history. Blood. 2003;102:2321–2333. doi: 10.1182/blood-2002-12-3817.
    1. Cazzaniga G, van Delft FW, Lo Nigro L, Ford AM, Score J, Iacobucci I, Mirabile E, Taj M, Colman SM, Biondi A, Greaves M. Developmental origins and impact of BCR-ABL1 fusion and IKZF1 deletions in monozygotic twins with Ph + acute lymphoblastic leukemia. Blood. 2011;118:5559–5564. doi: 10.1182/blood-2011-07-366542.
    1. Ma Y, Dobbins SE, Sherborne AL, Chubb D, Galbiati M, Cazzaniga G, Micalizzi C, Tearle R, Lloyd AL, Hain R, Greaves M, Houlston RS. Developmental timing of mutations revealed by whole-genome sequencing of twins with acute lymphoblastic leukemia. Proc Natl Acad Sci U S A. 2013;110:7429–7433. doi: 10.1073/pnas.1221099110.
    1. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G, Van Loo P, Yoon CJ, Ellis P, Wedge DC, Pellagatti A, Shlien A, Groves MJ, Forbes SA, Raine K, Hinton J, Mudie LJ, McLaren S, Hardy C, Latimer C, Della Porta MG, O'Meara S, Ambaglio I, Galli A, Butler AP, Walldin G, Teague JW, Quek L, Sternberg A, Gambacorti-Passerini C, Cross NC, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122:3616–3627. doi: 10.1182/blood-2013-08-518886.
    1. Blount ZD, Borland CZ, Lenski RE. Historical contingency and the evolution of a key innovation in an experimental population of Escherichia coli. Proc Natl Acad Sci U S A. 2008;105:7899–7906. doi: 10.1073/pnas.0803151105.
    1. Blount ZD, Barrick JE, Davidson CJ, Lenski RE. Genomic analysis of a key innovation in an experimental Escherichia coli population. Nature. 2012;489:513–518. doi: 10.1038/nature11514.
    1. Harms MJ, Thornton JW. Historical contingency and its biophysical basis in glucocorticoid receptor evolution. Nature. 2014;512:203–207. doi: 10.1038/nature13410.
    1. Lui VW, Hedberg ML, Li H, Vangara BS, Pendleton K, Zeng Y, Lu Y, Zhang Q, Du Y, Gilbert BR, Freilino M, Sauerwein S, Peyser ND, Xiao D, Diergaarde B, Wang L, Chiosea S, Seethala R, Johnson JT, Kim S, Duvvuri U, Ferris RL, Romkes M, Nukui T, Kwok-Shing Ng P, Garraway LA, Hammerman PS, Mills GB, Grandis JR. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 2013;3:761–769. doi: 10.1158/-13-0103.
    1. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP. Integrative genomics viewer. Nat Biotechnol. 2011;29:24–26. doi: 10.1038/nbt.1754.
    1. Nakamura K, Oshima T, Morimoto T, Ikeda S, Yoshikawa H, Shiwa Y, Ishikawa S, Linak MC, Hirai A, Takahashi H, Altaf-Ul-Amin M, Ogasawara N, Kanaya S. Sequence-specific error profile of Illumina sequencers. Nucleic Acids Res. 2011;39:e90. doi: 10.1093/nar/gkr344.
    1. Liu X, Jian X, Boerwinkle E. dbNSFP v2.0: a database of human non-synonymous SNVs and their functional predictions and annotations. Hum Mutat. 2013;34:e2393–e2402. doi: 10.1002/humu.22376.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: Functional annotation of genetic variants from next-generation sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603.
    1. Ye K, Schulz MH, Long Q, Apweiler R, Ning Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics. 2009;25:2865–2871. doi: 10.1093/bioinformatics/btp394.
    1. Ion Ampliseq Designer. [].
    1. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L, Wilson RK. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576. doi: 10.1101/gr.129684.111.
    1. Nilsen G, Liestol K, Van Loo P, Moen Vollan HK, Eide MB, Rueda OM, Chin SF, Russell R, Baumbusch LO, Caldas C, Borresen-Dale AL, Lingjaerde OC. Copynumber: Efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics. 2012;13:591. doi: 10.1186/1471-2164-13-591.
    1. Pihur V, Datta S, Datta S. RankAggreg, an R package for weighted rank aggregation. BMC Bioinformatics. 2009;10:62. doi: 10.1186/1471-2105-10-62.
    1. TCGA Copy Number Portal. []
    1. Landau DA, Carter SL, Stojanov P, McKenna A, Stevenson K, Lawrence MS, Sougnez C, Stewart C, Sivachenko A, Wang L, Wan Y, Zhang W, Shukla SA, Vartanov A, Fernandes SM, Saksena G, Cibulskis K, Tesar B, Gabriel S, Hacohen N, Meyerson M, Lander ES, Neuberg D, Brown JR, Getz G, Wu CJ. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152:714–726. doi: 10.1016/j.cell.2013.01.019.
    1. Fraley C, Raftery AE, Murphy TB, Scrucca L: mclust version 4 for R: Normal Mixture Modeling for Model-Based Clustering, Classification and Density Estimation. University of Washington; 2012.
    1. Van Loo P, Nordgard SH, Lingjaerde OC, Russnes HG, Rye IH, Sun W, Weigman VJ, Marynen P, Zetterberg A, Naume B, Perou CM, Borresen-Dale AL, Kristensen VN. Allele-specific copy number analysis of tumors. Proc Natl Acad Sci U S A. 2010;107:16910–16915. doi: 10.1073/pnas.1009843107.
    1. PyMOL. []
    1. Zhou X, Tan M, Stone Hawthorne V, Klos KS, Lan KH, Yang Y, Yang W, Smith TL, Shi D, Yu D. Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers. Clin Cancer Res. 2004;10:6779–6788. doi: 10.1158/1078-0432.CCR-04-0112.

Source: PubMed

3
구독하다