D. Hanahan and R. A. Weinberg, The Hallmarks of Cancer, Cell, vol.100, issue.1, pp.57-70, 2000.
DOI : 10.1016/S0092-8674(00)81683-9

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, pp.646-674, 2011.
DOI : 10.1016/j.cell.2011.02.013

P. Kaatsch, Epidemiology of childhood cancer, Cancer Treatment Reviews, vol.36, issue.4, pp.277-285, 2010.
DOI : 10.1016/j.ctrv.2010.02.003

M. Jiang, J. Stanke, and J. M. Lahti, The Connections Between Neural Crest Development and Neuroblastoma, Current topics in developmental biology, vol.94, pp.77-127, 2011.
DOI : 10.1016/B978-0-12-380916-2.00004-8

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3633592

A. D. Cohn, J. M. Pearson, and . Maris, International consensus for neuroblastoma molecular diagnostics: report from the International Neuroblastoma Risk Group (INRG) Biology Committee, British Journal of Cancer, vol.100, issue.9, pp.1471-1482, 2009.

J. J. Koster, D. A. Zwijnenburg, L. J. Peter-van-sluis, I. Valentijn, M. Van-der-ploeg et al., Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes, Nature, issue.7391, pp.483589-593, 2012.

J. M. Maris, M. J. Weiss, Y. Mosse, G. Hii, C. Guo et al., Evidence for a Hereditary Neuroblastoma Predisposition Locus at Chromosome 16p12?13, Cancer Research, issue.22, pp.626651-6658, 2002.

P. Yaël, M. Mossé, L. Laudenslager, K. A. Longo, A. Cole et al., Identification of ALK as a major familial neuroblastoma predisposition gene, Nature, issue.7215, pp.455930-935, 2008.

L. Tweddle, A. Chen, L. Humphreys, A. Turnbull, G. Bellini et al., Identification of different ALK mutations in a pair of neuroblastoma cell lines established at diagnosis and relapse, Oncotarget, issue.52, pp.787301-87311, 2016.

D. R. Lohmann, B. Brandt, W. Höpping, E. Passarge, and B. Horsthemke, The spectrum of RB1 germ-line mutations in hereditary retinoblastoma, American Journal of Human Genetics, vol.58, issue.5, pp.940-949, 1996.

K. Sippel, R. Fraioli, G. Smith, . Schalkoff, . Sutherland et al., Frequency of Somatic and Germ-Line Mosaicism in Retinoblastoma: Implications for Genetic Counseling, The American Journal of Human Genetics, vol.62, issue.3, pp.610-619, 1998.
DOI : 10.1086/301766

B. Canard and R. S. Sarfati, DNA polymerase fluorescent substrates with reversible 3???-tags, Gene, vol.148, issue.1, pp.1-6, 1994.
DOI : 10.1016/0378-1119(94)90226-7

H. Thorvaldsdottir, J. T. Robinson, and J. P. Mesirov, Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration, Briefings in Bioinformatics, vol.14, issue.2, pp.178-192, 2013.
DOI : 10.1093/bib/bbs017

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3603213

P. Dehal and J. L. Boore, Two Rounds of Whole Genome Duplication in the Ancestral Vertebrate, PLoS Biology, vol.18, issue.10, p.314, 2005.
DOI : 10.1371/journal.pbio.0030314.st001

C. Daniel, D. E. Koboldt, R. K. Larson, D. Wilson-andreas, and . Baxevanis, Using VarScan 2 for Germline Variant Calling and Somatic Mutation Detection, Current protocols in bioinformatics, vol.44, pp.15-19, 2013.

K. Cibulskis, M. S. Lawrence, S. L. Carter, A. Sivachenko, D. Jaffe et al., Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples, Nature Biotechnology, vol.27, issue.3, pp.31213-219, 2013.
DOI : 10.1093/bioinformatics/btr446

C. T. Saunders, W. S. Wong, S. Swamy, J. Becq, L. J. Murray et al., Strelka: accurate somatic small-variant calling from sequenced tumor???normal sample pairs, Bioinformatics, vol.28, issue.14, pp.281811-1817, 2012.
DOI : 10.1093/bioinformatics/bts271

A. Ronald and . Fisher, On the interpretation of ?2 from contingency tables, and the calculation of p, Journal of the Royal Statistical Society, pp.87-94, 1922.

B. Zeitouni, V. Boeva, I. Janoueix-lerosey, S. Loeillet, P. Legoix-né et al., SVDetect: a tool to identify genomic structural variations from paired-end and mate-pair sequencing data, Bioinformatics, vol.26, issue.15, pp.261895-1896, 2010.
DOI : 10.1093/bioinformatics/btq293

URL : https://hal.archives-ouvertes.fr/inserm-00508372

K. Chen, J. W. Wallis, M. D. Mclellan, D. E. Larson, J. M. Kalicki et al., BreakDancer: an algorithm for high-resolution mapping of genomic structural variation, Nature Methods, vol.453, issue.9, pp.677-681, 2009.
DOI : 10.1101/gr.074492.107

M. Tom and . Mitchell, Machine Learning. McGraw-Hill series in computer science, 1997.

W. Walter and . Piegorsch, Statistical data analytics: foundations for data mining, informatics, and knowledge discovery, 2015.

D. Matthew, R. Zeiler, and . Fergus, Visualizing and understanding convolutional networks, European conference on computer vision, pp.818-833, 2014.

H. William, . Wolberg, L. Olvi, and . Mangasarian, Multisurface method of pattern separation for medical diagnosis applied to breast cytology, Proceedings of the national academy of sciences, pp.9193-9196, 1990.

L. Olvi, . Mangasarian, W. Setiono, and . Wolberg, Pattern recognition via linear programming: Theory and application to medical diagnosis. Largescale numerical optimization, pp.22-31, 1990.

P. Kristin, . Bennett, L. Olvi, and . Mangasarian, Robust linear programming discrimination of two linearly inseparable sets. Optimization methods and software, pp.23-34, 1992.

L. Olvi, N. Mangasarian, . Street, H. William, and . Wolberg, Breast cancer diagnosis and prognosis via linear programming, Operations Research, vol.43, issue.4, pp.570-577, 1995.

J. Zhang, Selecting Typical Instances in Instance-Based Learning, Proceedings of the Ninth International Machine Learning Conference, pp.470-479, 1992.
DOI : 10.1016/B978-1-55860-247-2.50066-8

H. Joe and . Ward-jr, Hierarchical grouping to optimize an objective function, Journal of the American statistical association, vol.58, issue.301, pp.236-244, 1963.

S. Tuffery, Data Mining et Statistique Decisionnelle: L'intelligence des données, p.912492491, 2012.

S. Lloyd, Least squares quantization in PCM, IEEE Transactions on Information Theory, vol.28, issue.2, pp.129-137, 1982.
DOI : 10.1109/TIT.1982.1056489

URL : http://www.cs.toronto.edu/~roweis/csc2515-2006/readings/lloyd57.pdf

L. Kaufman and P. J. Rousseeuw, Finding groups in data: an introduction to cluster analysis. Wiley series in probability and mathematical statistics, 2005.
DOI : 10.1002/9780470316801

J. Peter and . Rousseeuw, Silhouettes: A graphical aid to the interpretation and validation of cluster analysis, Journal of Computational and Applied Mathematics, vol.20, pp.53-65, 1987.

C. Wang, R. Machiraju, and K. Huang, Breast cancer patient stratification using a molecular regularized consensus clustering method, Methods, vol.67, issue.3, pp.304-312, 2014.
DOI : 10.1016/j.ymeth.2014.03.005

A. Yates, . Pancreatic-cancer-genome-initiative, . Icgc-breast-cancer, I. Consortium, . Mmml-seq et al., Signatures of mutational processes in human cancer, Nature, issue.7463, pp.500415-421, 2013.

L. Martignetti, L. Calzone, E. Bonnet, E. Barillot, and A. Zinovyev, ROMA: Representation and Quantification of Module Activity from Target Expression Data, Frontiers in Genetics, vol.430, 2016.
DOI : 10.1016/j.bbrc.2012.12.043

M. Kimura, The Number of Heterozygous Nucleotide Sites Maintained in a Finite Population Due to Steady Flux of Mutations, Genetics, vol.61, issue.4, pp.893-903, 1969.

F. Tajima, Infinite-allele model and infinite-site model in population genetics, Journal of Genetics, vol.7, issue.1, p.27, 1996.
DOI : 10.1017/CBO9780511623486

V. Boeva, T. Popova, K. Bleakley, P. Chiche, J. Cappo et al., Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data, Bioinformatics, vol.28, issue.3, pp.28423-425, 2012.
DOI : 10.1093/bioinformatics/btr670

L. Oesper, A. Mahmoody, J. Benjamin, and . Raphael, THetA: inferring intra-tumor heterogeneity from high-throughput DNA sequencing data, Genome Biology, vol.14, issue.7, p.80, 2013.
DOI : 10.1093/nar/gkh103

URL : http://doi.org/10.1186/gb-2013-14-7-r80

G. Ha, A. Roth, J. Khattra, J. Ho, D. Yap et al., TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data, Genome Research, vol.11, issue.11, pp.241881-1893, 2014.
DOI : 10.1093/bioinformatics/btt416

F. Favero, T. Joshi, A. M. Marquard, N. J. Birkbak, M. Krzystanek et al., Sequenza: allele-specific copy number and mutation profiles from tumor sequencing data, Annals of Oncology, vol.26, issue.1, pp.64-70, 2015.
DOI : 10.1093/annonc/mdu479

URL : https://academic.oup.com/annonc/article-pdf/26/1/64/8869154/mdu479.pdf

. Sciclone, Inferring Clonal Architecture and Tracking the Spatial and Temporal Patterns of Tumor Evolution, PLoS Comput Biol, vol.10, issue.8, p.1003665, 2014.

A. Roth, J. Khattra, D. Yap, A. Wan, E. Laks et al., PyClone: statistical inference of clonal population structure in cancer, Nature Methods, vol.11, issue.4, pp.396-398, 2014.
DOI : 10.1093/bioinformatics/btp698

I. Hajirasouliha, A. Mahmoody, and B. J. Raphael, A combinatorial approach for analyzing intra-tumor heterogeneity from high-throughput sequencing data, Bioinformatics, vol.30, issue.12, pp.78-86, 2014.
DOI : 10.1093/bioinformatics/btu284

Y. Qiao, A. R. Quinlan, A. Amir, . Jazaeri, G. Roeland et al., SubcloneSeeker: a computational framework for reconstructing tumor clone structure for cancer variant interpretation and prioritization, Genome Biology, vol.17, issue.8, p.15443, 2014.
DOI : 10.1093/bioinformatics/17.10.977

N. Beerenwinkel, R. F. Schwarz, M. Gerstung, and F. Markowetz, Cancer Evolution: Mathematical Models and Computational Inference, Systematic Biology, vol.64, issue.1, pp.1-25, 2015.
DOI : 10.1093/sysbio/syu081

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4265145

W. Jiao, S. Vembu, G. Amit, L. Deshwar, Q. Stein et al., Inferring clonal evolution of tumors from single nucleotide somatic mutations, BMC Bioinformatics, vol.15, issue.1, p.35, 2014.
DOI : 10.1111/j.2041-210X.2011.00131.x

URL : http://doi.org/10.1186/1471-2105-15-35

H. Zare, J. Wang, A. Hu, K. Weber, J. Smith et al., Inferring Clonal Composition from Multiple Sections of a Breast Cancer, PLoS Computational Biology, vol.29, issue.7, p.1003703, 2014.
DOI : 10.1371/journal.pcbi.1003703.s004

F. Strino, F. Parisi, M. Micsinai, and Y. Kluger, TrAp: a tree approach for fingerprinting subclonal tumor composition, Nucleic Acids Research, vol.41, issue.17, p.165, 2013.
DOI : 10.1093/nar/gkt641

A. Fischer, I. Vazquez-garcía, C. J. Illingworth, and V. Mustonen, High-Definition Reconstruction of Clonal Composition in Cancer, Cell Reports, vol.7, issue.5, pp.1740-1752, 2014.
DOI : 10.1016/j.celrep.2014.04.055

E. Purdom, C. Ho, C. S. Grasso, M. J. Quist, R. J. Cho et al., Methods and challenges in timing chromosomal abnormalities within cancer samples, Bioinformatics, vol.29, issue.24, pp.293113-3120, 2013.
DOI : 10.1093/bioinformatics/btt546

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3842754

. Campbell, Estimation of rearrangement phylogeny for cancer genomes

R. F. Schwarz, A. Trinh, B. Sipos, J. D. Brenton, N. Goldman et al., Phylogenetic Quantification of Intra-tumour Heterogeneity, PLoS Computational Biology, vol.11, issue.6, p.1003535, 2014.
DOI : 10.1371/journal.pcbi.1003535.g006

E. Letouzé, Y. Allory, A. Marc, F. Bollet, F. Radvanyi et al., Analysis of the copy number profiles of several tumor samples from the same patient reveals the successive steps in tumorigenesis, Genome Biology, issue.7, pp.11-76, 2010.

S. Wendl, M. A. Heath, D. C. Watson, M. H. Link, W. D. Tomasson et al., Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing, Nature, issue.7382, pp.481506-510, 2012.

P. Eirew, A. Steif, J. Khattra, G. Ha, D. Yap et al., Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution, Nature, vol.208, issue.7539, pp.518422-426, 2015.
DOI : 10.1084/jem.20110105

K. M. Hardiman, P. J. Ulintz, R. Kuick, D. H. Hovelson, C. M. Gates et al., Intratumor Genetic Heterogeneity in Rectal Cancer investigation; a journal of technical methods and pathology, Laboratory, vol.96, issue.1, pp.4-15, 2016.

N. Andor, T. A. Graham, M. Jansen, L. C. Xia, C. A. Aktipis et al., Pan-cancer analysis of the extent and consequences of intratumor heterogeneity, Nature Medicine, vol.372, issue.1, pp.105-113, 2016.
DOI : 10.1038/nature12634

P. Lamy, I. Nordentoft, K. Birkenkamp-demtröder, M. Borg-houlberg-thomsen, P. Villesen et al., Paired Exome Analysis Reveals Clonal Evolution and Potential Therapeutic Targets in Urothelial Carcinoma, Cancer Research, vol.76, issue.19, pp.765894-5906, 2016.
DOI : 10.1158/0008-5472.CAN-16-0436

C. M. Bishop, Pattern recognition and machine learning. Information science and statistics, 2006.

F. Thomas, D. A. Eleveld, V. B. Oldridge, L. C. Koster, S. J. Daage et al., Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations, Nature Genetics, issue.8, pp.47864-871, 2015.

J. T. Robinson, H. Thorvaldsdóttir, W. Winckler, M. Guttman, E. S. Lander et al., Integrative genomics viewer, Nature Biotechnology, vol.306, issue.1, pp.24-26, 2011.
DOI : 10.1093/bioinformatics/btp472

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3346182

F. Vandin, P. Clay, E. Upfal, and B. J. Raphael, DISCOVERY OF MUTATED SUBNETWORKS ASSOCIATED WITH CLINICAL DATA IN CANCER, Biocomputing 2012, pp.55-66, 2011.
DOI : 10.1142/9789814366496_0006

M. Hofree, J. P. Shen, H. Carter, A. Gross, and T. Ideker, Network-based stratification of tumor mutations, Nature Methods, vol.39, issue.11, pp.1108-1115, 2013.
DOI : 10.1101/gr.229102. Article published online before print in May 2002

URL : http://www.nature.com/nmeth/journal/v10/n11/pdf/nmeth.2651.pdf

M. Le-morvan, A. Zinovyev, and J. Vert, NetNorM: Capturing cancer-relevant information in somatic exome mutation data with gene networks for cancer stratification and prognosis, PLOS Computational Biology, vol.4, issue.1, 2016.
DOI : 10.1371/journal.pcbi.1005573.s010

URL : https://hal.archives-ouvertes.fr/hal-01341856

P. Deveau, E. Barillot, V. Boeva, A. Zinovyev, and E. Bonnet, Calculating biological module enrichment or depletion and visualizing data on large-scale molecular maps with ACSNMineR and RNaviCell packages, The R Journal, vol.8, issue.2, pp.293-306, 2016.
DOI : 10.1101/064469

P. D. Thomas, M. J. Campbell, A. Kejariwal, H. Mi, B. Karlak et al., PANTHER: A Library of Protein Families and Subfamilies Indexed by Function, Genome Research, vol.13, issue.9, pp.2129-2141, 2003.
DOI : 10.1101/gr.772403

H. Mi, Q. Dong, A. Muruganujan, P. Gaudet, S. Lewis et al., PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium, Nucleic Acids Research, vol.38, issue.suppl_1, pp.204-210, 2010.
DOI : 10.1093/nar/gkp1019

URL : http://doi.org/10.1093/nar/gkp1019

B. T. Da-wei-huang, R. A. Sherman, and . Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature Protocols, vol.99, issue.1, pp.44-57, 2009.
DOI : 10.6026/97320630002428

B. T. Da-wei-huang, R. A. Sherman, and . Lempicki, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Research, vol.37, issue.1, pp.1-13, 2009.
DOI : 10.1093/nar/gkn923

C. Kimchi-sarfaty, J. M. Oh, I. Kim, Z. E. Sauna, A. M. Calcagno et al., A "Silent" Polymorphism in the MDR1 Gene Changes Substrate Specificity, Science, vol.315, issue.5811, pp.315525-528, 2007.
DOI : 10.1126/science.1135308

G. Loughran, M. Chou, I. P. Ivanov, I. Jungreis, M. Kellis et al., Evidence of efficient stop codon readthrough in four mammalian genes, Nucleic Acids Research, vol.42, issue.14, pp.8928-8938, 2014.
DOI : 10.1093/nar/gku608

T. Kalstrup and R. Blunck, Reinitiation at non-canonical start codons leads to leak expression when incorporating unnatural amino acids, Scientific Reports, vol.257, issue.1, p.11866, 2015.
DOI : 10.1016/S0896-6273(00)80143-9

URL : http://doi.org/10.1038/srep11866

P. Kumar, S. Henikoff, and P. C. Ng, Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm, Nature Protocols, vol.4, issue.8, pp.1073-1081, 2009.
DOI : 10.1101/gr.772403

C. Pauline, S. Ng, and . Henikoff, SIFT: predicting amino acid changes that affect protein function, Nucleic Acids Research, vol.31, issue.13, pp.3812-3814, 2003.

I. Adzhubei, D. M. Jordan, and S. R. Sunyaev, Predicting Functional Effect of Human Missense Mutations Using PolyPhen-2. Current protocols in human genetics, p.0

M. Levo and E. Segal, In pursuit of design principles of regulatory sequences, Nature Reviews Genetics, vol.49, issue.7, pp.453-468, 2014.
DOI : 10.1016/j.celrep.2013.03.014

E. Khurana, Y. Fu, V. Colonna, X. J. Mu, H. M. Kang et al., Integrative Annotation of Variants from 1092 Humans: Application to Cancer Genomics, Science, vol.342, issue.6154, p.3421235587, 2013.
DOI : 10.1186/1479-7364-5-6-709

Y. Fu, Z. Liu, S. Lou, J. Bedford, X. J. Mu et al., FunSeq2: a framework for prioritizing noncoding regulatory variants in cancer, Genome Biology, vol.28, issue.10, pp.15-2014
DOI : 10.1093/bioinformatics/bts368

F. Vandin, E. Upfal, and B. J. Raphael, Algorithms for Detecting Significantly Mutated Pathways in Cancer, Journal of Computational Biology, vol.18, issue.3, pp.507-522, 2011.
DOI : 10.1089/cmb.2010.0265

W. Qiu, J. Wu, E. M. Walsh, Y. Zhang, C. Chen et al., Retinoblastoma protein modulates gankyrin???MDM2 in regulation of p53 stability and chemosensitivity in cancer cells, Oncogene, vol.92, issue.29, pp.274034-4043, 2008.
DOI : 10.1038/onc.2008.43

H. Wu and G. Lozano, NF-kappa B activation of p53. A potential mechanism for suppressing cell growth in response to stress, Journal of Biological Chemistry, vol.269, issue.31, pp.20067-20074, 1994.

P. M. Flatt, L. J. Tang, C. D. Scatena, S. T. Szak, and J. A. Pietenpol, p53 Regulation of G2 Checkpoint Is Retinoblastoma Protein Dependent, Molecular and Cellular Biology, vol.20, issue.12, pp.4210-4223, 2000.
DOI : 10.1128/MCB.20.12.4210-4223.2000

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC85790/pdf

X. Liu, Y. Zhang, M. Tong, X. Liu, G. Luo et al., Identification of a small molecule 1,4-bis-[4-(3-phenoxy-propoxy)-but-2-ynyl]-piperazine as a novel inhibitor of the transcription factor p53, Acta Pharmacologica Sinica, vol.5, issue.6
DOI : 10.1016/j.stem.2009.11.003

H. Mi, A. Muruganujan, J. T. Casagrande, and P. D. Thomas, Large-scale gene function analysis with the PANTHER classification system, Nature Protocols, vol.5, issue.8, pp.1551-1566, 2013.
DOI : 10.1186/1471-2105-8-426

M. Kanehisa, S. Goto, Y. Sato, M. Furumichi, and M. Tanabe, KEGG for integration and interpretation of large-scale molecular data sets, Nucleic Acids Research, vol.40, issue.D1, p.988, 2011.
DOI : 10.1093/nar/gkr988

D. Nishimura and . Biocarta, BioCarta, Biotech Software & Internet Report, vol.2, issue.3, pp.117-120, 2001.
DOI : 10.1089/152791601750294344

I. Kuperstein, . Bonnet, . Ha-nguyen, . Cohen, . Viara et al., Atlas of Cancer Signalling Network: a systems biology resource for integrative analysis of cancer data with Google Maps, Oncogenesis, vol.1046, issue.7, p.160, 2015.
DOI : 10.1093/annonc/mdr304

A. Ronald and . Fisher, Statistical methods for research workers, 1934.

I. Rivals, L. Personnaz, L. Taing, and M. Potier, Enrichment or depletion of a GO category within a class of genes: which test?, Bioinformatics, vol.23, issue.4, pp.401-407, 2007.
DOI : 10.1093/bioinformatics/btl633

URL : https://hal.archives-ouvertes.fr/hal-00801557

A. Reiner, D. Yekutieli, and Y. Benjamini, Identifying differentially expressed genes using false discovery rate controlling procedures, Bioinformatics, vol.19, issue.3, pp.368-375, 2003.
DOI : 10.1093/bioinformatics/btf877

URL : https://academic.oup.com/bioinformatics/article-pdf/19/3/368/717620/btf877.pdf

H. Wickham, ggplot2: elegant graphics for data analysis, 2009.

P. Sohrab, A. Shah, R. Roth, A. Goya, G. Oloumi et al., The clonal and mutational evolution spectrum of primary triple-negative breast cancers, Nature, issue.7403, pp.486395-399, 2012.

. Mirny, Impact of deleterious passenger mutations on cancer progression, Proceedings of the National Academy of Sciences, vol.110, issue.8, pp.2910-2915, 2013.

G. Chen, W. A. Mulla, A. Kucharavy, H. Tsai, B. Rubinstein et al., Targeting the Adaptability of Heterogeneous Aneuploids, Cell, vol.160, issue.4, pp.771-784, 2015.
DOI : 10.1016/j.cell.2015.01.026

URL : https://hal.archives-ouvertes.fr/hal-01528517

A. Marc, N. Bollet, P. Servant, C. Neuvial, I. Decraene et al., High-Resolution Mapping of DNA Breakpoints to Define True Recurrences Among Ipsilateral Breast Cancers, Journal of the National Cancer Institute, vol.100, issue.1, pp.48-58, 2008.

M. E. Bibliography, M. Adriaens, A. Jaillard, S. L. Waagmeester, A. R. Coort et al., The public road to high-quality curated biological pathways, Drug Discovery Today, vol.13, pp.856-862, 2008.

B. Aranda, H. Blankenburg, S. Kerrien, F. S. Brinkman, A. Ceol et al., PSICQUIC and PSISCORE: accessing and scoring molecular interactions, Nature Methods, vol.8, issue.7, pp.528-529, 2011.
DOI : 10.1002/pmic.200700658

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3246345

E. Barillot, L. Calzone, P. Hupe, J. Vert, and A. Zinovyev, Computational systems biology of cancer, pp.2012-293

J. Barretina, G. Caponigro, N. Stransky, K. Venkatesan, A. A. Margolin et al., The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity, Nature, vol.53, issue.7391, pp.483603-607, 2012.
DOI : 10.1002/pbc.20697

E. Bonnet, E. Viara, I. Kuperstein, L. Calzone, D. P. Cohen et al., NaviCell Web Service for network-based data visualization, Nucleic Acids Research, vol.43, issue.W1, pp.450-2015
DOI : 10.1093/nar/gkv450

URL : http://doi.org/10.1093/nar/gkv450

J. P. Cogswell, J. M. Ward, I. A. Taylor, M. Waters, Y. Shi et al., Identification of miRNA Changes in Alzheimer's Disease Brain and CSF Yields Putative Biomarkers and Insights into Disease Pathways, Journal of Alzheimer's Disease, vol.14, issue.1, pp.27-41, 2008.
DOI : 10.3233/JAD-2008-14103

D. Croft, A. F. Mundo, R. Haw, M. Milacic, J. Weiser et al., The Reactome pathway knowledgebase, Nucleic Acids Research, vol.42, issue.D1, pp.472-477, 2014.
DOI : 10.1093/nar/gkt1102

S. Falcon and R. Gentleman, Using GOstats to test gene lists for GO term association, Bioinformatics, vol.23, issue.2, pp.257-258, 2007.
DOI : 10.1093/bioinformatics/btl567

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.183.9047

R. A. Fisher, On the interpretation of ?2 from contingency tables, and the calculation of p, Journal of the Royal Statistical Society, pp.87-94, 1922.

R. A. Fisher, Statistical methods for research workers, 1934.

R. D. Hawkins, G. C. Hon, and B. Ren, Next-generation genomics: an integrative approach, Nature Reviews Genetics, vol.17, issue.7, pp.476-486, 2010.
DOI : 10.1101/gr.229102. Article published online before print in May 2002

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3321268

M. Kanehisa, S. Goto, Y. Sato, M. Furumichi, and M. Tanabe, KEGG for integration and interpretation of large-scale molecular data sets, Nucleic Acids Research, vol.40, issue.D1, pp.988-2011
DOI : 10.1093/nar/gkr988

T. Kelder, M. P. Van-iersel, K. Hanspers, M. Kutmon, B. R. Conklin et al., WikiPathways: building research communities on biological pathways, Nucleic Acids Research, vol.40, issue.D1, pp.1301-1307, 2012.
DOI : 10.1093/nar/gkr1074

URL : http://doi.org/10.1093/nar/gkr1074

I. Kuperstein, D. P. Cohen, S. Pook, E. Viara, L. Calzone et al., NaviCell: a web-based environment for navigation, curation and maintenance of large molecular interaction maps, BMC Systems Biology, vol.7, issue.1, p.100, 2013.
DOI : 10.1093/nar/gkr1097

URL : https://hal.archives-ouvertes.fr/inserm-00872428

I. Kuperstein, E. Bonnet, H. Nguyen, D. Cohen, E. Viara et al., Atlas of Cancer Signalling Network: a systems biology resource for integrative analysis of cancer data with Google Maps, Oncogenesis, vol.1046, issue.7, pp.160-2015
DOI : 10.1093/annonc/mdr304

D. T. Lang, RJSONIO: Serialize R objects to JSON, JavaScript Object Notation, 2014.

D. T. Lang, Client Interface for R, 2015. URL http, RCurl: General Network

A. Liberzon, A. Subramanian, R. Pinchback, H. Thorvaldsdóttir, P. Tamayo et al., Molecular signatures database (MSigDB) 3.0, Bioinformatics, vol.27, issue.12, pp.1739-1740, 2011.
DOI : 10.1093/bioinformatics/btr260

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3106198

. Luo, . Weijun, . Friedman, . Michael, . Shedden et al., GAGE: generally applicable gene set enrichment for pathway analysis, BMC Bioinformatics, vol.10, issue.1, p.161, 2009.
DOI : 10.1186/1471-2105-10-161

E. A. Maher, F. B. Furnari, R. M. Bachoo, D. H. Rowitch, D. N. Louis et al., Malignant glioma: genetics and biology of a grave matter, Genes & Development, vol.15, issue.11, pp.1311-1333, 2001.
DOI : 10.1101/gad.891601

R. Mclendon, A. Friedman, D. Bigner, E. G. Van-meir, D. J. Brat et al., Comprehensive genomic characterization defines human glioblastoma genes and core pathways, Nature, vol.123, issue.7216, pp.4551061-1068, 2008.
DOI : 10.2174/187152008784220276

A. Reiner, D. Yekutieli, and Y. Benjamini, Identifying differentially expressed genes using false discovery rate controlling procedures, Bioinformatics, vol.19, issue.3, pp.368-375, 2003.
DOI : 10.1093/bioinformatics/btf877

URL : https://academic.oup.com/bioinformatics/article-pdf/19/3/368/717620/btf877.pdf

I. Rivals, L. Personnaz, L. Taing, and M. Potier, Enrichment or depletion of a GO category within a class of genes: which test?, Bioinformatics, vol.23, issue.4, pp.401-407, 2007.
DOI : 10.1093/bioinformatics/btl633

URL : https://hal.archives-ouvertes.fr/hal-00801557

M. Schmidt, D. Böhm, C. Von-törne, E. Steiner, A. Puhl et al., The Humoral Immune System Has a Key Prognostic Impact in Node-Negative Breast Cancer, Cancer Research, vol.68, issue.13, pp.685405-5413, 2008.
DOI : 10.1158/0008-5472.CAN-07-5206

H. Wickham, ggplot2: elegant graphics for data analysis, 2009.

H. Wickham, Reference classes, 2015.

G. Yu and Q. He, ReactomePA: an R/Bioconductor package for reactome pathway analysis and visualization, C5MB00663E. [p303, 2015.
DOI : 10.1186/gb-2014-15-2-r23

G. Yu, L. Wang, Y. Han, and Q. He, clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters, OMICS: A Journal of Integrative Biology, vol.16, issue.5, pp.284-287, 2012.
DOI : 10.1089/omi.2011.0118

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3339379

A. Zanzoni, L. Montecchi-palazzi, M. Quondam, G. Ausiello, M. Helmer-citterich et al., MINT: a Molecular INTeraction database, FEBS Letters, vol.28, issue.1, pp.135-140, 2002.
DOI : 10.1093/nar/28.1.37

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1751541/pdf

I. Curie, . Psl-research-university, . Inserm-u830, and R. Laboratoire, Département de recherche translationnelle 4 Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA 5 Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Recherche Translationelle en Oncologie Pédiatrique), vol.75005

. Clone, Similarly, when ve samples were provided, we observed a 74

. France, the review board at the Children's Hospital of Philadelphia and review boards at other Children's 349

. Eleveld, Illumina paired-end sequencing for 7 novel neuroblastoma patients (the total of 21 patients corresponding to 355 a tumor at diagnosis and relapse, and a matching blood sample per patient) Data for 15 patients were taken 356 and reanalyzed from the previous study DNA from 7 patients from the previous study 357 and 7 new ones have been sequenced using Illumina HiSeq 2500 instruments to an average depth of coverage 358 of 80× by, BGI) and the Centre National de Génotypage (CNG) respectively. For 359, 2015.

. Mckenna, Complete Genomics tools for Illumina and Complete Genomics datasets respectively Reads from datasets 366 sequenced using the Illumina platform were realigned around indels with the, Genome Analysis ToolKit, vol.367, 2010.

. Boeva, Copy number analysis 390 Copy number alterations in patients were detected using the Control-FREEC method We selected the main ploidy value so that the predicted copy number and 392 B-allele frequency proles were consistent. Control-FREEC also provided estimations of the level of contam- 393 ination by normal cells, which, after manual conrmation, was further used for the clonal reconstruction, p.391, 2012.

. Quantumclone, QuantumCat simulates genomic variants, copy number alterations and 402 corresponding VAFs. It relies on the following set of rules: 403 1. A binary phylogenetic tree is created to simulate the clonal architecture of the tumor. The mutation 404 cellular prevalence values correspond to the nodes and leaves of the phylogenetic tree

M. Carlo, beta parameters in the Beta base measure for Dirichlet Process set to 1, concentra- 425 tion prior shape set to 1 and the rate parameter in the Gamma prior on the concentration parameter set to 426 0.001. We used the default Beta binomial distribution with precision parameter set to 1000, prior shape set 427 to 1, rate to 0.0001, and proposal precision set to 0, p.1

I. Curie, This study was also funded by the Associations Enfants et Santé

. Enfance, L. Cancer, and . Bagouz-À-manon, Les amis de Claire. VB and her team were supported by the 540

I. Adzhubei, D. M. Jordan, S. R. Sunyaev, L. Jonathan, and . Haines, Predicting Functional Eect of Human Missense Mutations Using PolyPhen-2. Current protocols in human genetics, p.7, 2013.

V. Boeva, T. Popova, K. Bleakley, P. Chiche, J. Cappo et al., Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data, Bioinformatics, vol.28, issue.3, p.423425
DOI : 10.1093/bioinformatics/btr670

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3268243

M. A. Bollet, N. Servant, P. Neuvial, C. Decraene, I. Lebigot et al., High-Resolution Mapping of DNA Breakpoints to Dene True Recurrences Among Ipsilateral Breast Cancers, Journal of the National Cancer Institute, vol.100, issue.1, p.4858, 2008.

E. C. Bruin, N. Mcgranahan, R. Mitter, M. Salm, D. C. Wedge et al., Spatial and temporal diversity in genomic instability processes denes lung cancer evolution, Science, issue.6206, p.346251256, 2014.

G. Chen, W. A. Mulla, A. Kucharavy, H. Tsai, B. Rubinstein et al., Targeting the Adaptability of Heterogeneous Aneuploids, Cell, vol.160, issue.4, p.771784, 2015.
DOI : 10.1016/j.cell.2015.01.026

URL : https://hal.archives-ouvertes.fr/hal-01528517

P. Deveau, E. Barillot, V. Boeva, A. Zinovyev, and E. Bonnet, Calculating biological module enrichment or depletion and visualizing data on large-scale molecular maps with ACSNMineR and RNaviCell packages, The R Journal, vol.8, issue.2, p.293306, 2016.
DOI : 10.1101/064469

T. F. Eleveld, D. A. Oldridge, V. Bernard, J. Koster, L. C. Daage et al., Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations, Nature Genetics, vol.52, issue.8, p.47864871, 2015.
DOI : 10.1038/nbt.1523

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4775079

A. Fischer, I. Vázquez-garcía, C. J. Illingworth, and V. Mustonen, High-Denition Reconstruction of Clonal Composition in Cancer, Cell Reports, vol.7, issue.5, p.17401752, 2014.

P. A. Futreal, L. Coin, M. Marshall, T. Down, T. Hubbard et al., A census of human cancer genes, Nature Reviews Cancer, vol.4, issue.3, p.177183, 2004.
DOI : 10.1038/nrc1299

I. Hajirasouliha, A. Mahmoody, R. , and B. J. , A combinatorial approach for analyzing intratumor heterogeneity from high-throughput sequencing data, Bioinformatics, issue.12, pp.30-78, 2014.

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, p.646674, 2011.
DOI : 10.1016/j.cell.2011.02.013

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Research, vol.37, issue.1, p.113, 2009.
DOI : 10.1093/nar/gkn923

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature Protocols, vol.99, issue.1, p.4457, 2009.
DOI : 10.6026/97320630002428

W. Jiao, S. Vembu, A. G. Deshwar, L. Stein, M. et al., Inferring clonal evolution of tumors from single nucleotide somatic mutations, BMC Bioinformatics, vol.15, issue.1, p.35, 2014.
DOI : 10.1111/j.2041-210X.2011.00131.x

T. B. Kepler, Reconstructing a B-cell clonal lineage. I. Statistical inference of unobserved ancestors, F1000Research, 1000.
DOI : 10.6084/m9.figshare.656793

E. Khurana, Y. Fu, V. Colonna, X. J. Mu, H. M. Kang et al., Integrative Annotation of Variants from 1092 Humans: Application to Cancer Genomics, Science, vol.342, issue.6154, p.3421235587, 2013.
DOI : 10.1186/1479-7364-5-6-709

D. C. Koboldt, D. E. Larson, W. , R. K. Andreas, and D. Baxevanis, Using VarScan 2 for Germline Variant Calling and Somatic Mutation Detection. Current protocols in bioinformatics, 2013.

I. Kuperstein, E. Bonnet, H. Nguyen, D. Cohen, E. Viara et al., Atlas of Cancer Signalling Network: a systems biology resource for integrative analysis of cancer data with Google Maps, Oncogenesis, vol.1046, issue.7, p.160, 2015.
DOI : 10.1093/annonc/mdr304

H. Li and R. Durbin, Fast and accurate short read alignment with BurrowsWheeler transform, Bioinformatics, issue.14, p.2517541760, 2009.
DOI : 10.1093/bioinformatics/btp324

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2705234

S. Malikic, A. W. Mcpherson, N. Donmez, and C. S. Sahinalp, Clonality inference in multiple tumor samples using phylogeny, Bioinformatics, vol.31, issue.9, p.3113491356, 2015.
DOI : 10.1093/bioinformatics/btv003

URL : http://summit.sfu.ca/system/files/iritems1/14463/etd8573_SMalikic.pdf

C. D. Manning, P. Raghavan, and H. Schütze, Introduction to information retrieval, 2008.
DOI : 10.1017/CBO9780511809071

A. Marusyk, D. P. Tabassum, P. M. Altrock, V. Almendro, F. Michor et al., Non-cellautonomous driving of tumour growth supports sub-clonal heterogeneity, Nature, vol.2014, issue.7520, p.5145458
DOI : 10.1038/nature13556

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4184961

C. D. Mcfarland, K. S. Korolev, G. V. Kryukov, S. R. Sunyaev, and L. A. Mirny, Impact of deleterious passenger mutations on cancer progression, Proceedings of the National Academy of Sciences, p.11029102915, 2013.
DOI : 10.1038/nature07485

A. Mckenna, M. Hanna, E. Banks, A. Sivachenko, K. Cibulskis et al., The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data, Genome Research, vol.20, issue.9, p.12971303, 2010.
DOI : 10.1101/gr.107524.110

H. Mi, Q. Dong, A. Muruganujan, P. Gaudet, S. Lewis et al., PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium, Nucleic Acids Research, vol.38, issue.suppl_1, pp.204-210, 2010.
DOI : 10.1093/nar/gkp1019

URL : http://doi.org/10.1093/nar/gkp1019

C. A. Miller, B. S. White, N. D. Dees, M. Grith, J. S. Welch et al., SciClone: Inferring Clonal Architecture and Tracking the Spatial and Temporal Patterns of Tumor Evolution, PLoS Computational Biology, vol.10, issue.8, pp.10-1003665, 2014.
DOI : 10.1371/journal.pcbi.1003665.s010

J. J. Molenaar, J. Koster, D. A. Zwijnenburg, P. Van-sluis, L. J. Valentijn et al., Sequencing of neuroblastoma identies chromothripsis and defects in neuritogenesis genes, Nature, issue.7391, p.483589593, 2012.

P. C. Ng and S. Heniko, SIFT: predicting amino acid changes that affect protein function, Nucleic Acids Research, vol.31, issue.13, p.3138123814, 2003.
DOI : 10.1093/nar/gkg509

URL : https://academic.oup.com/nar/article-pdf/31/13/3812/9487105/gkg509.pdf

Y. Qiao, A. R. Quinlan, A. A. Jazaeri, R. Verhaak, D. A. Wheeler et al., Subclone- Seeker: a computational framework for reconstructing tumor clone structure for cancer variant interpretation and prioritization, Genome Biology, issue.8, p.15443, 2014.
DOI : 10.1186/preaccept-1691129011290725

URL : http://doi.org/10.1186/preaccept-1691129011290725

A. Roth, J. Khattra, D. Yap, A. Wan, E. Laks et al., PyClone: statistical inference of clonal population structure in cancer, Nature Methods, vol.11, issue.4, p.396398, 2014.
DOI : 10.1093/bioinformatics/btp698

A. Schramm, J. Köster, Y. Assenov, K. Altho, M. Peifer et al., Mutational dynamics between primary and relapse neuroblastomas, Nature Genetics, vol.4, issue.8, p.47872877, 2015.
DOI : 10.1016/j.ccr.2008.12.005

R. F. Schwarz, A. Trinh, B. Sipos, J. D. Brenton, N. Goldman et al., Phylogenetic Quantification of Intra-tumour Heterogeneity, PLoS Computational Biology, vol.11, issue.6, p.1003535, 2014.
DOI : 10.1371/journal.pcbi.1003535.g006

URL : http://doi.org/10.1371/journal.pcbi.1003535

P. D. Thomas, M. J. Campbell, A. Kejariwal, H. Mi, B. Karlak et al., PANTHER: A Library of Protein Families and Subfamilies Indexed by Function, Genome Research, vol.13, issue.9, p.21292141, 2003.
DOI : 10.1101/gr.772403

C. Tomasetti and B. Vogelstein, Variation in cancer risk among tissues can be explained by the number of stem cell divisions, Science, vol.10, issue.4, p.3477881, 2015.
DOI : 10.1038/ng0895-383

M. J. Williams, B. Werner, C. P. Barnes, T. A. Graham, and A. Sottoriva, Identication of neutral tumor evolution across cancer types, Nature Genetics, issue.3, p.48238244, 2016.
DOI : 10.1038/ng.3489

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4934603