F. Michor and K. Polyak, The Origins and Implications of Intratumor Heterogeneity, Cancer Prevention Research, vol.3, issue.11, pp.1361-1365, 2010.
DOI : 10.1158/1940-6207.CAPR-10-0234

J. Visvader, Cells of origin in cancer, Nature, vol.28, issue.7330, pp.314-336, 2011.
DOI : 10.1038/nature09781

R. Fisher, L. Pusztai, and C. Swanton, Cancer heterogeneity: implications for targeted therapeutics, British Journal of Cancer, vol.25, issue.3, pp.479-85, 2013.
DOI : 10.1126/scitranslmed.3003854

C. Alix-panabieres and K. Pantel, Circulating Tumor Cells: Liquid Biopsy of Cancer, Clinical Chemistry, vol.59, issue.1, pp.60-64
DOI : 10.1373/clinchem.2012.194258

M. Gabriel, L. Calleja, A. Chalopin, B. Ory, and D. Heymann, Circulating Tumor Cells: A Review of Non-EpCAM-Based Approaches for Cell Enrichment and Isolation, Clinical Chemistry, vol.62, issue.4, pp.571-81, 2016.
DOI : 10.1373/clinchem.2015.249706

URL : https://hal.archives-ouvertes.fr/inserm-01466107

I. Fidler and I. Hart, Biological diversity in metastatic neoplasms: origins and implications, Science, vol.217, issue.4564, pp.998-1003, 1982.
DOI : 10.1126/science.7112116

D. Hayes and C. Paoletti, Circulating tumour cells: insights into tumour heterogeneity, Journal of Internal Medicine, vol.21, issue.suppl, pp.137-180, 2013.
DOI : 10.1111/joim.12047

P. Friedl and A. S. , Cancer Invasion and the Microenvironment: Plasticity and Reciprocity, Cell, vol.147, issue.5, pp.992-1009, 2011.
DOI : 10.1016/j.cell.2011.11.016

E. Andrechek, J. Nevins, R. Somasundaram, J. Villanueva, and M. Herlyn, Mouse models of cancers: opportunities to address heterogeneity of human cancer and evaluate therapeutic strategies Intratumoral heterogeneity as a therapy resistance mechanism: role of melanoma subpopulations Intratumoral heterogeneity of immunohistochemical marker expression in breast carcinoma: a tissue microarray-based study, Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions, pp.1095-100335, 2008.

P. Nowell, The clonal evolution of tumor cell populations, Science, vol.194, issue.4260, pp.23-31, 1976.
DOI : 10.1126/science.959840

T. Mazor, A. Pankov, J. Song, and J. Costello, Intratumoral Heterogeneity of the Epigenome Portela A, Esteller M. Epigenetic modifications and human diseaseEvaluation of biological effects and possible mechanisms of action of static magnetic field], Cancer Cell. Nat Biotechnol. Fiziol Zh, vol.292858, issue.163, pp.440-511057, 2010.

J. Cassidy, C. Caldas, and A. Bruna, Maintaining Tumor Heterogeneity in Patient-Derived Tumor Xenografts, Cancer Research, vol.75, issue.15, pp.2963-2971, 2015.
DOI : 10.1158/0008-5472.CAN-15-0727

T. Tlsty and L. Coussens, TUMOR STROMA AND REGULATION OF CANCER DEVELOPMENT, Annual Review of Pathology: Mechanisms of Disease, vol.1, issue.1, pp.119-50, 2006.
DOI : 10.1146/annurev.pathol.1.110304.100224

N. Turner and J. Reis-filho, Genetic heterogeneity and cancer drug resistance, The Lancet Oncology, vol.13, issue.4, pp.178-85, 2012.
DOI : 10.1016/S1470-2045(11)70335-7

J. Wu, M. Fackler, M. Halushka, D. Molavi, M. Taylor et al., Heterogeneity of Breast Cancer Metastases: Comparison of Therapeutic Target Expression and Promoter Methylation Between Primary Tumors and Their Multifocal Metastases, Wicha MS. Cancer stem cell heterogeneity in hereditary breast cancer, pp.1938-46105, 2008.
DOI : 10.1158/1078-0432.CCR-07-4082

A. Guidi, D. Berry, G. Broadwater, M. Perloff, L. Norton et al., Association of Angiogenesis in Lymph Node Metastases With Outcome of Breast Cancer, JNCI: Journal of the National Cancer Institute, vol.92, issue.6, pp.486-92, 2000.
DOI : 10.1093/jnci/92.6.486

M. Greaves and C. Maley, Clonal evolution in cancer, Nature, vol.69, issue.7381, pp.306-319, 2012.
DOI : 10.1038/nature10762

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3367003

G. Gundem, V. Loo, P. Kremeyer, B. Alexandrov, L. Tubio et al., The evolutionary history of lethal metastatic prostate cancer, Nature, vol.4, issue.7547, pp.353-360, 2015.
DOI : 10.1038/nature14347

M. Hong, G. Macintyre, D. Wedge, V. Loo, P. Patel et al., Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer Genomic instability--an evolving hallmark of cancer, Nat Commun. Nat Rev Mol Cell Biol, vol.611, issue.3, pp.220-228, 2010.

D. Hanahan and R. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, pp.646-74, 2011.
DOI : 10.1016/j.cell.2011.02.013

G. Dakubo, J. Jakupciak, M. Birch-machin, R. Parr, A. Marusyk et al., Clinical implications and utility of field cancerization 29. Lichtenstein AV. Clonal heterogeneity of tumor may be due to continuous influx of newly transformed cells Intra-tumour heterogeneity: a looking glass for cancer?, Cancer Cell Int. Cancer Biol Ther. Nat Rev Cancer, vol.7512, issue.305, pp.1263-4323, 2006.

G. Assie, E. Letouze, M. Fassnacht, A. Jouinot, W. Luscap et al., Integrated genomic characterization of adrenocortical carcinoma Wilting RH, Dannenberg JH Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance Intratumoral Heterogeneity of MicroRNA Expression in Rectal Cancer, ):e0156919. 34. Berdasco M, Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry, pp.607-1221, 2010.

R. Place, L. Li, D. Pookot, E. Noonan, R. Dahiya et al., MicroRNA-373 induces expression of genes with complementary promoter sequences Primary colorectal cancers and their subsequent hepatic metastases are genetically different: implications for selection of patients for targeted treatment, Proceedings of the National Academy of Sciences of the United States of America, pp.1608-13688, 2008.

E. Vakiani, M. Janakiraman, R. Shen, R. Sinha, Z. Zeng et al., Comparative Genomic Analysis of Primary Versus Metastatic Colorectal Carcinomas, Journal of Clinical Oncology, vol.30, issue.24, pp.2956-62, 2012.
DOI : 10.1200/JCO.2011.38.2994

S. Ekinci, A. Demirci, U. , C. Oksuzoglu, B. Ozturk et al., KRAS discordance between primary and metastatic tumor in patients with metastatic colorectal carcinoma, J BUON, vol.20, issue.1, pp.128-163, 2015.

M. Colombino, M. Capone, A. Lissia, A. Cossu, C. Rubino et al., Mutation Frequencies Among Primary Tumors and Metastases in Patients With Melanoma, Journal of Clinical Oncology, vol.30, issue.20, pp.2522-2531, 2012.
DOI : 10.1200/JCO.2011.41.2452

C. Mao, X. Wu, Z. Yang, D. Threapleton, J. Yuan et al., Concordant analysis of KRAS, BRAF, PIK3CA mutations, and PTEN expression between primary colorectal cancer and matched metastases. Sci Rep Analysis of the concordance in the EGFR pathway status between primary tumors and related metastases of colorectal cancer patients:implications for cancer therapy, Curr Cancer Drug Targets, vol.512, issue.2, pp.8065-41124, 2012.

Z. Chen, W. Zhong, X. Zhang, J. Su, X. Yang et al., EGFR Mutation Heterogeneity and the Mixed Response to EGFR Tyrosine Kinase Inhibitors of Lung Adenocarcinomas, The Oncologist, vol.17, issue.7, pp.978-85, 2012.
DOI : 10.1634/theoncologist.2011-0385

P. Steeg, Tumor metastasis: mechanistic insights and clinical challenges, Nature Medicine, vol.96, issue.8, pp.895-904, 2006.
DOI : 10.1038/nm1469

E. Heitzer, M. Auer, C. Gasch, M. Pichler, P. Ulz et al., Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by Array-CGH and Next-Generation Sequencing, Cancer Research, vol.73, issue.10, pp.2965-75, 2013.
DOI : 10.1158/0008-5472.CAN-12-4140

B. Mostert, Y. Jiang, A. Sieuwerts, H. Wang, J. Bolt-de-vries et al., mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue, International Journal of Cancer, vol.7, issue.1, pp.130-171, 2013.
DOI : 10.1002/ijc.27987

M. Suhaimi, N. Fy, D. Lee, W. Phyo, I. Cima et al., Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients Heterogeneous proliferative potential of occult metastatic cells in bone marrow of patients with solid epithelial tumors, Proceedings of the National Academy of Sciences of the United States of America, pp.2246-51, 2002.

J. Kraus, K. Pantel, D. Pinkel, D. Albertson, M. Speicher et al., High-resolution genomic profiling of occult micrometastatic tumor cells High level of chromosomal instability in circulating tumor cells of ROS1-rearranged non-small-cell lung cancer, Genes Chromosomes Cancer. Ann Oncol, vol.3626, issue.27, pp.159-661408, 2003.

G. Middleton, L. Crack, S. Popat, C. Swanton, S. Hollingsworth et al., The National Lung Matrix Trial: translating the biology of stratification in advanced non-small-cell lung cancer, Annals of Oncology, vol.26, issue.12, pp.2464-2473, 2015.
DOI : 10.1093/annonc/mdv394

M. Jamal-hanjani, A. Hackshaw, Y. Ngai, J. Shaw, C. Dive et al., Tracking Genomic Cancer Evolution for Precision Medicine: The Lung TRACERx Study, PLoS Biology, vol.3, issue.7, pp.1001906-52, 2014.
DOI : 10.1371/journal.pbio.1001906.g002

C. Alix-panabieres and K. Pantel, Technologies for detection of circulating tumor cells: facts and vision, Lab Chip, vol.20, issue.1, pp.57-62, 2014.
DOI : 10.1002/hed.23439

S. Joosse, T. Gorges, and K. Pantel, Biology, detection, and clinical implications of circulating tumor cells, EMBO Molecular Medicine, vol.7, issue.1, pp.1-11, 2015.
DOI : 10.15252/emmm.201303698

S. Au, B. Storey, J. Moore, Q. Tang, Y. Chen et al., Clusters of circulating tumor cells traverse capillary-sized vessels, Proceedings of the National Academy of Sciences, vol.113, issue.18, pp.4947-52, 2016.
DOI : 10.1073/pnas.1524448113

A. Place, J. Huh, S. Polyak, and K. , The microenvironment in breast cancer progression: biology and implications for treatment, Breast Cancer Research, vol.116, issue.Suppl 2, pp.227-56, 2011.
DOI : 10.1172/JCI26532

C. Alix-panabieres, K. Pantel, C. Gasch, K. Pantel, S. Riethdorf et al., Circulating tumor cells: liquid biopsy of cancer Whole Genome Amplification in Genomic Analysis of Single Circulating Tumor Cells CTC enumeration and characterization: moving toward personalized medicine, Clin Chem. Methods Mol Biol. Ann Transl Med, vol.5913472, issue.111, pp.110-118, 2013.

F. Fabbri, S. Carloni, W. Zoli, P. Ulivi, G. Gallerani et al., Detection and recovery of circulating colon cancer cells using a dielectrophoresis-based device: KRAS mutation status in pure CTCs, Cancer Letters, vol.335, issue.1, pp.225-256, 2013.
DOI : 10.1016/j.canlet.2013.02.015

X. Ni, M. Zhuo, Z. Su, J. Duan, Y. Gao et al., Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients TP53 mutations detected in circulating tumor cells present in the blood of metastatic triple negative breast cancer patients, Proceedings of the National Academy of Sciences of the United States of America):445. 62. Pestrin M al. Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients, pp.21083-21091, 2013.

K. Sakaizawa, Y. Goto, Y. Kiniwa, A. Uchiyama, K. Harada et al., Mutation analysis of BRAF and KIT in circulating melanoma cells at the single cell level Serial blood-based analysis of AR-V7 in men with advanced prostate cancer, Mol Oncol. Br J Cancer. Nakazawa M Ann Oncol, vol.910626, issue.649, pp.749-57939, 2012.

S. Kobayashi, H. Canepa, A. Bailey, S. Nakayama, N. Yamaguchi et al., Compound EGFR Mutations and Response to EGFR Tyrosine Kinase Inhibitors, Journal of Thoracic Oncology, vol.8, issue.1, pp.45-51, 2013.
DOI : 10.1097/JTO.0b013e3182781e35

URL : http://doi.org/10.1097/jto.0b013e3182781e35

M. Yu, A. Bardia, N. Aceto, F. Bersani, M. Madden et al., Cancer therapy Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility PIK3CA mutational status in circulating tumor cells can change during disease recurrence or progression in patients with breast cancer, Science. Clin Cancer Res, vol.34520, issue.619322, pp.216-205823, 2014.

M. Kovac, C. Blattmann, S. Ribi, J. Smida, N. Mueller et al., Exome sequencing of osteosarcoma reveals mutation signatures reminiscent of BRCA deficiency:8940. 69. Itzkovitz S, van Oudenaarden A. Validating transcripts with probes and imaging technology, Nat Commun. Nature methods, vol.68, issue.4, pp.12-21, 2011.

B. Summersgill, J. Clark, and J. Shipley, Fluorescence and chromogenic in situ hybridization to detect genetic aberrations in formalin-fixed paraffin embedded material, including tissue microarrays, Nature Protocols, vol.27, issue.2, pp.220-254, 2008.
DOI : 10.1038/nprot.2007.534

H. Fiegler, J. Geigl, S. Langer, D. Rigler, K. Porter et al., High resolution array-CGH analysis of single cells. Nucleic acids research Microarray-based comparative genomic hybridization and its applications in human genetics, Clinical genetics, vol.3566, issue.36, pp.488-95, 2004.

S. Park, M. Gonen, H. Kim, F. Michor, and K. Polyak, Cellular and genetic diversity in the progression of in situ human breast carcinomas to an invasive phenotype, Journal of Clinical Investigation, vol.120, issue.2, pp.636-680, 2010.
DOI : 10.1172/JCI40724DS1

F. Wang, J. Flanagan, N. Su, L. Wang, S. Bui et al., RNAscope, The Journal of Molecular Diagnostics, vol.14, issue.1, pp.22-31, 2012.
DOI : 10.1016/j.jmoldx.2011.08.002

URL : http://doi.org/10.1016/j.jmoldx.2011.08.002

J. Lee, E. Daugharthy, J. Scheiman, R. Kalhor, T. Ferrante et al., Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues, Nature Protocols, vol.536, issue.3, pp.442-58, 2015.
DOI : 10.1038/nmeth.2047

M. Janiszewska, L. Liu, V. Almendro, Y. Kuang, C. Paweletz et al., In situ single-cell analysis identifies heterogeneity for PIK3CA mutation and HER2 amplification in HER2-positive breast cancer, Nature Genetics, vol.134, issue.10, pp.1212-1221, 2015.
DOI : 10.1002/path.3032

M. Setou, K. Shrivas, M. Sroyraya, H. Yang, Y. Sugiura et al., Developments and applications of mass microscopy, Medical Molecular Morphology, vol.81, issue.1, pp.1-5, 2010.
DOI : 10.1007/s00795-009-0489-0

V. Almendro, A. Marusyk, and K. Polyak, Cellular Heterogeneity and Molecular Evolution in Cancer, Annual Review of Pathology: Mechanisms of Disease, vol.8, issue.1
DOI : 10.1146/annurev-pathol-020712-163923

X. Xu, Y. Hou, X. Yin, L. Bao, A. Tang et al., Single-cell exome sequencing reveals singlenucleotide mutation characteristics of a kidney tumor Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution Tumour evolution inferred by single-cell sequencing Future medical applications of single-cell sequencing in cancer Genomic profiling defines subtypes of prostate cancer with the potential for therapeutic stratification, Cell. Nature. Nature. Genome Med. Schoenborn JR Clin Cancer Res, vol.148461472319, issue.82515, pp.886-95809, 2009.

F. Thege, T. Lannin, T. Saha, S. Tsai, M. Kochman et al., Microfluidic immunocapture of circulating pancreatic cells using parallel EpCAM and MUC1 capture: characterization, optimization and downstream analysis Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer, Lab Chip. Clin Chem. Science, vol.1459274, issue.865289, pp.1775-84252, 1996.

C. Gawad, W. Koh, and S. Quake, Single-cell genome sequencing: current state of the science, Nature Reviews Genetics, vol.148, issue.3, pp.175-88, 2016.
DOI : 10.1038/onc.2013.29

C. Gawad, W. Koh, S. Quake, K. Yeung, C. Fraley et al., Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics Model-based clustering and data transformations for gene expression data Single-cell RNA-seq: advances and future challenges, Proceedings of the National Academy of Sciences of the United States of America. Bioinformatics. Nucleic acids research, vol.1111742, issue.9014, pp.17947-52977, 2001.

L. Wen, F. Tang, K. Chen, A. Boettiger, J. Moffitt et al., Single-cell sequencing in stem cell biology RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells Cancer genomics: one cell at a time, aaa6090. 93. Navin NE, pp.71-92452, 2014.

D. Luca, F. Rotunno, G. Salvianti, F. Galardi, F. Pestrin et al., Mutational analysis of single circulating tumor cells by next generation sequencing in metastatic breast cancer, Oncotarget, vol.7, issue.18, pp.26107-26126, 2016.
DOI : 10.18632/oncotarget.8431

R. Jiang, Y. Lu, H. Ho, B. Li, J. Chen et al., A comparison of isolated circulating tumor cells and tissue biopsies using whole-genome sequencing in prostate cancer, Oncotarget, vol.6, issue.42, pp.44781-93, 2015.

A. Powell, A. Talasaz, H. Zhang, M. Coram, A. Reddy et al., Single cell profiling of circulating tumor cells: transcriptional heterogeneity and diversity from breast cancer cell lines Expression profiling of circulating tumor cells in metastatic breast cancer Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state al. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics, PloS one. Breast Cancer Res Treat. Nat Biotechnol. Farlik M Cell Rep, vol.71493310, issue.998, pp.33788-97121, 2012.

R. Doherty and C. Couldrey, Exploring genome wide bisulfite sequencing for DNA methylation analysis in livestock: a technical assessment, Frontiers in Genetics, vol.102, issue.11., p.126, 2014.
DOI : 10.1016/j.ygeno.2013.04.014

T. Morris and S. Beck, Analysis pipelines and packages for Infinium HumanMethylation450 BeadChip (450k) data, Methods, vol.72, pp.3-8, 2015.
DOI : 10.1016/j.ymeth.2014.08.011

URL : http://doi.org/10.1016/j.ymeth.2014.08.011

D. Wlodkowic and Z. Darzynkiewicz, Rise of the Micromachines: Microfluidics and the Future of Cytometry, Methods Cell Biol, vol.102, pp.105-130, 2011.
DOI : 10.1016/B978-0-12-374912-3.00005-5

M. Werner, F. Merenda, J. Piguet, R. Salathe, and H. Vogel, Microfluidic array cytometer based on refractive optical tweezers for parallel trapping, imaging and sorting of individual cells, Lab on a Chip, vol.10, issue.14, pp.2432-2441, 2011.
DOI : 10.1039/c1lc20181f

J. Sun, M. Masterman-smith, N. Graham, J. Jiao, J. Mottahedeh et al., A Microfluidic Platform for Systems Pathology: Multiparameter Single-Cell Signaling Measurements of Clinical Brain Tumor Specimens, Cancer Research, vol.70, issue.15, pp.6128-6166, 2010.
DOI : 10.1158/0008-5472.CAN-10-0076

N. Kotecha, N. Flores, J. Irish, E. Simonds, D. Sakai et al., Single-Cell Profiling Identifies Aberrant STAT5 Activation in Myeloid Malignancies with Specific Clinical and Biologic Correlates, Cancer Cell, vol.14, issue.4, pp.335-378, 2008.
DOI : 10.1016/j.ccr.2008.08.014

J. Irish, J. Myklebust, A. Alizadeh, R. Houot, J. Sharman et al., B-cell signaling networks reveal a negative prognostic human lymphoma cell subset that emerges during tumor progression, Proceedings of the National Academy of Sciences, vol.107, issue.29, pp.12747-54, 2010.
DOI : 10.1073/pnas.1002057107

A. Palazzo, E. Evensen, Y. Huang, A. Cesano, G. Nolan et al., Association of Reactive Oxygen Species-Mediated Signal Transduction with In Vitro Apoptosis Sensitivity in Chronic Lymphocytic Leukemia B Cells, PLoS ONE, vol.112, issue.10, p.24592, 2011.
DOI : 10.1371/journal.pone.0024592.s007

D. Bandura, V. Baranov, O. Ornatsky, A. Antonov, R. Kinach et al., Mass Cytometry: Technique for Real Time Single Cell Multitarget Immunoassay Based on Inductively Coupled Plasma Time-of-Flight Mass Spectrometry, Analytical Chemistry, vol.81, issue.16, pp.6813-6835, 2009.
DOI : 10.1021/ac901049w

A. Alizadeh, V. Aranda, A. Bardelli, C. Blanpain, C. Bock et al., Toward understanding and exploiting tumor heterogeneity, Nature Medicine, vol.21, issue.8, pp.846-53, 2015.
DOI : 10.1038/nm.3915

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4785013

P. Qiu, E. Simonds, S. Bendall, K. Gibbs, J. Bruggner et al., Extracting a cellular hierarchy from high-dimensional cytometry data with SPADE, Nature Biotechnology, vol.49, issue.10, pp.886-91, 2011.
DOI : 10.1371/journal.pcbi.1001123

L. Han, P. Qiu, Z. Zeng, J. Jorgensen, D. Mak et al., Single-cell mass cytometry reveals intracellular survival/proliferative signaling in FLT3-ITD-mutated AML stem/progenitor cells, Cytometry Part A, vol.304, issue.4, pp.346-56, 2015.
DOI : 10.1002/cyto.a.22628

E. Carpenter, J. Rader, J. Ruden, E. Rappaport, K. Hunter et al., Dielectrophoretic Capture and Genetic Analysis of Single Neuroblastoma Tumor Cells, Frontiers in Oncology, vol.338, p.201, 2014.
DOI : 10.1126/science.1229164

B. Polzer, G. Medoro, S. Pasch, F. Fontana, L. Zorzino et al., Molecular profiling of single circulating tumor cells with diagnostic intention, EMBO Molecular Medicine, vol.6, issue.11, pp.1371-86, 2014.
DOI : 10.15252/emmm.201404033

V. Murlidhar, N. Ramnath, S. Nagrath, and R. Reddy, Optimizing the Detection of Circulating Markers to Aid in Early Lung Cancer Detection, Cancers, vol.8, issue.7, 2016.
DOI : 10.3390/cancers8070061

L. Martelotto, C. Ng, S. Piscuoglio, B. Weigelt, and J. Reis-filho, Breast cancer intra-tumor heterogeneity, Breast Cancer Research, vol.497, issue.3, p.210, 2014.
DOI : 10.1038/nature12065

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4053234

S. Dey, L. Kester, B. Spanjaard, M. Bienko, and A. Van-oudenaarden, Integrated genome and transcriptome sequencing of the same cell, Nature Biotechnology, vol.10, issue.3, pp.285-294, 2015.
DOI : 10.1038/nbt.3129

M. Gerdes, A. Sood, C. Sevinsky, A. Pris, M. Zavodszky et al., Emerging Understanding of Multiscale Tumor Heterogeneity, Frontiers in Oncology, vol.20, issue.Suppl 6, p.366, 2014.
DOI : 10.1038/nm.3488