M. Lopez-lazaro, The stem cell division theory of cancer, Crit Rev Oncol Hematol, vol.123, pp.95-113, 2018.

S. Paget, The distribution of secondary growths in cancer of the breast. 1889, Cancer Metastasis Rev, vol.8, pp.98-101, 1998.

M. Tellez-gabriel, C. Charrier, and B. Brounais-le-roeyr, Analysis of gap junctional intercellular communications using a dielectrophoresis-based microchip, Eur J Cell Biol, vol.96, pp.110-128, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01466071

I. Wee, N. Syn, and G. Sethi, Role of tumor-derived exosomes in cancer metastasis, Biochim Biophys Acta Rev Cancer, vol.1871, pp.12-21, 2018.

F. M. Vallette, C. Olivier, and F. Lézot, Dormant, quiescent, tolerant and persister cells: four synonyms for the same target in cancer, Biochem Pharmacol, vol.162, pp.169-176, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-01947474

T. Courau, D. Nehar-belaid, and L. Florez, TGF? and VEGF cooperatively control the immunotolerant environment and the efficacy of cancer immunotherapies, JCI Insight, vol.1, p.85974, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01617546

C. L. Mackall, P. S. Meltzer, and L. J. Helman, Focus on sarcomas, Cancer Cell, vol.2, pp.175-183, 2002.

M. Lopez-lazaro, Cancer etiology: Variation in cancer risk among tissues is poorly explained by the number of gene mutations, Genes Chromosomes Cancer, vol.57, pp.281-93, 2018.

C. Tomasetti, L. Macrhionni, and M. A. Nowak, Only three driver gene mutations are required for the development of lung and colorectal cancers, Proc Natl Acad Sci, vol.112, pp.118-141, 2015.

B. Vogelstein and K. W. Kinzler, The path to cancer --three strikes and You're out, N Engl J Med, vol.373, pp.1895-1903, 2015.

A. Chatterjee, E. J. Rodger, and M. R. Eccles, Epigenetic drivers of tumourigenesis and cancer metastasis, Semin Cancer Biol, vol.51, pp.149-59, 2018.

A. M. Noone, K. A. Cronin, and S. F. Altekruse, Cancer incidence and survival trends by subtype using data from the surveillance epidemiology and end results program, 1992-2013, Cancer Epidemiol Biomarkers Prev, vol.26, pp.632-673, 2017.

A. Vertii, P. D. Kaufman, and H. Hehnly, New dimensions of asymmetric division in vertebrates, Cytoskeleton, vol.75, pp.87-102, 2018.

A. Santoro, T. Vlachou, and M. Carminati, Molecular mechanisms of asymmetric divisions in mammary stem cells, EMBO Rep, vol.17, pp.1700-720, 2016.

C. Tomasetti, L. Li, and B. Vogelstein, Stem cell divisions, somatic mutations, cancer etiology, and cancer prevention, Science, vol.355, pp.1330-334, 2017.

C. Tomasetti and B. Vogelstein, Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions, Science, vol.347, pp.78-81, 2015.

F. Mitelman, B. Johansson, and F. Mertens, Cancer chromosome breakpoints cluster in generich genomic regions, Genes Chromosomes Cancer, vol.58, pp.149-54, 2019.

T. M. Grzywa, W. Praskal, and P. K. Wlodarski, Intratumor and Intertumor Heterogeneity in Melanoma, Transl Oncol, vol.10, pp.956-75, 2017.

H. K. Brown, M. Tellez-gabriel, and P. F. Cartron, Characterization of circulating tumor cells as a reflection of the tumor heterogeneity: myth or reality?, Drug Discov Today
URL : https://hal.archives-ouvertes.fr/inserm-01947472

G. Gundem, P. Van-loo, and B. Kremeyer, The evolutionary history of lethal metastatic prostate cancer, Nature, vol.520, pp.353-357, 2015.

M. K. Hong, G. Macintyre, and D. C. Wedge, Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer, Nat Commun, vol.6, p.6605, 2015.

I. Bedzhov, . Graham, . Sj, and C. Y. Leung, Developmental plasticity, cell fate specification and morphogenesis in the early mouse embryo, Philos Trans R Soc Lond B Biol Sci, vol.369, p.1657, 2014.

M. A. Nieto, R. Y. Huang, and R. A. Jackson, Cell, vol.166, pp.21-45, 2016.

M. T. Gabriel, L. R. Calleja, and A. Chalopin, Circulating tumor cells: a review of non-EpCAMbased approaches for cell enrichment and isolation, Clin Chem, vol.62, pp.571-81, 2016.

G. A. Franzetti, K. Laud-duval, and W. Van-der-ent, Cell-to cell heterogeneity of EWSR1-FLI1 activity determines proliferation/migration choices in Ewing sarcoma cells, Oncogene, vol.36, pp.3505-514, 2017.

S. Gambera, A. Abarrategi, and F. Gonzalez-camacho, Clonal dynamics in osteosarcoma defined by RGB marking, Nat Commun, vol.9, p.3994, 2018.

H. Zahreddine and K. L. Borden, Mechanisms and insights into drug resistance in cancer, Front Pharmacol, vol.4, p.28, 2013.

J. Zhu, L. Liang, and Y. Jiao, Enhanced invasion of metastatic cancer cells via extracellular matrix interface, PLoS One, vol.10, p.118058, 2015.

L. G. Martelotto, C. K. Ng, and S. Piscuoglio, Breast cancer intra-tumor heterogeneity, Breast Cancer Res, vol.16, p.210, 2014.

A. Jimenez-sanchez, D. Memon, and S. Pourpe, Heterogeneous tumor-immune microenvironments among differentially growing metastases in an ovarian cancer patient, Cell, vol.170, pp.927-938, 2017.

G. Malandrino, S. T. Finocchiaro, and P. Rossi, Multifunctional cadmium single source precursor for the selective deposition of CdO or CdS by a solution route, Chem Commun. (Camb), vol.45, pp.5681-683, 2005.

S. Josson, M. Gururajan, and S. Y. Sung, Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis, Oncogene, vol.34, pp.2690-699, 2015.

M. Chittezhath, M. K. Dhillon, and J. Y. Lim, Molecular profiling reveals a tumor-promoting phenotype of monocytes and macrophages in human cancer progression, Immunity, vol.41, pp.815-844, 2014.

K. Cyll, E. Ersvaer, and L. Vlatkovic, Tumour heterogeneity poses a significant challenge to cancer biomarker research, Br J Cancer, vol.117, pp.367-75, 2017.

I. Kümler, E. Balslev, and A. S. Knop, Expression patterns of biomarkers in primary tumors and corresponding metastases in breast cancer, Appl Immunohistochem Mol Morphol, vol.26, pp.13-19, 2018.

G. U. Erdem, K. Altundag, and N. Y. Ozdemir, Comparative study of receptor discordance between primary and corresponding metastatic lesions in breast cancer, J BUON, vol.22, pp.365-76, 2017.

A. , C. Pantel, and K. , Challenges in circulating tumour cell research, Nat Rev Cancer, vol.14, pp.623-654, 2014.

F. C. Bidard, S. Michiels, and S. Riethdorf, Circulating tumor cells in breast cancer patients treated by neoadjuvant chemotherapy: a meta-analysis, J Natl Cancer Inst, vol.110, pp.560-67, 2018.

S. De-wit, M. Manicone, and E. Rossi, EpCAM high and EpCAM low circulating tumor cells in metastatic prostate and breast cancer patients, Oncotarget, vol.9, pp.35705-716, 2018.

E. Munzone, F. Nolé, and A. Goldhirsch, Changes of HER2 status in circulating tumor cells compared with the primary tumor during treatment for advanced breast cancer, Clin Breast Cancer, vol.10, pp.392-97, 2010.

E. A. Punnoose, S. K. Atwal, and J. M. Spoerke, Molecular biomarker analyses using circulating tumor cells, PLoS One, vol.5, p.12517, 2010.

N. Beije, W. Onstenk, and J. Krann, Prognostic impact of HER2 and ER status of circulating tumor cells in metastatic breast cancer patients with a HER2-negative primary tumor, Neoplasia, vol.18, pp.647-53, 2016.

M. Wallwiener, A. D. Hartkopf, and S. Riethdorf, The impact of HER2 phenotype of circulating tumor cells in metastatic breast cancer: a retrospective study in 107 patients, BMC Cancer, vol.15, p.403, 2015.

M. Suhaimi, N. A. Foong, Y. M. Lee, and D. Y. , Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients, Mol Oncol, vol.9, pp.850-60, 2015.

C. Bingham, S. V. Fernandez, and P. Fittipaldi, Mutational studies on single circulating tumor cells isolated from the blood of inflammatory breast cancer patients, Breast Cancer Res Treat, vol.163, pp.219-249, 2017.

M. Pestrin, F. Salvianti, and F. Galardi, Heterogeneity of PIK3CA mutational status at the single cell level in circulating tumor cells from metastatic breast cancer patients, Mol Oncol, vol.9, pp.749-57, 2015.

E. Pailler, N. Auger, and C. R. Lindsay, High level of chromosomal instability in circulating tumor cells of ROS1-rearranged non-small-cell lung cancer, Ann Oncol, vol.26, pp.1408-1423, 2015.

S. C. Tsao, W. J. Wang, and Y. , Characterising the phenotypic evolution of circulating tumour cells during treatment, Nat Commun, vol.9, p.1482, 2018.

L. Kermanshah, M. Poudineh, and S. Ahmed, Dynamic CTC phenotypes in metastatic prostate cancer models visualized using magnetic ranking cytometry, Lab Chip, vol.18, pp.2055-64, 2018.

J. Lack, M. Gillard, and M. Cam, Circulating tumor cells capture disease evolution in advanced prostate cancer, J Transl Med, vol.15, p.44, 2017.

F. De-luca, G. Rotunno, and F. Savianti, Mutational analysis of single circulating tumor cells by next generation sequencing in metastatic breast cancer, Oncotarget, vol.7, pp.26107-26126, 2016.

D. T. Miyamoto, Y. Zheng, and B. S. Wittner, RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance, Science, vol.349, pp.1351-1357, 2015.

E. Pailler, M. Oulhen, and I. Borget, Circulating tumor cells with aberrant ALK copy number predict progression-free survival during crizotinib treatment in ALK-rearranged non-small cell lung cancer patients, Cancer Res, vol.77, pp.2222-230, 2017.

C. Paolillo, Detection of activating estrogen receptor gene (ESR1) mutations in single circulating tumor cells, Clin Cancer Res, vol.23, pp.6086-93, 2017.

I. Tirosh, B. Izar, and S. M. Prakadan, Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq, Science, vol.352, pp.189-96, 2016.

S. H. Au, B. D. Storey, and J. C. Moore, Clusters of circulating tumor cells traverse capillarysized vessels, Proc Natl Acad Sci, vol.113, pp.4947-4952, 2016.

N. Aceto, A. Bardia, and D. T. Miyamoto, Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis, Cell, vol.158, pp.1110-1122, 2014.

A. F. Sarioglu, N. Aceto, and N. Kojic, A microfluidic device for label-free, physical capture of circulating tumor cell clusters, Nat Methods, vol.12, pp.685-691, 2015.

T. Satake, A. Suetsugu, and M. Nakamura, Color-coded imaging of the circulating tumor cell microenvironment, Anticancer Res, vol.38, pp.5635-5638, 2018.

B. M. Szczerba, F. Castro-giner, and M. Vetter, Neutrophils escort circulating tumour cells to enable cell cycle progression, Nature, vol.566, pp.553-557, 2019.
DOI : 10.1038/s41586-019-0915-y

L. Dyck and K. Mills, Immune checkpoints and their inhibition in cancer and infectious diseases, Eur J Immunol, vol.47, pp.765-779, 2017.

M. F. Heymann, F. Lézot, and D. Heymann, The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma, Cell Immunol
URL : https://hal.archives-ouvertes.fr/inserm-01644725

D. K. Mishra, H. J. Rocha, and R. Miller, Immune cells inhibit the tumor metastasis in the 4D cellular lung model by reducing the number of live circulating tumor cells, Sci Rep, vol.8, p.16569, 2018.

M. Mazel, W. Jacot, and K. Pantel, Frequent expression of PD-L1 on circulating breast cancer cells, Mol Oncol, vol.9, pp.1773-82, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01162382

W. W. Sun, Z. H. Xu, and P. Lian, Characteristics of circulating tumor cells in organ metastases, prognosis, and T lymphocyte mediated immune response, Onco Targets Ther, vol.10, pp.2413-2424, 2017.

J. P. Arnoletti, N. Fanaian, and J. Reza, Pancreatic and bile duct cancer circulating tumor cells (CTC) form immune-resistant multi-cell type clusters in the portal venous circulation, Cancer Biol Ther, vol.19, pp.887-897, 2018.

X. Wang, Q. Sun, and Q. Liu, CTC immune escape mediated by Med Hypotheses, vol.93, pp.138-147, 2016.
DOI : 10.1016/j.mehy.2016.05.022

Z. Shen, A. Wu, and X. Chen, Current detection technologies for circulating tumor cells, Chem Soc Rev, vol.46, pp.2038-56, 2017.
DOI : 10.1039/c6cs00803h

URL : http://europepmc.org/articles/pmc5598784?pdf=render

M. Tellez-gabriel, B. Ory, F. Lamoureux, M. F. Heymann, and D. Heymann, Tumour heterogeneity: the key advantages of single-cell analysis, Int J Mol Sci, vol.17, p.2142, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01466086

C. Gawad, W. Koh, and S. R. Quake, Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics, Proc Natl Acad Sci, vol.111, pp.17947-52, 2014.

J. G. Lohr, V. A. Adalsteinsson, and K. Cibulskis, Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer, Nat Biotechnol, vol.32, pp.479-84, 2014.

R. Jiang, Y. T. Lu, and H. Ho, A comparison of isolated circulating tumor cells and tissue biopsies using whole-genome sequencing in prostate cancer, Oncotarget, vol.6, pp.44781-93, 2015.

C. F. De-bourcy, D. Vlaminck, and J. N. Kanbar, A quantitative comparison of single-cell whole genome amplification methods, PLoS One, vol.9, p.105585, 2014.

E. Borgström, M. Paterlini, and J. E. Mold, Comparison of whole genome amplification techniques for human single cell exome sequencing, PLoS One, vol.12, p.171566, 2017.

L. Deleye, L. Tilleman, V. Plaetsen, and A. S. , Performance of four modern whole genome amplification methods for copy number variant detection in single cells, Sci Rep, vol.7, p.3422, 2017.

B. Polzer, G. Medoro, and S. Pasch, Molecular profiling of single circulating tumor cells with diagnostic intention, EMBO Mol Med, vol.6, pp.1371-86, 2014.

M. Janiszewska, L. Liu, and V. Almendro, In situ single-cell analysis identifies heterogeneity for PIK3CA mutation and HER2 amplification in HER2-positive breast cancer, Nat Genet, vol.47, pp.1212-1221, 2015.

C. Li, S. Wu, and Z. Yang, Single-cell exome sequencing identifies mutations in KCP, LOC440040, and LOC440563 as drivers in renal cell carcinoma stem cells, Cell Res, vol.27, pp.590-93, 2017.

M. Liu, Y. Liu, and J. Di, Multi-region and single-cell sequencing reveal variable genomic heterogeneity in rectal cancer, BMC Cancer, vol.17, p.787, 2017.

X. Zhang, S. L. Marjani, and Z. Hu, Single-cell sequencing for precise cancer research: progress and prospects, Cancer Res, vol.76, pp.1305-1317, 2016.
DOI : 10.1158/0008-5472.can-15-1907

URL : http://cancerres.aacrjournals.org/content/76/6/1305.full.pdf

G. Russo, A. Patrignani, and L. Poveda, Highly sensitive, non-invasive detection of colorectal cancer mutations using single molecule, third generation sequencing, Appl Transl Genom, vol.7, pp.32-41, 2015.

A. Kilianski, P. A. Roth, and A. T. Liem, Use of unamplified RNA/cDNA-hybrid nanopore sequencing for rapid detection and characterization of RNA viruses, Emerg Infect Dis, vol.22, pp.1448-51, 2016.

D. Yalcin, Z. M. Hakguder, and H. H. Out, Bioinformatics approaches to single-cell analysis in developmental biology, Mol Hum Reprod, vol.22, pp.182-92, 2016.
DOI : 10.1093/molehr/gav050

URL : https://academic.oup.com/molehr/article-pdf/22/3/182/7427336/gav050.pdf

C. Chen, D. Xing, and L. Tan, Single-cell whole-genome analyses by linear amplification via Transposon Insertion (LIANTI), Science, vol.356, pp.189-94, 2017.
DOI : 10.1126/science.aak9787

URL : http://europepmc.org/articles/pmc5538131?pdf=render

T. N. Vu, Q. F. Wills, and K. R. Kalari, Beta-Poisson model for single-cell RNA-seq data analyses, Bioinformatics, vol.32, pp.2128-2163, 2016.
DOI : 10.1093/bioinformatics/btw202

URL : https://academic.oup.com/bioinformatics/article-pdf/32/14/2128/19568119/btw202.pdf

H. Guo, P. Zhu, and X. Wu, Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing, Genome Res, vol.23, pp.2126-2161, 2013.

S. A. Smallwood, H. J. Lee, and C. Angermueller, Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity, Nat Methods, vol.11, pp.817-837, 2014.

M. Farlik, N. C. Sheffield, and A. Nuzzo, Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics, Cell Rep, vol.10, pp.1386-97, 2015.

A. Rotem, O. Ram, and N. Shoresh, Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state, Nat Biotechnol, vol.33, pp.1165-72, 2015.
DOI : 10.1038/nbt.3383

URL : http://europepmc.org/articles/pmc4636926?pdf=render

J. Kind, L. Pagie, and S. S. De-vries, Genome-wide maps of nuclear lamina interactions in single human cells, Cell, vol.63, pp.134-181, 2015.

D. A. Cusanovich, R. Daza, and A. Adey, Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing, Science, vol.348, pp.910-914, 2015.

W. Jin, Q. Tang, and M. Wan, Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples, Nature, vol.28, pp.142-148, 2105.

T. Nagano, Y. Lubling, and T. J. Stevens, Single-cell Hi-C reveals cell-to-cell variability in chromosome structure, Nature, vol.502, pp.59-64, 2013.

S. J. Clark, H. J. Lee, and S. A. Smallwood, Single-cell epigenomics: powerful new methods for understanding gene regulation and cell identity, Genome Biol, vol.17, p.72, 2016.

C. F. Pixberg, K. Raba, and F. Müller, Analysis of DNA methylation in single circulating tumor cells, Oncogene, vol.36, pp.3223-231, 2017.

S. Zhu, T. Qing, and Y. Zheng, Advances in single-cell RNA sequencing and its applications in cancer research, Oncotarget, vol.8, pp.53763-779, 2017.

C. Ziegenhain, B. Vieth, and S. Parkh, Comparative analysis of single-cell RNA sequencing methods, Mol Cell, vol.65, pp.631-643, 2017.

M. A. Ortega, O. Poirion, and X. Zhu, Using single-cell multiple omics approaches to resolve tumor heterogeneity, Clin Transl Med, vol.6, p.46, 2017.

A. P. Patel, I. Tirsoh, and S. J. Trombetta, Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma, Science, vol.344, pp.1396-401, 2014.

W. Chung, H. H. Eum, and H. O. Lee, Single-cell RNA-seq enables comprehensive tumour and immune cell profiling in primary breast cancer, Nat Commun, vol.8, p.15081, 2017.

A. Bhan, M. Soleimani, and S. S. Mandal, Long noncoding RNA and cancer: a new paradigm, Cancer Res, vol.77, pp.3965-981, 2017.

M. Acunzo, G. Romano, and D. Wermicke, MicroRNA and cancer--a brief overview, Adv Biol Regul, vol.57, pp.1-9, 2015.

J. Abernathy and K. Overturf, Comparison of ribosomal RNA removal methods for transcriptome sequencing workflows in teleost fish, Anim Biotechnol, vol.27, pp.60-65, 2016.

A. R. Wu, N. F. Neff, and T. Kalisky, Quantitative assessment of single-cell RNA-sequencing methods, Nat Methods, vol.11, pp.41-47, 2014.

A. Suzuki, K. Matsushima, and H. Makinoshima, Single-cell analysis of lung adenocarcinoma cell lines reveals diverse expression patterns of individual cells invoked by a molecular target drug treatment, Genome Biol, vol.16, p.66, 2015.

C. A. Vallejos, D. Risso, and A. Sciadone, Normalizing single-cell RNA sequencing data: challenges and opportunities, Nat Methods, vol.14, pp.565-571, 2017.

Z. Wu, Y. Zhang, and M. L. Stitzel, Two-phase differential expression analysis for single cell RNA-seq, Bioinformatics, vol.34, pp.3340-348, 2018.

Z. Miao, K. Deng, and X. Wang, DEsingle for detecting three types of differential expression in single-cell RNA-seq data, Bioinformatics, vol.34, pp.3223-224, 2018.

J. R. Heath, A. Ribas, and P. S. Mischel, Single-cell analysis tools for drug discovery and development, Nat Rev Drug Discov, vol.15, pp.204-220, 2016.

A. V. Ullal, V. Peterson, and S. S. Agasti, Cancer cell profiling by barcoding allows multiplexed protein analysis in fine-needle aspirates, Sci Transl Med, vol.6, pp.219-228, 2014.

C. Giesen, H. A. Wang, and D. Schapiro, Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry, Nat Methods, vol.11, pp.417-439, 2014.

E. Sinkala, E. Sollier-christen, and C. Renier, Profiling protein expression in circulating tumour cells using microfluidic western blotting, Nat Commun, vol.8, p.14622, 2017.

N. Gulbahce, M. Magbanua, and R. Chin, Quantitative whole genome sequencing of circulating tumor cells enables personalized combination therapy of metastatic cancer, Cancer Res, vol.77, pp.4530-541, 2017.

R. Gao, A. Davis, and T. O. Mcdonald, Punctuated copy number evolution and clonal stasis in triple-negative breast cancer, Nat Genet, vol.48, pp.1119-1149, 2016.

C. Paoletti, A. K. Cani, and J. M. Larios, Comprehensive mutation and copy number profiling in archived circulating breast cancer tumor cells documents heterogeneous resistance mechanisms, Cancer Res, vol.78, pp.1110-122, 2018.

A. Carlsson, Paired high-content analysis of prostate cancer cells in bone marrow and blood characterizes increased androgen receptor expression in tumor cell clusters, Clin Cancer Res, vol.23, pp.1722-732, 2017.

Y. Liu, S. Meucci, and L. Sheng, Meta-analysis of the mutational status of circulation tumor cells and paired primary tumor tissues from colorectal cancer patients, Oncotarget, vol.8, pp.77928-941, 2017.

N. Guibert, M. Delaunay, and A. Lusque, PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab, Lung Cancer, vol.120, pp.108-120, 2018.

E. V. Kaigorodova, O. E. Savelieva, and L. A. Tashireva, Heterogeneity of circulating tumor cells in neoadjuvant chemotherapy of breast cancer, Molecules, p.23, 2018.

A. Jakabova, Z. Biecikova, and E. Pospisilova, Molecular characterization and heterogeneity of circulating tumor cells in breast cancer, Breast Cancer Res Treat, vol.166, pp.695-700, 2017.

K. E. Aaltonen, V. Novosadova, and P. O. Bendahl, Molecular characterization of circulating tumor cells from patients with metastatic breast cancer reflects evolutionary changes in gene expression under the pressure of systemic therapy, Oncotarget, vol.8, pp.45544-565, 2017.

C. Gasch, T. Oldopp, and O. Mauermann, Frequent detection of PIK3CA mutations in single circulating tumor cells of patients suffering from HER2-negative metastatic breast cancer

, Mol Oncol, vol.10, pp.1330-1373, 2016.

H. I. Scher, R. P. Graf, and N. A. Schreiber, Phenotypic heterogeneity of circulating tumor cells informs clinical decisions between AR signaling inhibitors and taxanes in metastatic prostate cancer, Cancer Res, vol.277, pp.5687-98, 2017.

L. Kermanshah, M. Poudineh, and S. Ahmed, Dynamic CTC phenotypes in metastatic prostate cancer models visualized using magnetic ranking cytometry, Lab Chip, issue.18, pp.2055-64, 2018.

A. Markou, M. Lazaridou, and P. Paraskevopoulos, Multiplex gene expression profiling of in vivo isolated circulating tumor cells in high-risk prostate cancer patients, Clin Chem, vol.264, pp.297-306, 2018.

Y. F. Sun, W. Guo, and Y. Xu, Circulating tumor cells from different vascular sites exhibit spatial heterogeneity in epithelial and mesenchymal composition and distinct clinical significance in hepatocellular carcinoma, Clin Cancer Res, vol.24, pp.547-59, 2018.

Y. Kondo, K. Hayashi, and K. Kawakami, KRAS mutation analysis of single circulating tumor cells from patients with metastatic colorectal cancer, BMC Cancer, vol.17, p.311, 2017.

K. Tan, S. M. Leong, and Z. Kee, Longitudinal monitoring reveals dynamic changes in circulating tumor cells (CTCs) and CTC-associated miRNAs in response to chemotherapy in metastatic colorectal cancer patients, Cancer Lett, vol.423, pp.1-8, 2018.

S. A. Joosse, F. R. Souche, and A. Babayan, Chromosomal aberrations associated with sequential steps of the metastatic Cascade in colorectal cancer patients, Clin Chem, vol.64, pp.1505-1512, 2018.

I. Messaritakis, D. Stoltidis, and A. Kotsakis, TTF-1-and/or CD56-positive circulating tumor cells in patients with small cell lung cancer (SCLC), Sci Rep, vol.7, p.45351, 2017.

A. Chalopin, M. Tellez-gabriel, and H. K. Brown, Isolation of circulating tumor cells in preclinical model of osteosarcoma: effect of chemotherapy, J Bone Oncol, vol.12, pp.83-90, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01851614

Y. Hou, H. Huo, and C. Cao, Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas, Cell Res, vol.26, pp.304-323, 2016.

D. Han, K. Chen, and J. Che, Detection of epithelial-mesenchymal transition status of circulating tumor cells in patients with esophageal squamous carcinoma, Biomed Res Int, p.7610154, 2018.

H. Polioudaki, S. Agelaki, and R. Chiotaki, Variable expression levels of keratin and vimentin reveal differential EMT status of circulating tumor cells and correlation with clinical characteristics and outcome of patients with metastatic breast cancer, BMC Cancer, vol.15, p.399, 2015.

S. Li, Q. Chen, and H. Li, Mesenchymal circulating tumor cells (CTCs) and OCT4 mRNA expression in CTCs for prognosis prediction in patients with non-small-cell lung cancer

, Clin Transl Oncol, vol.19, pp.1147-1153, 2017.

A. Markiewicz, M. We?nicka-ja?kiewicz, and B. Seroczy?ska, Epithelial-mesenchymal transition markers in lymph node metastases and primary breast tumors -relation to dissemination and proliferation, Am J Transl Res, vol.6, issue.6, pp.793-808, 2014.

S. Kasimir-bauer and O. Hoffmann, Expression of stem cell and epithelial-mesenchymal transition markers in primary breast cancer patients with circulating tumor cells, Breast Cancer Res, vol.14, p.15, 2012.

W. J. Allard, J. Matera, and M. C. Miller, Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases, Clin Cancer Res, vol.10, pp.6897-6904, 2004.

L. E. Lowes, D. Goodale, and Y. Xia, Epithelial-to-mesenchymal transition leads to diseasestage differences in circulating tumor cell detection and metastasis in pre-clinical models of prostate cancer, Oncotarget, vol.7, issue.46, pp.76125-76139, 2016.

M. A. Juratli, M. Sarimollaoglu, and E. R. Siegel, Real-time monitoring of circulating tumor cell release during tumor manipulation using in vivo photoacoustic and fluorescent flow cytometry, Head Neck, vol.36, pp.1207-1222, 2014.

L. S. Sasportas, S. S. Hori, and G. Pratx, Detection and quantitation of circulating tumor cell dynamics by bioluminescence imaging in an orthotopic mammary carcinoma model, PLoS One, vol.9, p.105079, 2014.

P. Savas, B. Virassamy, and C. Ye, Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis, Nat Med, vol.24, pp.986-93, 2018.

E. Castellanos, E. Feld, and L. Horn, Driven by Mutations: The predictive value of mutation subtype in EGFR-mutated non-small cell lung cancer, J Thorac Oncol, vol.12, pp.612-623, 2017.