H. K. Brown, K. Schiavonne, F. Gouin, M. F. Heymann, and D. Heymann, Biology of Bone Sarcomas and New Therapeutic Developments, Calcified Tissue International, vol.14, issue.4, pp.174-195, 2018.
DOI : 10.1016/S1470-2045(13)70277-8

URL : https://hal.archives-ouvertes.fr/inserm-01653600

A. B. Mohseny, K. Szuhai, S. Romeo, A. P. Buddingh, I. Briaire-de-bruijn et al., Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2, J. Pathol, pp.219-294, 2009.

A. J. Mutsaers and C. R. Walkley, Cells of origin in osteosarcoma: Mesenchymal stem cells or osteoblast committed cells?, Bone, vol.62, pp.56-63, 2014.
DOI : 10.1016/j.bone.2014.02.003

A. I. Ségaliny, M. Tellez-gabriel, M. F. Heymann, and D. Heymann, Receptor tyrosine kinases: Characterisation, mechanism of action and therapeutic interests for bone cancers, Journal of Bone Oncology, vol.4, issue.1
DOI : 10.1016/j.jbo.2015.01.001

, Bone Oncol, vol.4, pp.1-12, 2015.

M. F. Heymann, H. K. Brown, and D. Heymann, Drugs in early clinical development for the treatment of osteosarcoma, Expert Opinion on Investigational Drugs, vol.50, issue.6, pp.1265-1280, 2016.
DOI : 10.1038/srep20944

URL : https://hal.archives-ouvertes.fr/inserm-01466096

A. Luetke, P. A. Meyers, I. Lewis, and H. Juergens, Osteosarcoma treatment ??? Where do we stand? A state of the art review, Cancer Treatment Reviews, vol.40, issue.4, pp.40-523, 2014.
DOI : 10.1016/j.ctrv.2013.11.006

A. Alfranca, L. Martinez-cruzado, J. Tornin, A. Abarrategi, T. Amaral et al.,

J. Menendez, R. Garcia-castro, and . Rodriguez, Bone microenvironment signals in osteosarcoma 17 development, Cell Mol. Life Sci, vol.72, pp.3097-3113, 2015.

M. F. Heymann, F. Lezot, and D. Heymann, The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma, Cellular Immunology
DOI : 10.1016/j.cellimm.2017.10.011

URL : https://hal.archives-ouvertes.fr/inserm-01644725

Q. Liu, H. Zhang, X. Jiang, C. Qian, Z. Liu et al., Factors involved in cancer metastasis: a better understanding to ???seed and soil??? hypothesis, Molecular Cancer, vol.501, issue.7467, 2017.
DOI : 10.1038/nature12517

R. S. Chiang, J. E. Cabrera, and . Segall, Am. J. Physiol. Cell. Physiol, pp.311-312, 2016.

X. X. Jie, C. J. Zhang, and . Xu, Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications, Oncotarget, vol.8, issue.46, pp.81558-81571, 2017.
DOI : 10.18632/oncotarget.18277

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=18277&path%5B%5D=58605

M. K. Pantel and . Speicher, The biology of circulating tumor cells, Oncogene, vol.195, issue.10, pp.1216-1240, 2016.
DOI : 10.1158/0008-5472.CAN-13-2030

K. C. Alix-panabières, Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy, Cancer Discovery, vol.6, issue.5, pp.479-491, 2016.
DOI : 10.1158/2159-8290.CD-15-1483

K. Pantel and C. , Liquid biopsy: Potential and challenges, Molecular Oncology, vol.73, issue.992-1003, pp.371-373, 2016.
DOI : 10.1158/0008-5472.CAN-12-0326

URL : https://hal.archives-ouvertes.fr/hal-01877343

A. Satelli, A. Mitra, J. J. Cutrera, M. Devarie, X. Xia et al.,

K. E. Somaiah, V. Torres, J. A. Ravi, E. S. Ludwig, S. Kleinerman et al., Universal marker and detection tool for human sarcoma circulating tumor cells, Cancer Res, pp.74-1645, 2014.

H. Ø. Bruland, G. Høifødt, S. Saeter, O. Smeland, and . Fodstad, Hematogenous Micrometastases in Osteosarcoma Patients, Clinical Cancer Research, vol.11, issue.13, pp.4666-4673, 2005.
DOI : 10.1158/1078-0432.CCR-05-0165

URL : http://clincancerres.aacrjournals.org/content/11/13/4666.full.pdf

Ø. S. Bruland, H. Høifødt, K. S. Hall, S. Smeland, and Ø. Fodstad, Bone Marrow Micrometastases Studied by an Immunomagnetic Isolation Procedure in Extremity Localized Non-metastatic Osteosarcoma Patients, Cancer Treat. Res, pp.152-509, 2009.
DOI : 10.1007/978-1-4419-0284-9_30

G. L. Chang, S. M. Asatrian, A. W. Dry, and . James, Circulating tumor cells in sarcomas: a brief review, Medical Oncology, vol.7, issue.3, pp.32-430, 2015.
DOI : 10.2147/OTT.S62349

M. Tellez-gabriel, H. K. Brown, R. Young, M. F. Heymann, and D. Heymann, The Challenges of Detecting Circulating Tumor Cells in Sarcoma, Frontiers in Oncology, vol.3, issue.10, 2016.
DOI : 10.1371/journal.pone.0003469

M. T. Gabriel, L. R. 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.62-571, 2016.
DOI : 10.1373/clinchem.2015.249706

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

C. L. Rodriguez-calleja, F. Jacques, and M. Lamoureux,

D. Quillard, P. Sahay, J. Perrot, C. Amiaud, and R. Charrier,

L. W. Heymann, B. Ellissen, and . Ory, ?Np63? silences a miRNA program to aberrantly initiate a 19

, wound-healing program that promotes TGF?-induced metastasis, Cancer Res, vol.76, pp.3236-3251, 2016.

A. A. Ségaliny, B. Mohamadi, A. Dizier, R. Lokajczyk, and R. Brion,

C. Charrier, D. Boisson-vidal, and . Heymann, Interleukin-34 promotes tumor progression and metastatic process in osteosarcoma through induction of angiogenesis and macrophage recruitment, Int. J. Cancer, vol.137, pp.73-85, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01644787

S. K. Sommer, I. H. Peters, M. Robins, G. J. Raap, S. Wiedemann et al., A preclinical model for experimental chemotherapy of human head and neck cancer, International Journal of Oncology, vol.18, pp.1145-1149, 2001.
DOI : 10.3892/ijo.18.6.1145

G. A. Odri, S. Dumoucel, G. Picarda, S. Battaglia, F. Lamoureux et al.,

F. Rousseau, K. Tirode, O. Laud, F. Delattre, D. Gouin et al., Zoledronic acid as a new adjuvant therapeutic strategy for Ewing's sarcoma patients, Cancer Res, vol.70, pp.7610-7619, 2010.

L. Hanly, R. Figueredo, M. J. Rieder, J. Koropatnick, and G. Koren, The effects of Nacetylcysteine on ifosfamide efficacy in a mouse xenograft model, Anticancer Res, vol.32, pp.3791-3798, 2012.

M. B. Gobin, F. Baudhuin, B. Lamoureux, C. Ory, and R. Charrier,

F. Redini, F. Lezot, D. Blanchard, and . Heymann, BYL719, a new ?-specific PI3K inhibitor: single administration and in combination with conventional chemotherapy for the treatment of osteosarcoma, Int. J. Cancer, vol.136, pp.784-796, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01644799

M. S. Regan-shaw, N. Nihal, and . Ahmad, Dose translation from animal to human studies revisited, The FASEB Journal, vol.22, issue.3, pp.659-661, 2007.
DOI : 10.1093/jnci/94.24.1883

M. Tellez-gabriel, C. Charrier, B. Brounais-le-royer, M. Mullard, H. K. Brown et al.,

D. Verrecchia and . Heymann, Analysis of gap junctional intercellular communications using a dielectrophoresis-based microchip, Eur. J. Cell Biol, vol.96, pp.110-118, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01466071

L. T. Chinen, C. A. Mello, E. A. Abdallah, L. M. Ocea, M. E. Buim et al.,

J. Gasparini, M. F. Fanelli, and P. Paterlini-bréchot, Isolation, detection, and immunomorphological characterization of circulating tumor cells (CTCs) from patients with different types of sarcoma using isolation by size of tumor cells: a window on sarcoma-cell invasion, OncoTargets Ther, pp.1609-1617, 2014.

Q. H. Li, H. Meng, I. S. Noh, N. Batth, K. E. Somaiah et al., Detection of circulating tumor cells from cryopreserved human sarcoma peripheral blood mononuclear cells, Cancer Letters, vol.403, pp.406-216, 2017.
DOI : 10.1016/j.canlet.2017.05.032

P. H. Zhang, X. Gao, M. Xiao, L. Heger, B. Geng et al.,

Y. Liu, X. Hu, S. Yu, L. Wu, Z. A. Wang et al., A liquid biopsy-based method for the detection and quantification of circulating tumor cells in surgical osteosarcoma patients, Int. J

, Oncol. in press

M. H. Zhang, Z. Li, P. H. Wang, Y. J. Lan, G. J. Lu et al., Detection and clinical significance of circulating tumor cells in osteosarcoma using immunofluorescence 21

, situ hybridization, pp.485-489, 2017.

M. Yu, S. Stott, M. Toner, S. Maheswaran, and D. A. Haber, Circulating tumor cells: approaches to isolation and characterization, The Journal of Cell Biology, vol.28, issue.3, pp.373-382, 2011.
DOI : 10.1016/j.chroma.2007.05.064

R. S. Stottl, S. Lee, M. Nagrath, D. T. Yu, L. Miyamoto et al.,

S. Ulman, Z. Springer, A. L. Nakamura, D. I. Moore, M. E. Tsukrov et al.,

. Maheswaran, Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer, Sci. Transl. Med, vol.2, 2010.

J. H. Jürgens, K. Treuner, and U. Winkler,

, Semin. Oncol, vol.16, pp.46-50, 1989.

S. D. Micalizzi, D. A. Maheswaran, and . Haber, A conduit to metastasis: circulating tumor cell biology, Genes & Development, vol.8, issue.18, pp.31-1827, 2017.
DOI : 10.1038/ncomms14344

L. Zhou, D. T. Dicker, E. Matthew, W. S. El-deiry, and R. K. Alpaugh, Circulating tumor cells: silent predictors of metastasis, F1000Research, vol.12, p.1445, 2017.
DOI : 10.1007/s11888-016-0320-y

M. S. Goldstein, C. M. Hayashi, K. W. Albert, D. M. Jackson, and . Loeb, An orthotopic xenograft model with survival hindlimb amputation allows investigation of the effect of tumor microenvironment on sarcoma metastasis, Clinical & Experimental Metastasis, vol.332, issue.9, pp.703-715, 2015.
DOI : 10.1016/j.jim.2007.11.012

M. E. Aanstoos, D. P. Regan, R. J. Rose, L. S. Chubb, and N. P. Ehrhart, , p.22

, Stromal Cells Influence Microscopic Residual or Metastatic Osteosarcoma in a Murine Model?, Clin. Orthop. Relat. Res, vol.474, pp.707-715, 2016.

C. Maloney, M. C. Edelman, M. P. Kallis, S. Z. Soffer, M. Symons et al., Intratibial Injection Causes Direct Pulmonary Seeding of Osteosarcoma Cells and Is Not a Spontaneous Model of Metastasis, Clinical Orthopaedics and Related Research, vol.476, issue.7
DOI : 10.1007/s11999.0000000000000291