G. Ottaviani and N. Jaffe, The Epidemiology of Osteosarcoma, Cancer Treat. Res, vol.152, pp.3-13, 2009.
DOI : 10.1007/978-1-4419-0284-9_1

S. Mallone and A. Tavilla, Rare cancers are not so rare: The rare cancer burden in Europe, Eur. J. Cancer, vol.47, pp.2493-2511, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00719112

J. H. Schwab, D. S. Springfield, K. A. Raskin, H. J. Mankin, and F. J. Hornicek, What???s New in Primary Bone Tumors, The Journal of Bone & Joint Surgery, vol.94, issue.20, pp.1913-1919, 2012.
DOI : 10.2106/JBJS.L.00955

S. S. Bielack, B. Kempf-bielack, G. Delling, G. U. Exner, S. Flege et al., Prognostic Factors in High-Grade Osteosarcoma of the Extremities or Trunk: An Analysis of 1,702 Patients Treated on Neoadjuvant Cooperative Osteosarcoma Study Group Protocols, Journal of Clinical Oncology, vol.20, issue.3, pp.776-790, 2002.
DOI : 10.1200/JCO.2002.20.3.776

L. Wei, Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma, Cell Rep, vol.2014, issue.7, pp.104-112

A. B. Shaikh, F. Li, M. Li, B. He, X. He et al., Present Advances and Future Perspectives of Molecular Targeted Therapy for Osteosarcoma, International Journal of Molecular Sciences, vol.25, issue.12, pp.17-506, 2016.
DOI : 10.1002/pbc.25062

Y. Wittrant, S. Théoleyre, C. Chipoy, M. Padrines, F. Blanchard et al., New therapeutic targets in bone tumours and associated osteolysis Bone sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, European Sarcoma Network Working Group, pp.49-57, 2004.

S. Ferrari and M. Serra, An update on chemotherapy for osteosarcoma, Expert Opinion on Pharmacotherapy, vol.133, issue.11, pp.2727-2736
DOI : 10.1371/journal.pone.0123082

P. K. Wu, W. M. Chen, C. F. Chen, O. K. Lee, C. K. Haung et al., Primary Osteogenic Sarcoma with Pulmonary Metastasis: Clinical Results and Prognostic Factors in 91 Patients, Japanese Journal of Clinical Oncology, vol.9, issue.1, pp.514-522, 2009.
DOI : 10.1080/1120009X.1994.11741153

URL : https://academic.oup.com/jjco/article-pdf/39/8/514/5415316/hyp057.pdf

M. Kansara, M. W. Teng, M. J. Smyth, and D. M. Thomas, Translational biology of osteosarcoma, Nature Reviews Cancer, vol.34, issue.11, pp.722-735, 2014.
DOI : 10.3109/02841869509127199

J. Whelan, A. Mctiernan, N. Cooper, Y. K. Wong, M. Francis et al., Incidence and survival of malignant bone sarcomas in England 1979-2007, International Journal of Cancer, vol.45, issue.Suppl 7, pp.508-517, 1979.
DOI : 10.1016/j.ejca.2008.11.003

X. H. Feng and R. Derynck, SPECIFICITY AND VERSATILITY IN TGF-?? SIGNALING THROUGH SMADS, Annual Review of Cell and Developmental Biology, vol.21, issue.1, pp.659-693, 2005.
DOI : 10.1146/annurev.cellbio.21.022404.142018

C. A. Harrison, S. L. Musawi, and K. L. Walton, Prodomains regulate the synthesis, extracellular localisation and activity of TGF-?? superfamily ligands, Growth Factors, vol.121, issue.5, pp.174-186, 2011.
DOI : 10.1016/j.cell.2010.07.011

J. Cárcamo, A. Zentella, and J. Massagué, Disruption of transforming growth factor beta signaling by a mutation that prevents transphosphorylation within the receptor complex., Molecular and Cellular Biology, vol.15, issue.3, pp.1573-1581, 1995.
DOI : 10.1128/MCB.15.3.1573

A. Moustakas and C. H. Heldin, The regulation of TGF?? signal transduction, Development, vol.136, issue.22, pp.3699-3714, 2009.
DOI : 10.1242/dev.030338

J. Massagué, TGF?? signalling in context, Nature Reviews Molecular Cell Biology, vol.18, issue.10, pp.616-630
DOI : 10.1016/j.ccr.2010.10.023

A. Hata and Y. G. Chen, TGF-?? Signaling from Receptors to Smads, Cold Spring Harbor Perspectives in Biology, vol.8, issue.9, 2016.
DOI : 10.1101/cshperspect.a022061

URL : http://cshperspectives.cshlp.org/content/8/9/a022061.full.pdf

C. H. Heldin and A. Moustakas, Signaling Receptors for TGF-?? Family Members, Cold Spring Harbor Perspectives in Biology, vol.8, issue.8, 2016.
DOI : 10.1101/cshperspect.a022053

URL : http://cshperspectives.cshlp.org/content/8/8/a022053.full.pdf

C. Chang, Agonists and Antagonists of TGF-?? Family Ligands, Cold Spring Harbor Perspectives in Biology, vol.8, issue.8
DOI : 10.1101/cshperspect.a021923

S. Dennler, S. Itoh, D. Vivien, P. Ten-dijke, S. Huet et al., Direct binding of Smad3 and Smad4 to critical TGF beta-inducible elements in the promoter of human plasminogen activator inhibitor-type 1 gene

A. Weiss and L. Attisano, The TGFbeta Superfamily Signaling Pathway, Wiley Interdisciplinary Reviews: Developmental Biology, vol.39, issue.1, pp.47-63
DOI : 10.1016/j.molcel.2010.07.011

Y. Shi and J. Massagué, Mechanisms of TGF-?? Signaling from Cell Membrane to the Nucleus, Cell, vol.113, issue.6, pp.685-700, 2003.
DOI : 10.1016/S0092-8674(03)00432-X

W. Shi, C. Sun, B. He, W. Xiong, X. Shi et al., GADD34???PP1c recruited by Smad7 dephosphorylates TGF?? type I receptor, The Journal of Cell Biology, vol.14, issue.2, pp.291-300, 2004.
DOI : 10.1016/S0962-8924(99)01579-2

URL : http://jcb.rupress.org/content/jcb/164/2/291.full.pdf

M. Lutz and P. Knaus, Integration of the TGF-beta pathway into the cellular signalling network. Cell Signal, pp.977-988, 2002.

D. Javelaud and A. Mauviel, Crosstalk mechanisms between the mitogen-activated protein kinase pathways and Smad signaling downstream of TGF-??: implications for carcinogenesis, Oncogene, vol.24, issue.37, pp.5742-5750, 2005.
DOI : 10.1074/jbc.274.19.13133

URL : https://hal.archives-ouvertes.fr/inserm-00147399

S. M. Seyedin, T. C. Thomas, A. Y. Thompson, D. M. Rosen, and K. A. Piez, Purification and characterization of two cartilage-inducing factors from bovine demineralized bone., Proc. Natl. Acad. Sci, pp.2267-2271, 1985.
DOI : 10.1073/pnas.82.8.2267

T. Matsumoto and M. Abe, TGF-??-related mechanisms of bone destruction in multiple myeloma, Bone, vol.48, issue.1, pp.129-134, 2011.
DOI : 10.1016/j.bone.2010.05.036

M. Wu, G. Chen, and Y. P. Li, TGF-?? and BMP signaling in osteoblast, skeletal development and bone formation, homeostasis and disease, Bone Research, vol.126, issue.1, 2016.
DOI : 10.1371/journal.pone.0020780

URL : http://www.nature.com/articles/boneres20169.pdf

K. Janssens, P. Ten-dijke, S. Janssens, and W. Van-hul, Transforming Growth Factor-??1 to the Bone, Endocrine Reviews, vol.26, issue.6, pp.743-774, 2005.
DOI : 10.1210/er.2004-0001

P. Juárez and T. A. Guise, TGF-?? in cancer and bone: Implications for treatment of bone metastases, Bone, vol.48, issue.1, pp.23-29, 2011.
DOI : 10.1016/j.bone.2010.08.004

T. Alliston, L. Choy, P. Ducy, G. Karsenty, and R. Derynck, TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation, The EMBO Journal, vol.20, issue.9, pp.2254-2272, 2001.
DOI : 10.1093/emboj/20.9.2254

S. Maeda, M. Hayashi, S. Komiya, T. Imamura, and K. Miyazono, Endogenous TGF-?? signaling suppresses maturation of osteoblastic mesenchymal cells, The EMBO Journal, vol.23, issue.3, pp.552-563, 2004.
DOI : 10.1038/sj.emboj.7600067

M. Karst, G. Gorny, R. J. Galvin, and M. J. Oursler, Roles of stromal cell RANKL, OPG, and M-CSF expression in biphasic TGF-? regulation of osteoclast differentiation, Journal of Cellular Physiology, vol.145, issue.1, pp.99-106, 2004.
DOI : 10.1128/MCB.10.9.4473

J. L. Crane, L. Xian, and X. Cao, Role of TGF-?? Signaling in Coupling Bone Remodeling, Methods Mol. Biol, vol.1344, pp.287-300, 2016.
DOI : 10.1007/978-1-4939-2966-5_18

G. Balooch, M. Balooch, R. K. Nalla, S. Schilling, E. H. Filvaroff et al., TGF-?? regulates the mechanical properties and composition of bone matrix, Proc. Natl. Acad. Sci. USA 2005, pp.18813-18818
DOI : 10.1016/S1535-6108(03)00135-1

K. S. Mohammad, C. G. Chen, G. Balooch, E. Stebbins, C. R. Mckenna et al., Pharmacologic Inhibition of the TGF-?? Type I Receptor Kinase Has Anabolic and Anti-Catabolic Effects on Bone, PLoS ONE, vol.19, issue.4, p.5275, 2009.
DOI : 10.1371/journal.pone.0005275.s001

A. Lamora, J. Talbot, G. Bougras, J. Amiaud, M. Leduc et al., Overexpression of Smad7 Blocks Primary Tumor Growth and Lung Metastasis Development in Osteosarcoma, Clinical Cancer Research, vol.20, issue.19, pp.5097-5112, 2014.
DOI : 10.1158/1078-0432.CCR-13-3191

A. B. Roberts and L. M. Wakefield, The two faces of transforming growth factor beta in carcinogenesis, Proc. Natl. Acad. Sci. USA 2003, pp.8621-8623

L. H. Katz, Y. Li, J. S. Chen, N. M. Muñoz, A. Majumdar et al., Targeting TGF-?? signaling in cancer, Expert Opinion on Therapeutic Targets, vol.7, issue.6, pp.743-760, 2013.
DOI : 10.1371/journal.pone.0039684

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3745214/pdf

D. R. Principe, J. A. Doll, J. Bauer, B. Jung, H. G. Munshi et al., TGF-??: Duality of Function Between Tumor Prevention and Carcinogenesis, JNCI Journal of the National Cancer Institute, vol.72, issue.4, p.106, 2014.
DOI : 10.1016/j.urology.2008.04.017

J. Han, C. A. Alvarez-breckenridge, Q. E. Wang, and J. Yu, TGF-? signaling and its targeting for glioma treatment, Am. J. Cancer Res. 2015, vol.5, pp.945-955

J. Sheng, W. Chen, and H. J. Zhu, ) in human diseases, Growth Factors, vol.586, issue.2, pp.92-101, 2015.
DOI : 10.1016/j.febslet.2012.03.063

J. Wang, N. Shao, X. Ding, B. Tan, Q. Song et al., Crosstalk between transforming growth factor-?? signaling pathway and long non-coding RNAs in cancer, Cancer Letters, vol.370, issue.2, pp.296-301, 2016.
DOI : 10.1016/j.canlet.2015.11.007

K. Pardali, A. Kurisaki, A. Morén, P. Ten-dijke, D. Kardassis et al., Regulation by Transforming Growth Factor-??, Journal of Biological Chemistry, vol.10, issue.38, pp.29244-29256, 2000.
DOI : 10.1006/bbrc.1997.6909

P. Staller, K. Peukert, A. Kiermaier, J. Seoane, J. Lukas et al., Repression of p15INK4b expression by Myc through association with Miz-1, Nature Cell Biology, vol.19, issue.4, pp.392-399, 2001.
DOI : 10.1128/MCB.19.7.4672

J. Seoane, H. V. Le, L. Shen, S. A. Anderson, and J. Massagué, Integration of Smad and Forkhead Pathways in the Control of Neuroepithelial and Glioblastoma Cell Proliferation, Cell, vol.117, issue.2, pp.211-223, 2004.
DOI : 10.1016/S0092-8674(04)00298-3

P. M. Siegel and J. Massagué, Cytostatic and apoptotic actions of TGF-?? in homeostasis and cancer, Nature Reviews Cancer, vol.3, issue.11, pp.807-821, 2003.
DOI : 10.1038/nrc1208

S. Senturk, M. Mumcuoglu, O. Gursoy-yuzugullu, B. Cingoz, K. C. Akcali et al., Transforming growth factor-beta induces senescence in hepatocellular carcinoma cells and inhibits tumor growth, Hepatology, vol.62, issue.Suppl. 1, pp.966-974, 2010.
DOI : 10.1159/000048275

URL : http://onlinelibrary.wiley.com/doi/10.1002/hep.23769/pdf

L. Levy and C. S. Hill, Alterations in components of the TGF-?? superfamily signaling pathways in human cancer, Cytokine & Growth Factor Reviews, vol.17, issue.1-2, pp.41-58, 2006.
DOI : 10.1016/j.cytogfr.2005.09.009

Y. Drabsch, Ten Dijke, P. TGF-? signalling and its role in cancer progression and metastasis, Cancer Metastasis Rev, vol.2012, issue.31, pp.553-568

S. A. Hahn, M. Schutte, A. T. Hoque, C. A. Moskaluk, L. T. Da-costa et al., DPC4, A Candidate Tumor Suppressor Gene at Human Chromosome 18q21.1, Science, vol.271, issue.5247, pp.350-353, 1996.
DOI : 10.1126/science.271.5247.350

M. Miyaki, T. Iijima, M. Konishi, K. Sakai, A. Ishii et al., Higher frequency of Smad4 gene mutation in human colorectal cancer with distant metastasis, Oncogene, vol.18, issue.20, pp.3098-3103, 1999.
DOI : 10.1038/383168a0

S. Izumoto, N. Arita, T. Ohnishi, S. Hiraga, T. Taki et al., Microsatellite instability and mutated type II transforming growth factor-?? receptor gene in gliomas, Cancer Letters, vol.112, issue.2, pp.251-256, 1997.
DOI : 10.1016/S0304-3835(96)04583-1

M. G. Brattain, J. Chang, and S. J. Kim, Mutational inactivation of transforming growth factor-? receptor type II in microsatellite stable colon cancers, Cancer Res, vol.59, pp.320-324, 1999.

B. Bierie and H. L. Moses, TGF??: the molecular Jekyll and Hyde of cancer, Nature Reviews Cancer, vol.274, issue.7, pp.506-520, 2006.
DOI : 10.1126/science.274.5291.1363

R. A. Walker and S. J. Dearing, Transforming growth factor beta1 in ductal carcinoma in situ and invasive carcinomas of the breast, European Journal of Cancer, vol.28, issue.2-3, pp.641-644, 1992.
DOI : 10.1016/S0959-8049(05)80116-9

E. Friedman, L. I. Gold, D. Klimstra, Z. S. Zeng, S. Winawer et al., High levels of transforming growth factor-?1 correlate with disease progression in human colon cancer, Cancer Epidemiol. Biomark. Prev, vol.4, pp.549-554, 1995.

P. Wikström, P. Stattin, I. Franck-lissbrant, J. E. Damber, and A. Bergh, Transforming growth factor ??1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer, The Prostate, vol.104, issue.1, pp.19-29, 1998.
DOI : 10.1016/0304-3835(96)04243-7

J. Zavadil, L. Cermak, N. Soto-nieves, and E. P. Böttinger, Integration of TGF-??/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition, The EMBO Journal, vol.23, issue.5, pp.1155-1165, 2004.
DOI : 10.1038/sj.emboj.7600069

G. Han, S. L. Lu, A. G. Li, W. He, C. L. Corless et al., Distinct mechanisms of TGF-??1-mediated epithelial-to-mesenchymal transition and metastasis during skin carcinogenesis, Journal of Clinical Investigation, vol.115, issue.7, pp.1714-1723, 2005.
DOI : 10.1172/JCI24399

Y. Katsuno, S. Lamouille, and R. Derynck, TGF-?? signaling and epithelial???mesenchymal transition in cancer progression, Current Opinion in Oncology, vol.25, issue.1, pp.76-84, 2013.
DOI : 10.1097/CCO.0b013e32835b6371

URL : http://pdfs.journals.lww.com/co-oncology/2013/01000/TGF___signaling_and_epithelial_mesenchymal.14.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1506646807765;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzd1e6uV2h8gb8tPnf1XZERZ6/dAtCdx648Kq/I2MFrHNLanbakVBGnXm2eE4J7H209;hash|pP+SxBReV4FX6h5KwhfckQ==

R. Derynck, B. P. Muthusamy, and K. Y. Saeteurn, Signaling pathway cooperation in TGF-??-induced epithelial???mesenchymal transition, Current Opinion in Cell Biology, vol.31, issue.31, pp.56-66
DOI : 10.1016/j.ceb.2014.09.001

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4657734/pdf

H. Peinado, M. Quintanilla, and A. Cano, Transforming Growth Factor ??-1 Induces Snail Transcription Factor in Epithelial Cell Lines, Journal of Biological Chemistry, vol.47, issue.23, pp.21113-21123, 2003.
DOI : 10.1016/S0092-8674(00)00121-5

URL : http://www.jbc.org/content/278/23/21113.full.pdf

H. Peinado, E. Ballestar, M. Esteller, and A. Cano, Snail mediates E-cadherin repression by the recruitment of the Sin3A, HDAC1)/HDAC2 complex, pp.306-319, 2004.

R. Vogelmann, M. D. Nguyen-tat, K. Giehl, G. Adler, D. Wedlich et al., TGF??-induced downregulation of E-cadherin-based cell-cell adhesion depends on PI3-kinase and PTEN, Journal of Cell Science, vol.118, issue.20, pp.4901-4912, 2005.
DOI : 10.1242/jcs.02594

K. Polyak and R. A. Weinberg, Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits, Nature Reviews Cancer, vol.103, issue.4, pp.265-273, 2009.
DOI : 10.1038/nrc1279

E. C. Connolly, J. Freimuth, and R. J. Akhurst, Complexities of TGF-?? Targeted Cancer Therapy, International Journal of Biological Sciences, vol.8, issue.7, pp.964-978
DOI : 10.7150/ijbs.4564

H. G. Hagedorn, B. E. Bachmeier, and A. G. Nerlich, Synthesis and degradation of basement membranes and extracellular matrix and their regulation by TGF-?? in invasive carcinomas (Review), International Journal of Oncology, vol.18, pp.669-681, 2001.
DOI : 10.3892/ijo.18.4.669

D. Padua and J. Massagué, Roles of TGF?? in metastasis, Cell Research, vol.49, issue.1, pp.89-102, 2009.
DOI : 10.1038/nature03799

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. Carmeliet and R. K. Jain, Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases, Nature Reviews Drug Discovery, vol.7, issue.6, pp.417-427, 2011.
DOI : 10.1038/nmeth.1475

D. Jong, J. S. Van-diest, P. J. Van-der-valk, P. Baak, and J. P. , Expression of growth factors, growth-inhibiting factors, and their receptors in invasive breast cancer. II: Correlations with proliferation and angiogenesis, The Journal of Pathology, vol.12, issue.1, pp.53-57, 1998.
DOI : 10.1200/JCO.1994.12.3.454

N. Ito, S. Kawata, S. Tamura, Y. Shirai, S. Kiso et al., Positive correlation of plasma transforming growth factor-??1 levels with tumor vascularity in hepatocellular carcinoma, Cancer Letters, vol.89, issue.1, pp.45-48, 1995.
DOI : 10.1016/0304-3835(95)90156-6

V. Ivanovic, A. Melman, B. Davis-joseph, M. Valcic, and J. Geliebter, Elevated plasma levels of TGF-??1 in patients with invasive prostate cancer, Nature Medicine, vol.87, issue.4, pp.282-284, 1995.
DOI : 10.1111/j.1349-7006.1992.tb01964.x

U. Junker, B. Knoefel, K. Nuske, K. Rebstock, T. Steiner et al., Transforming Growth Factor Beta 1 is Significantly Elevated in Plasma of Patients Suffering from Renal Cell Carcinoma, Cytokine, vol.8, issue.10, pp.794-798, 1996.
DOI : 10.1006/cyto.1996.0105

M. M. Shull, I. Ormsby, A. B. Kier, S. Pawlowski, R. J. Diebold et al., Targeted disruption of the mouse transforming growth factor-??1 gene results in multifocal inflammatory disease, Nature, vol.359, issue.6397, pp.693-699, 1992.
DOI : 10.1038/359693a0

R. S. Yang, C. T. Wu, K. H. Lin, R. L. Hong, T. K. Liu et al., Relation between Histological Intensity of Transforming Growth Factor-.BETA. Isoforms in Human Osteosarcoma and the Rate of Lung Metastasis., The Tohoku Journal of Experimental Medicine, vol.184, issue.2, pp.133-142, 1998.
DOI : 10.1620/tjem.184.133

S. Xu, S. Yang, G. Sun, W. Huang, and Y. Zhang, Transforming Growth Factor-Beta Polymorphisms and Serum Level in the Development of Osteosarcoma, DNA and Cell Biology, vol.33, issue.11, pp.802-806, 2014.
DOI : 10.1089/dna.2014.2527

A. Franchi, L. Arganini, G. Baroni, A. Calzolari, R. Capanna et al., Expression of transforming growth factor ?? isoforms in osteosarcoma variants: association of tgf??1 with high-grade osteosarcomas, The Journal of Pathology, vol.11, issue.3, pp.284-289, 1998.
DOI : 10.1002/jbmr.5650110210

M. B. Mintz, R. Sowers, K. M. Brown, S. C. Hilmer, B. Mazza et al., An Expression Signature Classifies Chemotherapy-Resistant Pediatric Osteosarcoma, Cancer Research, vol.65, issue.5, pp.1748-1754, 2005.
DOI : 10.1158/0008-5472.CAN-04-2463

Y. Huang, Y. Yang, R. Gao, X. Yang, X. Yan et al., RLIM interacts with Smurf2 and promotes TGF-?? induced U2OS cell migration, Biochemical and Biophysical Research Communications, vol.414, issue.1, pp.181-185, 2011.
DOI : 10.1016/j.bbrc.2011.09.053

A. Kunita, T. G. Kashima, A. Ohazama, A. E. Grigoriadis, and M. Fukayama, Podoplanin Is Regulated by AP-1 and Promotes Platelet Aggregation and Cell Migration in Osteosarcoma, The American Journal of Pathology, vol.179, issue.2, pp.1041-1049, 2011.
DOI : 10.1016/j.ajpath.2011.04.027

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3157255/pdf

J. Chen, Y. Song, J. Yang, L. Gong, P. Zhao et al., The up-regulation of cysteine-rich protein 61 induced by transforming growth factor beta enhances osteosarcoma cell migration, Molecular and Cellular Biochemistry, vol.281, issue.1-2, pp.269-277, 2013.
DOI : 10.1074/jbc.M610172200

J. Y. Sung, S. Y. Park, J. H. Kim, H. G. Kang, J. H. Yoon et al., Interferon consensus sequence-binding protein (ICSBP) promotes epithelial-to-mesenchymal transition (EMT)-like phenomena, cell-motility and invasion via TGF-?? signaling in U2OS cells, Cell Death & Disease, vol.91, issue.5, p.1224, 2014.
DOI : 10.1006/excr.2000.4919

URL : http://www.nature.com/cddis/journal/v5/n5/pdf/cddis2014189a.pdf

G. M. Quan and P. Choong, Anti-angiogenic therapy for osteosarcoma, Cancer and Metastasis Reviews, vol.38, issue.4, pp.707-713, 2006.
DOI : 10.1177/002215549804600904

M. Tsubaki, T. Satou, T. Itoh, J. Kaneko, Y. Kidera et al., Blockade of the Ras/MEK/ERK and Ras/PI3K/Akt pathways by statins reduces the expression of bFGF, HGF, and TGF-?? as angiogenic factors in mouse osteosarcoma, ERK and Ras/PI3K/Akt pathways by statins reduces the expression of bFGF, HGF, and TGF-? as angiogenic factors in mouse osteosarcoma, pp.100-107, 2011.
DOI : 10.1016/j.cyto.2011.01.005

M. Kawano, I. Itonaga, T. Iwasaki, H. Tsuchiya, and H. Tsumura, Anti-TGF-?? Antibody Combined with Dendritic Cells Produce Antitumor Effects in Osteosarcoma, Clinical Orthopaedics and Related Research??, vol.16, issue.8, pp.2288-2294, 2012.
DOI : 10.1158/1078-0432.CCR-09-1634

A. Lamora, M. Mullard, J. Amiaud, R. Brion, D. Heymann et al., Anticancer activity of halofuginone in a preclinical model of osteosarcoma: inhibition of tumor growth and lung metastases, Oncotarget, vol.6, issue.16, pp.14413-14427, 2015.
DOI : 10.18632/oncotarget.3891

S. Yarkoni, Amelioration of radiation-induced fibrosis: Inhibition of transforming growth factor-beta signaling by halofuginone, J. Biol. Chem, vol.279, pp.15167-15176, 2004.

W. J. Boyle, W. S. Simonet, and D. L. Lacey, Osteoclast differentiation and activation, Nature, vol.20, issue.Suppl., pp.337-342, 2003.
DOI : 10.1007/s007740200049

URL : http://www.nature.com/nature/journal/v423/n6937/pdf/nature01658.pdf

J. J. Yin, K. Selander, J. M. Chirgwin, M. Dallas, B. G. Grubbs et al., TGF-?? signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development, Journal of Clinical Investigation, vol.103, issue.2, pp.197-206, 1999.
DOI : 10.1172/JCI3523

URL : http://www.jci.org/articles/view/3523/files/pdf

D. Javelaud, V. Delmas, M. Möller, P. Sextius, J. André et al., Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo, Oncogene, vol.24, issue.51, pp.7624-7629, 2005.
DOI : 10.1074/jbc.M309798200

URL : https://hal.archives-ouvertes.fr/inserm-00147458

D. Javelaud, K. S. Mohammad, C. R. Mckenna, P. Fournier, F. Luciani et al., Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis, Cancer Research, vol.67, issue.5, pp.2317-2324, 2007.
DOI : 10.1158/0008-5472.CAN-06-3950

URL : https://hal.archives-ouvertes.fr/inserm-00147397

K. S. Mohammad, D. Javelaud, P. G. Fournier, M. Niewolna, C. R. Mckenna et al., TGF-??-RI Kinase Inhibitor SD-208 Reduces the Development and Progression of Melanoma Bone Metastases, Cancer Research, vol.71, issue.1, pp.175-184, 2011.
DOI : 10.1158/0008-5472.CAN-10-2651

R. J. Akhurst and A. Hata, Targeting the TGF?? signalling pathway in disease, Nature Reviews Drug Discovery, vol.298, issue.10, pp.790-811
DOI : 10.1152/ajpheart.01048.2009

P. Hau, P. Jachimczak, R. Schlingensiepen, F. Schulmeyer, T. Jauch et al., 2 with AP 12009 in Recurrent Malignant Gliomas: From Preclinical to Phase I/II Studies, Oligonucleotides, vol.17, issue.2, pp.201-212, 2007.
DOI : 10.1089/oli.2006.0053

A. Balasubramaniam, S. Nair, V. Oliushine, and V. Parfenov, Targeted therapy for high-grade glioma with the TGF-?2 inhibitor trabedersen: Results of a randomized and controlled phase IIb study, Neuro-Oncology, vol.13, pp.132-142, 2011.

J. C. Morris, A. R. Tan, T. E. Olencki, G. I. Shapiro, B. J. Dezube et al., Phase I Study of GC1008 (Fresolimumab): A Human Anti-Transforming Growth Factor-Beta (TGF??) Monoclonal Antibody in Patients with Advanced Malignant Melanoma or Renal Cell Carcinoma, PLoS ONE, vol.133, issue.3, p.90353, 2014.
DOI : 10.1371/journal.pone.0090353.s005

R. Ahnert, J. Baselga, J. Calvo, E. Seoane, J. Brana et al., First human dose (FHD) study of the oral transforming growth factor-beta receptor I kinase inhibitor LY2157299 in patients with treatment-refractory malignant glioma, J. Clin. Oncol, pp.29-3011, 2011.