M. Macias, P. Martin-malpartida, and J. Massagué, Structural determinants of Smad function in TGF-?? signaling, Trends in Biochemical Sciences, vol.40, issue.6, pp.296-308, 2015.
DOI : 10.1016/j.tibs.2015.03.012

URL : http://europepmc.org/articles/pmc4485443?pdf=render

A. Hata and Y. 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. 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

T. Tsukazaki, T. Chiang, A. Davison, L. Attisano, and J. Wrana, SARA, a FYVE Domain Protein that Recruits Smad2 to the TGF?? Receptor, Cell, vol.95, issue.6, pp.779-91, 1998.
DOI : 10.1016/S0092-8674(00)81701-8

URL : https://doi.org/10.1016/s0092-8674(00)81701-8

S. Dennler, S. Itoh, D. Vivien, P. Ten-dijke, S. Huet et al., Direct binding of Smad3 and Smad4 to critical TGFbeta -inducible elements in the promoter of human plasminogen activator inhibitor-type 1gene, The EMBO Journal, vol.17, issue.11, pp.3091-100, 1998.
DOI : 10.1093/emboj/17.11.3091

A. Nakao, M. Afrakhte, A. Morén, T. Nakayama, J. Christian et al., Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling, Nature, vol.389, pp.631-636, 1997.

P. Kavsak, R. Rasmussen, C. Causing, S. Bonni, H. Zhu et al., Smad7 Binds to Smurf2 to Form an E3 Ubiquitin Ligase that Targets the TGF?? Receptor for Degradation, Molecular Cell, vol.6, issue.6, pp.1365-75, 2000.
DOI : 10.1016/S1097-2765(00)00134-9

URL : https://doi.org/10.1016/s1097-2765(00)00134-9

T. Ebisawa, M. Fukuchi, G. Murakami, T. Chiba, K. Tanaka et al., Smurf1 Interacts with Transforming Growth Factor-?? Type I Receptor through Smad7 and Induces Receptor Degradation, Journal of Biological Chemistry, vol.112, issue.16, pp.12477-80, 2001.
DOI : 10.1016/S1097-2765(00)00134-9

URL : http://www.jbc.org/content/276/16/12477.full.pdf

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

Y. Zhang, Mechanistic insight into contextual TGF-?? signaling, Current Opinion in Cell Biology, vol.51, 2017.
DOI : 10.1016/j.ceb.2017.10.001

K. Yamaguchi, K. Shirakabe, H. Shibuya, K. Irie, I. Oishi et al., Identification of a Member of the MAPKKK Family as a Potential Mediator of TGF-beta Signal Transduction, Science, vol.270, issue.5244, pp.2008-2019, 1995.
DOI : 10.1126/science.270.5244.2008

M. Yamashita, K. Fatyol, C. Jin, X. Wang, Z. Liu et al., TRAF6 Mediates Smad-Independent Activation of JNK and p38 by TGF-??, Molecular Cell, vol.31, issue.6, pp.918-942, 2008.
DOI : 10.1016/j.molcel.2008.09.002

URL : https://doi.org/10.1016/j.molcel.2008.09.002

A. Hamidi, J. Song, N. Thakur, S. Itoh, A. Marcusson et al., TGF-?? promotes PI3K-AKT signaling and prostate cancer cell migration through the TRAF6-mediated ubiquitylation of p85??, Science Signaling, vol.47, issue.486, 2017.
DOI : 10.1002/(SICI)1096-9888(199902)34:2<105::AID-JMS768>3.0.CO;2-4

A. Roberts and L. Wakefield, The two faces of transforming growth factor ?? in carcinogenesis, Proceedings of the National Academy of Sciences, vol.109, issue.12, pp.8621-8624, 2003.
DOI : 10.1172/JCI200215234

J. Huang and G. Blobe, Dichotomous roles of TGF-?? in human cancer, Biochemical Society Transactions, vol.44, issue.5, pp.1441-54, 2016.
DOI : 10.1042/BST20160065

T. Yokobori and M. Nishiyama, TGF-?? Signaling in Gastrointestinal Cancers: Progress in Basic and Clinical Research, Journal of Clinical Medicine, vol.61, issue.1, p.10, 2017.
DOI : 10.1186/1471-2407-12-239

URL : http://www.mdpi.com/2077-0383/6/1/11/pdf

B. Costanza, I. Umelo, J. Bellier, V. Castronovo, and A. Turtoi, Stromal Modulators of TGF-?? in Cancer, Journal of Clinical Medicine, vol.9, issue.1, p.10, 2017.
DOI : 10.1371/journal.pone.0083786

URL : http://www.mdpi.com/2077-0383/6/1/7/pdf

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.29244-56, 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
DOI : 10.1128/MCB.19.7.4672

, Nat Cell Biol, vol.3, pp.392-401, 2001.

S. Zelivianski, A. Cooley, R. Kall, and J. Jeruss, Cyclin-Dependent Kinase 4-Mediated Phosphorylation Inhibits Smad3 Activity in Cyclin D-Overexpressing Breast Cancer Cells, Molecular Cancer Research, vol.8, issue.10, pp.1375-87, 2010.
DOI : 10.1158/1541-7786.MCR-09-0537

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.3098-103, 1999.
DOI : 10.1038/383168a0

R. Verrecchia and . Signaling, Microenvironment in Osteosarcoma Frontiers in Oncology | www.frontiersin, p.133, 2018.

W. Grady, L. Myeroff, S. Swinler, A. Rajput, S. Thiagalingam et al., Mutational inactivation of transforming growth factor beta receptor type II in microsatellite stable colon cancers, Cancer Res, vol.59, pp.320-324, 1999.

S. Matsuyama, M. Iwadate, M. Kondo, M. Saitoh, A. Hanyu et al., SB-431542 and Gleevec inhibit transforming growth factor-beta-induced proliferation of human osteosarcoma cells, Cancer Res, vol.63, pp.7791-7799, 2003.

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.5097-112, 2014.
DOI : 10.1158/1078-0432.CCR-13-3191

R. Yang, C. Wu, K. Lin, R. Hong, T. 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.133-175, 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.802-808, 2014.
DOI : 10.1089/dna.2014.2527

C. Morrison, J. Parvani, and W. Schiemann, The relevance of the TGF-?? Paradox to EMT-MET programs, Cancer Letters, vol.341, issue.1, pp.30-40, 2013.
DOI : 10.1016/j.canlet.2013.02.048

X. Jie, X. Zhang, and C. Xu, Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications, Oncotarget, vol.8, issue.46, pp.81558-71, 2017.
DOI : 10.18632/oncotarget.18277

R. Derynck, B. Muthusamy, and K. Saeteurn, Signaling pathway cooperation in TGF-??-induced epithelial???mesenchymal transition, Current Opinion in Cell Biology, vol.31, pp.56-66, 2014.
DOI : 10.1016/j.ceb.2014.09.001

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

S. Lamouille, J. Xu, and R. Derynck, Molecular mechanisms of epithelial???mesenchymal transition, Nature Reviews Molecular Cell Biology, vol.22, issue.3, pp.178-96, 2014.
DOI : 10.1016/j.ccr.2012.10.012

G. Yang, J. Yuan, and K. Li, EMT transcription factors: implication in osteosarcoma, Medical Oncology, vol.9, issue.4, p.697, 2013.
DOI : 10.1158/1541-7786.MCR-11-0093

P. Lei, D. Ding, J. Xie, L. Wang, Q. Liao et al., Expression profile of Twist, vascular endothelial growth factor and CD34 in patients with different phases of osteosarcoma, Oncology Letters, vol.10, issue.1, pp.417-438, 2015.
DOI : 10.3892/ol.2015.3246

J. Wu, Q. Liao, H. He, D. Zhong, and K. Yin, TWIST interacts with ??-catenin signaling on osteosarcoma cell survival against cisplatin, Molecular Carcinogenesis, vol.222, issue.6, pp.440-446, 2014.
DOI : 10.1038/222385a0

A. Sharili, S. Allen, K. Smith, J. Hargreaves, J. Price et al., Expression of Snail2 in long bone osteosarcomas correlates with tumour malignancy, Tumor Biology, vol.454, issue.3, pp.515-541, 2011.
DOI : 10.1097/BLO.0b013e31802b683c

A. Shen, Y. Zhang, H. Yang, R. Xu, and G. Huang, Overexpression of ZEB1 relates to metastasis and invasion in osteosarcoma, Journal of Surgical Oncology, vol.13, issue.8, pp.830-834, 2012.
DOI : 10.1002/1520-6300(200102/03)13:2<235::AID-AJHB1034>3.0.CO;2-M

J. Sung, S. Park, J. Kim, H. Kang, J. 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, 2014.
DOI : 10.1006/excr.2000.4919

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, 2011.
DOI : 10.1016/j.bbrc.2011.09.053

A. Kunita, T. Kashima, A. Ohazama, A. 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, 2011.
DOI : 10.1016/j.ajpath.2011.04.027

URL : http://europepmc.org/articles/pmc3157255?pdf=render

J. Chen, Y. Song, Y. J. Gong, L. Zhao, P. Zhang 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.269-77, 2013.
DOI : 10.1074/jbc.M610172200

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.14413-14440, 2015.
DOI : 10.18632/oncotarget.3891

URL : https://hal.archives-ouvertes.fr/inserm-01793498

M. Pickup, S. Novitskiy, and H. Moses, The roles of TGF?? in the tumour microenvironment, Nature Reviews Cancer, vol.191, issue.11, pp.788-99300, 2013.
DOI : 10.1084/jem.191.7.1187

M. Pickup, P. Owens, and H. Moses, TGF-??, Bone Morphogenetic Protein, and Activin Signaling and the Tumor Microenvironment, Cold Spring Harbor Perspectives in Biology, vol.13, issue.5, 2017.
DOI : 10.1016/j.cellimm.2008.11.001

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

P. Carmeliet and R. Jain, Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases, Nature Reviews Drug Discovery, vol.7, issue.6, pp.417-444, 2011.
DOI : 10.1038/nmeth.1475

J. Franses, A. Baker, V. Chitalia, and E. Edelman, Stromal Endothelial Cells Directly Influence Cancer Progression, Science Translational Medicine, vol.335, issue.1, 2011.
DOI : 10.1007/s00441-008-0672-y

URL : http://europepmc.org/articles/pmc3076139?pdf=render

A. Chung and N. Ferrara, Developmental and Pathological Angiogenesis, Annual Review of Cell and Developmental Biology, vol.27, issue.1, pp.563-84, 2011.
DOI : 10.1146/annurev-cellbio-092910-154002

S. Tugues, S. Koch, L. Gualandi, X. Li, and L. Claesson-welsh, Vascular endothelial growth factors and receptors: Anti-angiogenic therapy in the treatment of cancer, Molecular Aspects of Medicine, vol.32, issue.2, pp.88-111, 2011.
DOI : 10.1016/j.mam.2011.04.004

C. Heldin, Targeting the PDGF signaling pathway in tumor treatment, Cell Communication and Signaling, vol.11, issue.1, pp.1478-811, 2013.
DOI : 10.1186/1478-811X-11-97

URL : https://biosignaling.biomedcentral.com/track/pdf/10.1186/1478-811X-11-97?site=biosignaling.biomedcentral.com

M. Dickson, J. Martin, F. Cousins, A. Kulkarni, S. Karlsson et al., Defective haematopoiesis and vasculogenesis in transforming growth factorbeta 1 knock out mice, Development, vol.121, pp.1845-54, 1995.

P. Bertolino, M. Deckers, F. Lebrin, and P. Ten-dijke, Transforming Growth Factor-?? Signal Transduction in Angiogenesis and Vascular Disorders, Chest, vol.128, issue.6, pp.585-90, 2005.
DOI : 10.1378/chest.128.6_suppl.585S

M. Oshima, H. Oshima, and M. Taketo, TGF-?? Receptor Type II Deficiency Results in Defects of Yolk Sac Hematopoiesis and Vasculogenesis, Developmental Biology, vol.179, issue.1, pp.297-302, 1996.
DOI : 10.1006/dbio.1996.0259

URL : https://doi.org/10.1006/dbio.1996.0259

S. Oh, T. Seki, K. Goss, T. Imamura, Y. Yi et al., Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis, Proceedings of the National Academy of Sciences, vol.196, issue.2, pp.2626-2657, 2000.
DOI : 10.1002/aja.1001960207

E. Pardali and P. Ten-dijke, Transforming growth factor-beta signaling and tumor angiogenesis, Frontiers in Bioscience, vol.14, issue.1, pp.4848-61, 2009.
DOI : 10.2741/3573

J. Tuxhorn, S. Mcalhany, F. Yang, T. Dang, and D. Rowley, Inhibition of transforming growth factor-beta activity decreases angiogenesis in a human prostate cancer-reactive stroma xenograft model, Cancer Res, vol.62, pp.6021-6026, 2002.

A. Mazzocca, E. Fransvea, G. Lavezzari, S. Antonaci, and G. Giannelli, Inhibition of transforming growth factor ?? receptor I kinase blocks hepatocellular carcinoma growth through neo-angiogenesis regulation, Hepatology, vol.69, issue.4, pp.1140-51, 2009.
DOI : 10.1016/j.cardiores.2005.07.007

URL : http://onlinelibrary.wiley.com/doi/10.1002/hep.23118/pdf

D. Donovan, J. Harmey, D. Toomey, D. Osborne, H. Redmond et al., TGF beta-1 regulation of VEGF production by breast cancer cells
DOI : 10.1007/bf02303745

, Ann Surg Oncol, vol.4, pp.621-628, 1997.

A. Fischer, E. Fuchs, M. Mikula, H. Huber, H. Beug et al., PDGF essentially links TGF-?? signaling to nuclear ??-catenin accumulation in hepatocellular carcinoma progression, Oncogene, vol.193, issue.23, pp.3395-405, 2007.
DOI : 10.1016/S0304-3835(03)00013-2

B. Xiong, L. Gong, F. Zhang, M. Hu, and H. Yuan, TGF beta1 expression and angiogenesis in colorectal cancer tissue, World J Gastroenterol, vol.8, 2002.

M. Petersen, E. Pardali, G. Van-der-horst, H. Cheung, C. Van-den-hoogen et al., Smad2 and Smad3 have opposing roles in breast cancer bone metastasis by differentially affecting tumor angiogenesis, Oncogene, vol.62, issue.9, pp.1351-61426, 2009.
DOI : 10.1172/JCI3523

J. Yang, D. Yang, Y. Sun, B. Sun, G. Wang et al., , in human osteosarcoma, Cancer, vol.50, issue.1 pt 1, pp.4925-4963, 2011.
DOI : 10.1002/pbc.21296

D. Chen, Y. Zhang, K. Zhu, and W. Wang, A systematic review of vascular endothelial growth factor expression as a biomarker of prognosis in patients with osteosarcoma, Tumor Biology, vol.39, issue.Suppl 7, pp.1895-1904, 2013.
DOI : 10.1007/s11033-012-1812-8

C. Betsholtz, B. Westermark, B. Ek, and C. Heldin, Coexpression of a PDGF-like growth factor and PDGF receptors in a human osteosarcoma cell line: Implications for autocrine receptor activation, Cell, vol.39, issue.3, pp.447-57, 1984.
DOI : 10.1016/0092-8674(84)90452-5

R. Verrecchia and . Signaling, Microenvironment in Osteosarcoma Frontiers in Oncology | www.frontiersin, p.133, 2018.

T. Kubo, S. Piperdi, J. Rosenblum, C. Antonescu, W. Chen et al., Platelet-derived growth factor receptor as a prognostic marker and a therapeutic target for imatinib mesylate therapy in osteosarcoma, Cancer, vol.24, issue.10, pp.2119-2148, 2008.
DOI : 10.1007/s10637-005-4543-z

M. Wu, G. Chen, and Y. 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

K. Janssens, P. Ten-dijke, S. Janssens, V. Hul, and W. , Transforming Growth Factor-??1 to the Bone, Endocrine Reviews, vol.26, issue.6, pp.743-74, 2005.
DOI : 10.1210/er.2004-0001

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.2254-72, 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.552-63, 2004.
DOI : 10.1038/sj.emboj.7600067

M. Karst, G. Gorny, R. Galvin, and M. 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 : 10.1038/345442a0

X. Guan, Cancer metastases: challenges and opportunities, Acta Pharmaceutica Sinica B, vol.5, issue.5, pp.402-420, 2015.
DOI : 10.1016/j.apsb.2015.07.005

K. Bjørnland, K. Flatmark, S. Pettersen, A. Aaasen, O. Fodstad et al., Matrix Metalloproteinases Participate in Osteosarcoma Invasion, Journal of Surgical Research, vol.127, issue.2, 2005.
DOI : 10.1016/j.jss.2004.12.016

O. Fromigué, Z. Hamidouche, and P. Marie, Blockade of the RhoA-JNK-c-Jun-MMP2 Cascade by Atorvastatin Reduces Osteosarcoma Cell Invasion, Journal of Biological Chemistry, vol.28, issue.45, pp.30549-56, 2008.
DOI : 10.1158/0008-5472.CAN-05-1149

M. Zhang and X. Zhang, Association of MMP-2 expression and prognosis in osteosarcoma patients, Int J Clin Exp Pathol, vol.8, pp.14965-70, 2015.

P. Kunz, H. Sähr, B. Lehner, C. Fischer, E. Seebach et al., Elevated ratio of MMP2/MMP9 activity is associated with poor response to chemotherapy in osteosarcoma, BMC Cancer, vol.97, issue.2, pp.223-233, 2016.
DOI : 10.1073/pnas.97.8.3884

J. Letterio and A. Roberts, REGULATION OF IMMUNE RESPONSES BY TGF-??, Annual Review of Immunology, vol.16, issue.1, pp.137-61, 1998.
DOI : 10.1146/annurev.immunol.16.1.137

M. Li and R. Flavell, TGF-??: A Master of All T Cell Trades, Cell, vol.134, issue.3, pp.392-404, 2008.
DOI : 10.1016/j.cell.2008.07.025

URL : https://doi.org/10.1016/j.cell.2008.07.025

R. Flavell, S. Sanjabi, S. Wrzesinski, and P. Licona-limón, The polarization of immune cells in the tumour environment by TGF??, Nature Reviews Immunology, vol.7, issue.8, pp.554-67, 2010.
DOI : 10.1172/JCI200215333

L. Gorelik and R. Flavell, Immune-mediated eradication of tumors through the blockade of transforming growth factor-?? signaling in T cells, Nature Medicine, vol.156, issue.10, pp.1118-1140, 2001.
DOI : 10.1084/jem.188.10.1849

E. Wilson, J. El-jawhari, A. Neilson, G. Hall, A. Melcher et al., Human tumor immune evasion via TGF-? blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumor activity, PLoS One, vol.6, issue.9, 2011.
DOI : 10.1371/journal.pone.0022842

URL : https://doi.org/10.1371/journal.pone.0022842

M. Papaspyridonos, I. Matei, Y. Huang, M. Do-rosario-andre, H. Brazier-mitouart et al., Id1 suppresses anti-tumour immune responses and promotes tumour progression by impairing myeloid cell maturation, Nature Communications, vol.20, issue.6, 2015.
DOI : 10.1016/j.semcancer.2012.02.003

URL : http://www.nature.com/articles/ncomms7840.pdf

C. Lewis and J. Pollard, Distinct Role of Macrophages in Different Tumor Microenvironments, Cancer Research, vol.66, issue.2, pp.605-617, 2006.
DOI : 10.1158/0008-5472.CAN-05-4005

URL : http://cancerres.aacrjournals.org/content/canres/66/2/605.full.pdf

B. Qian and J. Pollard, Macrophage Diversity Enhances Tumor Progression and Metastasis, Cell, vol.141, issue.1, pp.39-51, 2010.
DOI : 10.1016/j.cell.2010.03.014

URL : https://doi.org/10.1016/j.cell.2010.03.014

D. Gong, W. Shi, S. Yi, H. Chen, J. Groffen et al., TGF?? signaling plays a critical role in promoting alternative macrophage activation, BMC Immunology, vol.13, issue.1, pp.31-41, 2012.
DOI : 10.1002/gene.10046

URL : https://bmcimmunol.biomedcentral.com/track/pdf/10.1186/1471-2172-13-31?site=bmcimmunol.biomedcentral.com

M. Heymann, F. Lézot, and D. Heymann, The contribution of immune infiltrates and the local microenvironment in the pathogenesis of osteosarcoma, Cellular Immunology, 2017.
DOI : 10.1016/j.cellimm.2017.10.011

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

E. Buddingh, M. Kuijjer, R. Duim, H. Bürger, K. Agelopoulos et al., Tumor-Infiltrating Macrophages Are Associated with Metastasis Suppression in High-Grade Osteosarcoma: A Rationale for Treatment with Macrophage Activating Agents, Clinical Cancer Research, vol.17, issue.8, pp.2110-2119, 2011.
DOI : 10.1158/1078-0432.CCR-10-2047

URL : http://clincancerres.aacrjournals.org/content/clincanres/17/8/2110.full.pdf

C. Dumars, J. Ngyuen, A. Gaultier, R. Lanel, N. Corradini et al., Dysregulation of macrophage polarization is associated with the metastatic process in osteosarcoma, Oncotarget, vol.7, issue.48, pp.78343-54, 2016.
DOI : 10.18632/oncotarget.13055

URL : https://hal.archives-ouvertes.fr/inserm-01466103

A. Gomez-brouchet, C. Illac, J. Gilhodes, C. Bouvier, S. Aubert et al., CD163-positive tumor-associated macrophages and CD8-positive cytotoxic lymphocytes are powerful diagnostic markers for the therapeutic stratification of osteosarcoma patients: An immunohistochemical analysis of the biopsies fromthe French OS2006 phase 3 trial, OncoImmunology, vol.6, issue.9, 2017.
DOI : 10.1126/scitranslmed.3006504

B. Fritzsching, J. Fellenberg, L. Moskovszky, Z. Sápi, T. Krenacs et al., -ratio in osteosarcoma microenvironment separates survivors from non-survivors: a multicenter validated retrospective study, OncoImmunology, vol.31, issue.3, pp.990800-990810, 2014.
DOI : 10.1158/1078-0432.CCR-07-4534

URL : http://www.tandfonline.com/doi/pdf/10.4161/2162402X.2014.990800?needAccess=true

L. Katz, Y. Li, J. Chen, N. Muñoz, A. Majumdar et al., Targeting TGF-?? signaling in cancer, Expert Opinion on Therapeutic Targets, vol.7, issue.6, pp.743-60, 2013.
DOI : 10.1371/journal.pone.0039684

C. Neuzillet, A. Tijeras-raballand, R. Cohen, J. Cros, S. Faivre et al., Targeting the TGF?? pathway for cancer therapy, Pharmacology & Therapeutics, vol.147, pp.22-31, 2015.
DOI : 10.1016/j.pharmthera.2014.11.001

S. Colak, T. Dijke, and P. , Targeting TGF-?? Signaling in Cancer, Trends in Cancer, vol.3, issue.1, pp.56-71, 2017.
DOI : 10.1016/j.trecan.2016.11.008

U. Bogdahn, P. Hau, G. Stockhammer, N. Venkataramana, A. Mahapatra et al., Targeted therapy for high-grade glioma with the TGF-??2 inhibitor trabedersen: results of a randomized and controlled phase IIb study, Neuro-Oncology, vol.26, issue.13, pp.132-174, 2011.
DOI : 10.1200/JCO.2007.14.8163

J. Rodon, M. Carducci, J. Sepulveda-sánchez, A. Azaro, E. Calvo et al., First-in-Human Dose Study of the Novel Transforming Growth Factor-?? Receptor I Kinase Inhibitor LY2157299 Monohydrate in Patients with Advanced Cancer and Glioma, Clinical Cancer Research, vol.21, issue.3, pp.553-60, 2015.
DOI : 10.1158/1078-0432.CCR-14-1380