D. Dempster, Anatomy and functions of the adult skeleton In: Primer of the metabolic bone diseases and disorders of mineral metabolism, Ed. M.J. Favus, pp.7-11, 2006.

M. Favus, D. Bushinsky, L. Jr, and J. , Regulation of calcium, magnesium, and phosphate metabolism In: Primer of the metabolic bone diseases and disorders of mineral metabolism, Ed. M.J. Favus, pp.76-83, 2006.

F. Deschaseaux, L. Sensebe, and D. Heymann, Mechanisms of bone repair and regeneration, Trends in Molecular Medicine, vol.15, issue.9
DOI : 10.1016/j.molmed.2009.07.002

F. Ross, Osteoclast biology and bone resorption Primer of the metabolic bone diseases and disorders of mineral metabolism, pp.30-35, 2006.

A. Rousselle and D. Heymann, Osteoclastic acidification pathways during bone resorption, Bone, vol.30, issue.4, pp.533-573, 2002.
DOI : 10.1016/S8756-3282(02)00672-5

L. Filgueira, Osteoclast Differentiation and Function, Bone Cancer, pp.59-66, 2010.
DOI : 10.1016/B978-0-12-374895-9.00005-0

J. Billiard, Regulation of osteblast differentiation and bone cancers by Wnt and PTH signaling pathways, Bone Cancer, pp.47-58, 2010.

J. Aubin, J. Lian, and G. Stein, Bone formation maturation and functional activities of osteoblast lineage cells In: Primer of the metabolic bone diseases and disorders of mineral metabolism, pp.20-29, 2006.

N. Takahashi, OSTEOBLASTIC CELLS ARE INVOLVED IN OSTEOCLAST FORMATION, Endocrinology, vol.123, issue.5, pp.2600-2602, 1988.
DOI : 10.1210/endo-123-5-2600

E. Jimi, Osteoclast function is activated by osteoblastic cells through a mechanism involving cell-to-cell contact., Endocrinology, vol.137, issue.5, pp.2187-90, 1996.
DOI : 10.1210/endo.137.5.8612568

G. Mundy and F. Elefteriou, Boning up on Ephrin Signaling, Cell, vol.126, issue.3, pp.441-444, 2006.
DOI : 10.1016/j.cell.2006.07.015

E. Hauben and P. Hogendoorn, Epidemiology of primary bone tumors and economical aspects of bone metastases, Bone Cancer, pp.3-8, 2010.

Y. Wittrant, OPG: new therapeutic targets in bone tumours and associated osteolysis, Biochim Biophys Acta, vol.1704, pp.49-57, 2004.
URL : https://hal.archives-ouvertes.fr/inserm-00669006

K. Koeneman, F. Yeung, and L. Chung, Osteomimetic properties of prostate cancer cells: A hypothesis supporting the predilection of prostate cancer metastasis and growth in the bone environment, The Prostate, vol.93, issue.4, pp.246-61, 1999.
DOI : 10.1002/(SICI)1097-0045(19990601)39:4<246::AID-PROS5>3.0.CO;2-U

E. David, The Bone Niche of Chondrosarcoma: A Sanctuary for Drug Resistance, Tumour Growth and also a Source of New Therapeutic Targets, Sarcoma, vol.34, issue.4, p.932451, 2011.
DOI : 10.1002/ijc.21951

URL : https://hal.archives-ouvertes.fr/inserm-00667904

W. Simonet, Osteoprotegerin: A Novel Secreted Protein Involved in the Regulation of Bone Density, Cell, vol.89, issue.2, pp.309-328, 1997.
DOI : 10.1016/S0092-8674(00)80209-3

E. Tsuda, Identity of osteoclastogenesis inhibitory factor (OCIF) and osteoprotegerin (OPG): a mechanism by which OPG/OCIF inhibits osteoclastogenesis in vitro, Endocrinology, vol.139, pp.1329-1366, 1998.

D. Lacey, Osteoprotegerin Ligand Is a Cytokine that Regulates Osteoclast Differentiation and Activation, Cell, vol.93, issue.2, pp.165-76, 1998.
DOI : 10.1016/S0092-8674(00)81569-X

H. Yasuda, Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL, Proceedings of the National Academy of Sciences, vol.95, issue.7
DOI : 10.1073/pnas.95.7.3597

S. Theoleyre, Y. Wittrant, K. Tat, S. Fortun, Y. Redini et al., The molecular triad OPG/RANK/RANKL: involvement in the orchestration of pathophysiological bone remodeling, Cytokine & Growth Factor Reviews, vol.15, issue.6, pp.457-75, 2004.
DOI : 10.1016/j.cytogfr.2004.06.004

D. Heymann, Y. Fortun, F. Rédini, and M. Padrines, Osteolytic bone diseases: physiological analogues of bone resorption effectors as alternative therapeutic tools, Drug Discovery Today, vol.10, issue.4, pp.242-289, 2005.
DOI : 10.1016/S1359-6446(04)03265-9

K. Mori, K. Ando, D. Heymann, and F. Redini, Receptor activator of nuclear factor-kappa B ligand (RANKL) stimulates bone-associated tumours through functional RANK expressed on bone associated cancer cells?, Histol Histopathol, vol.24, pp.235-277, 2009.

M. Baud-'huin, F. Lamoureux, L. Duplomb, F. Rédini, and H. D. Rankl, osteoprotegerin: key partners of osteoimmunology and vascular diseases, Cell Mol Life Sci, vol.64, pp.2334-50, 2007.

H. Min, Osteoprotegerin Reverses Osteoporosis by Inhibiting Endosteal Osteoclasts and Prevents Vascular Calcification by Blocking a Process Resembling Osteoclastogenesis, The Journal of Experimental Medicine, vol.60, issue.4, pp.463-74, 2000.
DOI : 10.1038/386078a0

D. Anderson, A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function, Nature, vol.390, issue.6656, pp.175-184, 1997.
DOI : 10.1038/36593

W. Boyle, W. Simonet, and D. Lacey, Osteoclast differentiation and activation, Nature, vol.34, issue.6937, pp.337-379, 2003.
DOI : 10.1007/s007740200049

E. Grimaud, Receptor Activator of Nuclear Factor ??B Ligand (RANKL)/Osteoprotegerin (OPG) Ratio Is Increased in Severe Osteolysis, The American Journal of Pathology, vol.163, issue.5, pp.2021-2052, 2003.
DOI : 10.1016/S0002-9440(10)63560-2

K. Tat, S. Padrines, M. Theoleyre, S. Heymann, D. Fortun et al., IL-6, RANKL, TNF- alpha/IL-1: interrelations in bone resorption pathophysiology, Cytokine Growth Factor Rev, vol.15, pp.49-60, 2004.

C. Liu, Structural and Functional Insights of RANKL-RANK Interaction and Signaling, The Journal of Immunology, vol.184, issue.12, pp.6910-6919, 2010.
DOI : 10.4049/jimmunol.0904033

S. Ito and T. Hata, Crystal Structure of RANK Ligand Involved in Bone Metabolism, Vitam Horm, vol.67, pp.19-33, 2004.
DOI : 10.1016/S0083-6729(04)67002-6

J. Emery, Osteoprotegerin Is a Receptor for the Cytotoxic Ligand TRAIL, Journal of Biological Chemistry, vol.273, issue.23, pp.14363-14370, 1998.
DOI : 10.1074/jbc.273.23.14363

T. Standal, Osteoprotegerin is bound, internalized, and degraded by multiple myeloma cells, Blood, vol.100, issue.8, pp.3002-3009, 2002.
DOI : 10.1182/blood-2002-04-1190

S. Theoleyre, Cellular activity and signaling induced by osteoprotegerin in osteoclasts: involvement of receptor activator of nuclear factor ??B ligand and MAPK, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1644, issue.1
DOI : 10.1016/j.bbamcr.2003.10.005

F. Lamoureux, Glycosaminoglycans as Potential Regulators of Osteoprotegerin Therapeutic Activity in Osteosarcoma, Cancer Research, vol.69, issue.2, pp.526-562, 2009.
DOI : 10.1158/0008-5472.CAN-08-2648

A. Zannettino, Osteoprotegerin (OPG) is localized to the Weibel-Palade bodies of human vascular endothelial cells and is physically associated with von Willebrand factor, Journal of Cellular Physiology, vol.156, issue.2, pp.714-737, 2005.
DOI : 10.1002/jcp.20354

D. Heymann, Factor VIII/von Willebrand factor complex controls RANKL-induced osteoclastogenesis and cell survival, J Biol Chem, vol.264, pp.31704-31717, 2009.

G. Atkins, RANK Expression as a Cell Surface Marker of Human Osteoclast Precursors in Peripheral Blood, Bone Marrow, and Giant Cell Tumors of Bone, Journal of Bone and Mineral Research, vol.279, issue.Suppl 1, pp.1339-1388, 2006.
DOI : 10.1359/jbmr.060604

Y. Wittrant, K. Mori, A. Riet, A. Kamijo, D. Heymann et al., RANKL directly induces bone morphogenetic protein-2 expression in RANK-expressing POS-1 osteosarcoma cells, International Journal of Oncology, vol.28, pp.261-270, 2006.
DOI : 10.3892/ijo.28.1.261

URL : https://hal.archives-ouvertes.fr/inserm-00667510

K. Mori, Human osteosarcoma cells express functional receptor activator of nuclear factor-kappa B, The Journal of Pathology, vol.186, issue.5, pp.555-62, 2007.
DOI : 10.1002/path.2140

K. Mori, Receptor Activator of Nuclear Factor-kB Ligand (RANKL) directly modulates gene expression profile of RANK-positive Saos-2 human osteosarcoma cells

C. Hsu, Involvement of integrin up-regulation in RANKL/RANK pathway of chondrosarcomas migration, Journal of Cellular Biochemistry, vol.23, issue.1, pp.138-185, 2010.
DOI : 10.1002/jcb.22677

D. Santini, Receptor Activator of NF-kB (RANK) Expression in Primary Tumors Associates with Bone Metastasis Occurrence in Breast Cancer Patients, PLoS ONE, vol.34, issue.2, p.19234, 2011.
DOI : 10.1371/journal.pone.0019234.s002

D. Santini, Expression pattern of receptor activator of NF??B (RANK) in a series of primary solid tumors and related bone metastases, Journal of Cellular Physiology, vol.28, issue.3, pp.780-784, 2011.
DOI : 10.1002/jcp.22402

P. Bhatia, M. Sanders, and M. Hansen, Expression of receptor activator of nuclear factorkappaB is inversely correlated with metastatic phenotype in breast carcinoma, Clin Cancer Res, vol.11, pp.162-167, 2005.

D. Jones, Regulation of cancer cell migration and bone metastasis by RANKL, Nature, vol.17, issue.7084, pp.692-698, 2006.
DOI : 10.1038/nature04524

G. Chen, K. Sircar, A. Aprikian, A. Potti, D. Goltzman et al., Expression of RANKL/RANK/OPG in primary and metastatic human prostate cancer as markers of disease stage and functional regulation, Cancer, vol.18, issue.2, pp.289-98, 2006.
DOI : 10.1002/cncr.21978

K. Mori, L. Goff, B. Charrier, C. Battaglia, S. Heymann et al., DU145 human prostate cancer cells express functional receptor activator of NF??B: New insights in the prostate cancer bone metastasis process, Bone, vol.40, issue.4, pp.981-90, 2007.
DOI : 10.1016/j.bone.2006.11.006

A. Armstrong, R. Miller, J. Jones, J. Zhang, E. Keller et al., RANKL acts directly on RANK-expressing prostate tumor cells and mediates migration and expression of tumor metastasis genes, The Prostate, vol.32, issue.1, pp.92-104, 2008.
DOI : 10.1002/pros.20678

F. Chuang, S. Hsue, C. Wu, and Y. Chen, Immunohistochemical expression of RANKL, RANK, and OPG in human oral squamous cell carcinoma, Journal of Oral Pathology & Medicine, vol.34, issue.10, pp.753-761, 2009.
DOI : 10.1111/j.1600-0714.2009.00793.x

M. Shin, K. Matsuo, T. Tada, H. Fukushima, H. Furuta et al., The inhibition of RANKL/RANK signaling by osteoprotegerin suppresses bone invasion by oral squamous cell carcinoma cells, Carcinogenesis, vol.32, issue.11, pp.1634-1674, 2011.
DOI : 10.1093/carcin/bgr198

M. Heymann, A. Riet, L. Goff, B. Battaglia, S. Paineau et al., OPG, RANK and RANK ligand expression in thyroid lesions, Regulatory Peptides, vol.148, issue.1-3, pp.46-53, 2008.
DOI : 10.1016/j.regpep.2008.02.004

L. Chen, RANKL increases migration of human lung cancer cells through intercellular adhesion molecule-1 up-regulation, Journal of Cellular Biochemistry, vol.14, issue.3, pp.933-974, 2011.
DOI : 10.1002/jcb.23009

S. Mikami, Increased RANKL expression is related to tumour migration and metastasis of renal cell carcinomas, The Journal of Pathology, vol.107, issue.4, pp.530-539, 2009.
DOI : 10.1002/path.2567

V. Kupas, RANK Is Expressed in Metastatic Melanoma and Highly Upregulated on Melanoma-Initiating Cells, Journal of Investigative Dermatology, vol.131, issue.4, pp.944-55, 2011.
DOI : 10.1038/jid.2010.377

P. Fiumara, Functional expression of receptor activator of nuclear factor kappa B in Hodgkin disease cell lines, Blood, vol.98, issue.9, pp.2784-90, 2001.
DOI : 10.1182/blood.V98.9.2784

J. Lee, RANKL expression is related to treatment outcome of patients with localized, high-grade osteosarcoma, Pediatric Blood & Cancer, vol.116, issue.5, pp.738-781, 2011.
DOI : 10.1002/pbc.22720

N. Rucci, Receptor Activator of NF-??B Ligand Enhances Breast Cancer-Induced Osteolytic Lesions through Upregulation of Extracellular Matrix Metalloproteinase Inducer/CD147, Cancer Research, vol.70, issue.15, pp.6150-60, 2010.
DOI : 10.1158/0008-5472.CAN-09-2758

S. Cross, Expression of receptor activator of nuclear factor ???? ligand (RANKL) and tumour necrosis factor related, apoptosis inducing ligand (TRAIL) in breast cancer, and their relations with osteoprotegerin, oestrogen receptor, and clinicopathological variables, Journal of Clinical Pathology, vol.59, issue.7, pp.716-736, 2006.
DOI : 10.1136/jcp.2005.030031

C. Van-poznak, S. Cross, M. Saggese, C. Hudis, K. Panageas et al., Expression of osteoprotegerin (OPG), TNF related apoptosis inducing ligand (TRAIL), and receptor activator of nuclear factor ??B ligand (RANKL) in human breast tumours, Journal of Clinical Pathology, vol.59, issue.1, pp.56-63, 2006.
DOI : 10.1136/jcp.2005.026534

A. Sabbota, H. Kim, X. Zhe, R. Fridman, R. Bonfil et al., Shedding of RANKL by Tumor-Associated MT1-MMP Activates Src-Dependent Prostate Cancer Cell Migration, Cancer Research, vol.70, issue.13, pp.5558-66, 2010.
DOI : 10.1158/0008-5472.CAN-09-4416

V. Odero-marah, Receptor activator of NF-??B Ligand (RANKL) expression is associated with epithelial to mesenchymal transition in human prostate cancer cells, Cell Research, vol.122, issue.8, pp.858-70, 2008.
DOI : 10.1073/pnas.93.26.15152

S. Yuvaraj, A. Griffin, K. Sundaram, K. Kirkwood, J. Norris et al., A Novel Function of CXCL13 to Stimulate RANK Ligand Expression in Oral Squamous Cell Carcinoma Cells, Molecular Cancer Research, vol.7, issue.8
DOI : 10.1158/1541-7786.MCR-08-0589

H. Penno, O. Nilsson, H. Brändström, O. Winqvist, and O. Ljunggren, Expression of RANK???ligand in prostate cancer cell lines, Scandinavian Journal of Clinical and Laboratory Investigation, vol.39, issue.6, pp.151-156, 2009.
DOI : 10.1080/00365510802460466

S. Sood, S. Balasubramanian, S. Higham, M. Fernando, and B. Harrison, Osteoprotegerin (OPG) and Related Proteins (RANK, RANKL and TRAIL) in Thyroid Disease, World Journal of Surgery, vol.23, issue.9, pp.1984-92, 2011.
DOI : 10.1007/s00268-011-1185-5

E. Nakamura, RANKL-induced CCL22/macrophage-derived chemokine produced from osteoclasts potentially promotes the bone metastasis of lung cancer expressing its receptor CCR4, Clinical & Experimental Metastasis, vol.84, issue.1, pp.9-18, 2006.
DOI : 10.1007/s10585-006-9006-1

A. Sasaki, Receptor Activator of Nuclear Factor-??B Ligand (RANKL) Expression in Hepatocellular Carcinoma With Bone Metastasis, Annals of Surgical Oncology, vol.30, issue.3, pp.1191-1200, 2007.
DOI : 10.1245/s10434-006-9277-4

V. Barcala, RANKL expression in a case of follicular lymphoma, European Journal of Haematology, vol.98, issue.12, pp.417-426, 2003.
DOI : 10.1034/j.1600-0609.2003.00067.x

H. Shibata, Malignant B-Lymphoid Cells with Bone Lesions Express Receptor Activator of Nuclear Factor-??B Ligand and Vascular Endothelial Growth Factor to Enhance Osteoclastogenesis, Clinical Cancer Research, vol.11, issue.17, pp.6109-6124, 2005.
DOI : 10.1158/1078-0432.CCR-05-0181

S. Shu, C. Martin, N. Thudi, W. Dirksen, and T. Rosol, Osteolytic bone resorption in adult T-cell leukemia/lymphoma, Leukemia & Lymphoma, vol.140, issue.4, pp.702-716, 2010.
DOI : 10.1007/s00223-004-0161-6

O. Sezer, U. Heider, C. Jakob, J. Eucker, and K. Possinger, Human Bone Marrow Myeloma Cells Express RANKL, Journal of Clinical Oncology, vol.20, issue.1, pp.353-357, 2002.
DOI : 10.1200/JCO.2002.20.1.353

S. Roux, RANK (receptor activator of nuclear factor-kappaB) and RANKL expression in multiple myeloma, British Journal of Haematology, vol.74, issue.1, pp.86-92, 2002.
DOI : 10.1073/pnas.95.7.3597

A. Farrugia, Receptor activator of nuclear factor-kappaB ligand expression by human myeloma cells mediates osteoclast formation in vitro and correlates with bone destruction in vivo, Cancer Res, vol.63, pp.5438-5483, 2003.

U. Heider, Expression of receptor activator of NF-kappaB ligand (RANKL) mRNA in human multiple myeloma cells, J Cancer Res Clin Oncol, vol.130, pp.469-74, 2004.

C. Buckle, H. Neville-webbe, P. Croucher, and M. Lawson, Targeting RANK/RANKL in the Treatment of Solid Tumours and Myeloma, Current Pharmaceutical Design, vol.16, issue.11, pp.1272-83, 2010.
DOI : 10.2174/138161210791034021

D. Granchi, In vitro blockade of receptor activator of nuclear factor-?B ligand prevents osteoclastogenesis induced by neuroblastoma cells, International Journal of Cancer, vol.33, issue.6, pp.829-867, 2004.
DOI : 10.1002/ijc.20308

Z. Tang, F. Zhang, P. Tang, X. Qi, and J. Jiang, Hypoxia induces RANK and RANKL expression by activating HIF-1?? in breast cancer cells, Biochemical and Biophysical Research Communications, vol.408, issue.3, pp.411-417, 2011.
DOI : 10.1016/j.bbrc.2011.04.035

L. Zhang, Receptor activator for nuclear factor ?? B expression predicts poor prognosis in breast cancer patients with bone metastasis but not in patients with visceral metastasis, Journal of Clinical Pathology, vol.65, issue.1, pp.36-40, 2012.
DOI : 10.1136/jclinpath-2011-200312

W. Dougall, Molecular Pathways: Osteoclast-Dependent and Osteoclast-Independent Roles of the RANKL/RANK/OPG Pathway in Tumorigenesis and Metastasis, Clinical Cancer Research, vol.18, issue.2, pp.326-335, 2012.
DOI : 10.1158/1078-0432.CCR-10-2507

J. Fata, The Osteoclast Differentiation Factor Osteoprotegerin-Ligand Is Essential for Mammary Gland Development, Cell, vol.103, issue.1, pp.41-50, 2000.
DOI : 10.1016/S0092-8674(00)00103-3

A. Mukherjee, Targeting RANKL to a specific subset of murine mammary epithelial cells induces ordered branching morphogenesis and alveologenesis in the absence of progesterone receptor expression, The FASEB Journal, vol.24, issue.11, pp.4408-4427, 2010.
DOI : 10.1096/fj.10-157982

R. Fernandez-valdivia, The RANKL signaling axis is sufficient to elicit ductal side-branching and alveologenesis in the mammary gland of the virgin mouse, Developmental Biology, vol.328, issue.1, pp.127-166, 2009.
DOI : 10.1016/j.ydbio.2009.01.019

E. Gonzalez-suarez, RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis, Nature, vol.96, issue.7320, pp.103-140, 2010.
DOI : 10.1038/nature09495

P. Joshi, Progesterone induces adult mammary stem cell expansion, Nature, vol.314, issue.7299, pp.803-810, 2010.
DOI : 10.1038/nature09091

M. Asselin-labat, Control of mammary stem cell function by steroid hormone signalling, Nature, vol.21, issue.7299, pp.798-802, 2010.
DOI : 10.1038/nature09027

F. Lamoureux, Therapeutic Relevance of Osteoprotegerin Gene Therapy in Osteosarcoma: Blockade of the Vicious Cycle between Tumor Cell Proliferation and Bone Resorption, Cancer Research, vol.67, issue.15, pp.7308-7326, 2007.
DOI : 10.1158/0008-5472.CAN-06-4130

F. Lamoureux, Therapeutic efficacy of soluble receptor activator of NF-kB (RANK) delivered by non viral gene transfer in a mouse model of osteolytic osteosarcoma, Bone, vol.42, pp.3389-98, 2008.
DOI : 10.1016/j.bone.2007.12.193

J. Rousseau, Formulated siRNAs targeting Rankl prevent osteolysis and enhance chemotherapeutic response in osteosarcoma models, Journal of Bone and Mineral Research, vol.390, issue.10, pp.2452-62, 2011.
DOI : 10.1002/jbmr.455

P. Bekker, The Effect of a Single Dose of Osteoprotegerin in Postmenopausal Women, Journal of Bone and Mineral Research, vol.16, issue.2, pp.348-60, 2001.
DOI : 10.1359/jbmr.2001.16.2.348

J. Body, A Phase I study of AMGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases, Cancer, vol.14, issue.S3, pp.887-892, 2003.
DOI : 10.1002/cncr.11138

P. Bekker, A Single-Dose Placebo-Controlled Study of AMG 162, a Fully Human Monoclonal Antibody to RANKL, in Postmenopausal Women, Journal of Bone and Mineral Research, vol.273, issue.7, pp.1059-66, 2004.
DOI : 10.1359/JBMR.040305

M. Mcclung, Denosumab in Postmenopausal Women with Low Bone Mineral Density, New England Journal of Medicine, vol.354, issue.8, pp.821-852, 2006.
DOI : 10.1056/NEJMoa044459

P. Kostenuik, Denosumab, a Fully Human Monoclonal Antibody to RANKL, Inhibits Bone Resorption and Increases BMD in Knock-In Mice That Express Chimeric (Murine/Human) RANKL*, Journal of Bone and Mineral Research, vol.24, issue.2
DOI : 10.1359/jbmr.081112

J. Body, A Study of the Biological Receptor Activator of Nuclear Factor-??B Ligand Inhibitor, Denosumab, in Patients with Multiple Myeloma or Bone Metastases from Breast Cancer, Clinical Cancer Research, vol.12, issue.4, pp.1221-1229, 2006.
DOI : 10.1158/1078-0432.CCR-05-1933

D. Henry, Randomized, Double-Blind Study of Denosumab Versus Zoledronic Acid in the Treatment of Bone Metastases in Patients With Advanced Cancer (Excluding Breast and Prostate Cancer) or Multiple Myeloma, Journal of Clinical Oncology, vol.29, issue.9, pp.1125-1157, 2011.
DOI : 10.1200/JCO.2010.31.3304

A. Stopeck, Denosumab Compared With Zoledronic Acid for the Treatment of Bone Metastases in Patients With Advanced Breast Cancer: A Randomized, Double-Blind Study, Journal of Clinical Oncology, vol.28, issue.35, pp.5132-5171, 2010.
DOI : 10.1200/JCO.2010.29.7101

J. Body, Effects of denosumab in patients with bone metastases with and without previous bisphosphonate exposure, Journal of Bone and Mineral Research, vol.357, issue.3, pp.440-446, 2010.
DOI : 10.1359/jbmr.090810

A. Lipton, Extended Efficacy and Safety of Denosumab in Breast Cancer Patients with Bone Metastases Not Receiving Prior Bisphosphonate Therapy, Clinical Cancer Research, vol.14, issue.20, pp.6690-6696, 2008.
DOI : 10.1158/1078-0432.CCR-07-5234

A. Lipton, Randomized active-controlled phase II study of denosumad efficacy and safety in patients with breast cancer-related bone metatases, J Clin Oncol, vol.45, pp.4431-4438, 2007.

G. Ellis, Randomized Trial of Denosumab in Patients Receiving Adjuvant Aromatase Inhibitors for Nonmetastatic Breast Cancer, Journal of Clinical Oncology, vol.26, issue.30, pp.4875-82, 2008.
DOI : 10.1200/JCO.2008.16.3832

G. Ellis, Effect of denosumab on bone mineral density in women receiving adjuvant aromatase inhibitors for non-metastatic breast cancer: subgroup analyses of a phase 3 study, Breast Cancer Research and Treatment, vol.106, issue.suppl 1, pp.81-88, 2009.
DOI : 10.1007/s10549-009-0352-y

K. Fizazi, Randomized Phase II Trial of Denosumab in Patients With Bone Metastases From Prostate Cancer, Breast Cancer, or Other Neoplasms After Intravenous Bisphosphonates, Journal of Clinical Oncology, vol.27, issue.10, pp.1564-71, 2009.
DOI : 10.1200/JCO.2008.19.2146

R. Vij, An open-label, phase 2 trial of denosumab in the treatment of relapsed or plateau-phase multiple myeloma, American Journal of Hematology, vol.37, issue.10, pp.650-656, 2009.
DOI : 10.1002/ajh.21509

K. Fizazi, Denosumab Treatment of Prostate Cancer With Bone Metastases and Increased Urine N-Telopeptide Levels After Therapy With Intravenous Bisphosphonates: Results of a Randomized Phase II Trial, The Journal of Urology, vol.182, issue.2, pp.509-524, 2009.
DOI : 10.1016/j.juro.2009.04.023

M. Smith, Denosumab in Men Receiving Androgen-Deprivation Therapy for Prostate Cancer, New England Journal of Medicine, vol.361, issue.8, pp.745-55, 2009.
DOI : 10.1056/NEJMoa0809003

M. Smith, Effects of Denosumab on Bone Mineral Density in Men Receiving Androgen Deprivation Therapy for Prostate Cancer, The Journal of Urology, vol.182, issue.6, pp.2670-2675, 2009.
DOI : 10.1016/j.juro.2009.08.048

M. Smith, Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial, The Lancet, vol.379, issue.9810, pp.39-46, 2012.
DOI : 10.1016/S0140-6736(11)61226-9

K. Fizazi, Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study, The Lancet, vol.377, issue.9768, pp.813-835, 2011.
DOI : 10.1016/S0140-6736(10)62344-6

D. Thomas, Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study, The Lancet Oncology, vol.11, issue.3, pp.275-80, 2010.
DOI : 10.1016/S1470-2045(10)70010-3

S. Jamal, Effects of denosumab on fracture and bone mineral density by level of kidney function, Journal of Bone and Mineral Research, vol.76, issue.8, pp.1839-1874, 2011.
DOI : 10.1002/jbmr.403

R. Coleman, Zoledronic acid, Expert Opinion on Drug Safety, vol.188, issue.1, pp.133-178, 2011.
DOI : 10.1056/NEJMoa0806285

URL : https://hal.archives-ouvertes.fr/hal-00621619

C. Migliorati, J. Epstein, E. Abt, and J. Berenson, Osteonecrosis of the jaw and bisphosphonates in cancer: a narrative review, Nature Reviews Endocrinology, vol.68, issue.1, pp.34-42, 2011.
DOI : 10.1038/nrendo.2010.195

O. Filleul, E. Crompot, and S. Saussez, Bisphosphonate-induced osteonecrosis of the jaw: a review of 2,400 patient cases, Journal of Cancer Research and Clinical Oncology, vol.144, issue.8, pp.1117-1141, 2010.
DOI : 10.1007/s00432-010-0907-7

D. Heymann, Editorial [Hot topic: Bisphosphonates and Bone Diseases: Past, Present and Future (Guest Editor: Dominique HEYMANN)], Current Pharmaceutical Design, vol.16, issue.27, pp.2948-2957, 2010.
DOI : 10.2174/138161210793563572

J. Xie, Economic Evaluation of Denosumab Compared with Zoledronic Acid in Hormone-Refractory Prostate Cancer Patients with Bone Metastases, Journal of Managed Care Pharmacy, vol.17, issue.8, pp.621-664, 2011.
DOI : 10.18553/jmcp.2011.17.8.621

L. Van-de-wetering-de-rooij, Safety, pharmacokinetics and efficacy of anti- RANKL nanobody ALX-0141 in healthy postmenopausal wome, Ann Rheum Dis, vol.70, issue.3, p.136, 2011.

D. Heymann, B. Ory, F. Gouin, J. Green, and F. Rédini, Bisphosphonates: new therapeutic agents for the treatment of bone tumors, Trends in Molecular Medicine, vol.10, issue.7, pp.337-343, 2004.
DOI : 10.1016/j.molmed.2004.05.007

B. Ory, G. Moriceau, F. Rédini, and D. Heymann, mTOR Inhibitors (Rapamycin and its Derivatives) and Nitrogen Containing Bisphosphonates: Bi-Functional Compounds for the Treatment of Bone Tumours, Current Medicinal Chemistry, vol.14, issue.13, pp.1381-1387, 2007.
DOI : 10.2174/092986707780831159

R. Coleman and E. Mccloskey, Bisphosphonates in oncology, Bone, vol.49, issue.1, pp.71-77, 2011.
DOI : 10.1016/j.bone.2011.02.003

R. Coleman, The use of bisphosphonates in cancer treatment, Annals of the New York Academy of Sciences, vol.25, issue.3, pp.3-14, 2011.
DOI : 10.1111/j.1749-6632.2010.05766.x

B. Ory, Zoledronic acid suppresses lung metastases and prolongs overall survival of osteosarcoma-bearing mice, Cancer, vol.91, issue.11, pp.2522-2529, 2005.
DOI : 10.1002/cncr.21530

F. Gouin, F. Rédini, B. Ory, and D. Heymann, Zoledronic acid slows down rat primary chondrosarcoma development, recurrent tumor progression after intralesional curretage and increases overall survival, International Journal of Cancer, vol.43, issue.5, pp.980-984, 2006.
DOI : 10.1002/ijc.21951

B. Ory, F. Blanchard, S. Battaglia, F. Gouin, F. Rédini et al., Zoledronic Acid Activates the DNA S-Phase Checkpoint and Induces Osteosarcoma Cell Death Characterized by Apoptosis-Inducing Factor and Endonuclease-G Translocation Independently of p53 and Retinoblastoma Status, Molecular Pharmacology, vol.71, issue.1, pp.333-343, 2007.
DOI : 10.1124/mol.106.028837

I. Holen and R. Coleman, Bisphosphonates as Treatment of Bone Metastases, Current Pharmaceutical Design, vol.16, issue.11, pp.1262-71, 2010.
DOI : 10.2174/138161210791034003

F. Lamoureux, Relevance of a new rat model of osteoblastic metastases from prostate carcinoma for preclinical studies using zoledronic acid, International Journal of Cancer, vol.104, issue.4, pp.751-760, 2008.
DOI : 10.1002/ijc.23187

G. Moriceau, Therapeutic approach of primary bone tumors by bisphosphonates

B. Ory, G. Moriceau, F. Redini, and D. Heymann, mTOR Inhibitors (Rapamycin and its Derivatives) and Nitrogen Containing Bisphosphonates: Bi-Functional Compounds for the Treatment of Bone Tumours, Current Medicinal Chemistry, vol.14, issue.13, pp.1381-1388, 2007.
DOI : 10.2174/092986707780831159

D. Heymann, Enhanced tumor regression and tissue repair when zoledronic acid is combined with ifosfamide in rat osteosarcoma, Bone, vol.37, issue.1, pp.74-86, 2005.
DOI : 10.1016/j.bone.2005.02.020

G. Moriceau, Zoledronic Acid Potentiates mTOR Inhibition and Abolishes the Resistance of Osteosarcoma Cells to RAD001 (Everolimus): Pivotal Role of the Prenylation Process, Cancer Research, vol.70, issue.24, pp.10329-10368, 2010.
DOI : 10.1158/0008-5472.CAN-10-0578

URL : https://hal.archives-ouvertes.fr/inserm-00667504

S. Battaglia, Impact of oncopediatric dosing regimen of zoledronic acid on bone growth: Preclinical studies and case report of an osteosarcoma pediatric patient, Journal of Bone and Mineral Research, vol.397, issue.10, pp.2439-51, 2011.
DOI : 10.1002/jbmr.453

F. Daubiné, L. Gall, C. Gasser, J. Green, J. Clézardin et al., Antitumor Effects of Clinical Dosing Regimens of Bisphosphonates in Experimental Breast Cancer Bone Metastasis, JNCI Journal of the National Cancer Institute, vol.99, issue.4, pp.322-352, 2007.
DOI : 10.1093/jnci/djk054

P. Ottewell, H. Mönkkönen, M. Jones, D. Lefley, R. Coleman et al., Antitumor Effects of Doxorubicin Followed by Zoledronic Acid in a Mouse Model of Breast Cancer, JNCI Journal of the National Cancer Institute, vol.100, issue.16, pp.1167-78, 2008.
DOI : 10.1093/jnci/djn240

D. Heymann, Y. Fortun, F. Redini, and M. Padrines, Osteolytic bone diseases: physiological analogues of bone resorption effectors as alternative therapeutic tools, Drug Discovery Today, vol.10, issue.4, pp.242-249, 2005.
DOI : 10.1016/S1359-6446(04)03265-9

D. Heymann, . P. Book-ed, . Picci, and . Ruggieri, Novel targeted therapies of bone tumors and future directions Bone Metastasis management, Future Medicine

V. Engleman, A peptidomimetic antagonist of the alpha(v)beta3 integrin inhibits bone resorption in vitro and prevents osteoporosis in vivo., Journal of Clinical Investigation, vol.99, issue.9, pp.2284-92, 1997.
DOI : 10.1172/JCI119404

X. Cheng, Disabling of Receptor Activator of Nuclear Factor-??B (RANK) Receptor Complex by Novel Osteoprotegerin-like Peptidomimetics Restores Bone Loss in Vivo, Journal of Biological Chemistry, vol.279, issue.9, pp.8269-77, 2004.
DOI : 10.1074/jbc.M309690200

K. Aoki, A TNF receptor loop peptide mimic blocks RANK ligand???induced signaling, bone resorption, and bone loss, Journal of Clinical Investigation, vol.116, issue.6, pp.1525-1559, 2006.
DOI : 10.1172/JCI22513

D. Heath, An Osteoprotegerin-like Peptidomimetic Inhibits Osteoclastic Bone Resorption and Osteolytic Bone Disease in Myeloma, Cancer Research, vol.67, issue.1, pp.202-210, 2007.
DOI : 10.1158/0008-5472.CAN-06-1287

L. Penolazzi, Peptide nucleic acid-DNA decoy chimeras targeting NF-kappa B transcription factors: induction of apoptosis in human primary osteoclasts, Int J Mol Med, vol.14, pp.145-52, 2004.

L. Penolazzi, Decoy oligodeoxynucleotides targeting NF-kappaB transcription factors: induction of apoptosis in human primary osteoclasts, Biochemical Pharmacology, vol.66, issue.7, pp.1189-98, 2003.
DOI : 10.1016/S0006-2952(03)00470-2

J. Clohisy, NF-kB signaling blockade abolishes implant particle-induced osteoclastogenesis, Journal of Orthopaedic Research, vol.276, issue.1, pp.13-20, 2004.
DOI : 10.1016/S0736-0266(03)00156-6

J. Clohisy, Direct Inhibition of NF-??B Blocks Bone Erosion Associated with Inflammatory Arthritis, The Journal of Immunology, vol.171, issue.10, pp.5547-53, 2003.
DOI : 10.4049/jimmunol.171.10.5547

C. Carron, Peptidomimetic antagonists of alphavbeta3 inhibit bone resorption by inhibiting osteoclast bone resorptive activity, not osteoclast adhesion to bone, Journal of Endocrinology, vol.165, issue.3, pp.587-98, 2000.
DOI : 10.1677/joe.0.1650587

S. Bakewell, Platelet and osteoclast ??3 integrins are critical for bone metastasis, Proceedings of the National Academy of Sciences, vol.100, issue.24, pp.14205-14215, 2003.
DOI : 10.1073/pnas.2234372100

J. Harms, integrin suppresses MDA-MB-435 skeletal metastasis, Clinical & Experimental Metastasis, vol.21, issue.2, pp.119-147, 2004.
DOI : 10.1023/B:CLIN.0000024763.69809.64

G. Van-der-horst, Targeting of ??v-Integrins in Stem/Progenitor Cells and Supportive Microenvironment Impairs Bone Metastasis in Human Prostate Cancer, Neoplasia, vol.13, issue.6, pp.516-541, 2011.
DOI : 10.1593/neo.11122

Y. Zhao, Tumor ??v??3 Integrin Is a Therapeutic Target for Breast Cancer Bone Metastases, Cancer Research, vol.67, issue.12, pp.5821-5851, 2007.
DOI : 10.1158/0008-5472.CAN-06-4499

J. Harms, integrin suppresses MDA-MB-435 skeletal metastasis, Clinical & Experimental Metastasis, vol.21, issue.2, pp.119-147, 2004.
DOI : 10.1023/B:CLIN.0000024763.69809.64

O. Ishibashi, S. Niwa, K. Kadoyama, and T. Inui, MMP-9 antisense oligodeoxynucleotide exerts an inhibitory effect on osteoclastic bone resorption by suppressing cell migration, Life Sciences, vol.79, issue.17, pp.1657-60, 2006.
DOI : 10.1016/j.lfs.2006.05.024

A. Costa, N. Cusano, B. Silva, S. Cremers, and J. Bilezikian, Cathepsin K: its skeletal actions and role as a therapeutic target in osteoporosis, Nature Reviews Rheumatology, vol.10, issue.8, pp.447-56, 2011.
DOI : 10.1038/nrrheum.2011.77

J. Sturge, M. Caley, and J. Waxman, Bone metastasis in prostate cancer: emerging therapeutic strategies, Nat Rev Clin Oncol, vol.8, pp.357-68, 2011.

Y. Ochi, Effects of ONO-5334, a novel orally-active inhibitor of cathepsin K, on bone metabolism, Bone, vol.49, issue.6, pp.1351-1357, 2011.
DOI : 10.1016/j.bone.2011.09.041

L. Gall and C. , A Cathepsin K Inhibitor Reduces Breast Cancer Induced Osteolysis and Skeletal Tumor Burden, Cancer Research, vol.67, issue.20, pp.9894-902, 2007.
DOI : 10.1158/0008-5472.CAN-06-3940

A. Xiang, Changes in micro-CT 3D bone parameters reflect effects of a potent cathepsin K inhibitor (SB-553484) on bone resorption and cortical bone formation in ovariectomized mice, Bone, vol.40, issue.5, pp.1231-1238, 2007.
DOI : 10.1016/j.bone.2007.01.010

A. Jensen, The Cathepsin K Inhibitor Odanacatib Suppresses Bone Resorption in Women With Breast Cancer and Established Bone Metastases: Results of a 4-Week, Double-Blind, Randomized, Controlled Trial, Clinical Breast Cancer, vol.10, issue.6, pp.452-460, 2010.
DOI : 10.3816/CBC.2010.n.059

K. Weilbaecher, T. Guise, M. Cauley, and L. , Cancer to bone: a fatal attraction, Nature Reviews Cancer, vol.71, issue.6, pp.411-425, 2011.
DOI : 10.1038/nrc3055