M. Attal, V. Lauwers-cances, C. Hulin, X. Leleu, D. Caillot et al.,

B. Lenalidomide, Dexamethasone with Transplantation for Myeloma, N Engl J Med. 6 avr, vol.376, issue.14, pp.1311-1331, 2017.

R. M. Kyle and . Myeloma, The New England Journal of Medicine, vol.14, 2004.

P. Antonio and A. Kenneth, Multiple Myeloma. n engl j med, vol.15, 2011.

S. Y. Kristinsson, O. Landgren, P. W. Dickman, Å. R. Derolf, and M. Björkholm, Patterns of Survival in Multiple Myeloma: A Population-Based Study of Patients Diagnosed in Sweden From, Journal of Clinical Oncology. 20 mai, vol.25, issue.15, pp.1993-2002, 1973.

N. Howlader, A. Noone, M. Krapcho, D. Miller, A. Brest et al., Cancer Statistics Review, 1975.

A. Monnereau, L. Remontet, M. Maynadié, F. Binder-foucard, A. Belot et al., Estimation nationale de l'incidence et de la mortalité par cancer en France entre 1980 et 2012: étude à partir des registres des cancers du réseau Francim, 2013.

P. R. Greipp, J. S. Miguel, B. Durie, J. J. Crowley, B. Barlogie et al., International Staging System for Multiple Myeloma, Journal of Clinical Oncology. 20 mai, vol.23, issue.15, pp.3412-3432, 2005.

S. V. Rajkumar, M. A. Dimopoulos, A. Palumbo, J. Blade, G. Merlini et al., International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. The Lancet Oncology, vol.15, pp.538-586, 2014.

E. Campo, S. H. Swerdlow, N. L. Harris, S. Pileri, H. Stein et al., The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications, Blood. 12 mai, vol.117, pp.5019-5051, 2011.

, Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: a report of the International Myeloma Working Group, The International Myeloma Working Group, vol.121, pp.749-57, 2003.

A. Palumbo, H. Avet-loiseau, S. Oliva, H. M. Lokhorst, H. Goldschmidt et al., Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group, JCO. 10 sept, vol.33, issue.26, pp.2863-2872, 2015.

M. Chretien, J. Corre, V. Lauwers-cances, F. Magrangeas, A. Cleynen et al., Understanding the role of hyperdiploidy in myeloma prognosis: which trisomies really matter?, Blood. 17 déc, vol.126, issue.25, pp.2713-2722, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01813432

B. Hebraud, X. Leleu, V. Lauwers-cances, M. Roussel, D. Caillot et al., Deletion of the 1p32 region is a major independent prognostic factor in young patients with myeloma: the IFM experience on 1195 patients, Leukemia. mars, vol.28, issue.3, pp.675-684, 2014.

H. Avet-loiseau, M. Attal, L. Campion, D. Caillot, C. Hulin et al., Play a Major Role in Defining Long-Term Survival, Term Analysis of the IFM 99 Trials for Myeloma: Cytogenetic Abnormalities, vol.30, pp.1949-52, 2012.

S. V. Rajkumar, M. A. Dimopoulos, A. Palumbo, J. Blade, G. Merlini et al., International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. The Lancet Oncology, vol.15, pp.538-586, 2014.

R. A. Kyle, T. M. Therneau, S. V. Rajkumar, J. R. Offord, D. R. Larson et al., A Long-Term Study of Prognosis in Monoclonal Gammopathy of Undetermined Significance, New England Journal of Medicine. 21 févr, vol.346, issue.8, pp.564-573, 2002.

O. Landgren and B. M. Weiss, Patterns of monoclonal gammopathy of undetermined significance and multiple myeloma in various ethnic/racial groups: support for genetic factors in pathogenesis, Leukemia, vol.23, issue.10, pp.1691-1698, 2009.

B. M. Weiss, J. Abadie, P. Verma, R. S. Howard, and W. M. Kuehl, A monoclonal gammopathy precedes multiple myeloma in most patients, Blood. 28 mai, vol.113, issue.22, pp.5418-5440, 2009.

O. Landgren, B. I. Graubard, J. A. Katzmann, R. A. Kyle, I. Ahmadizadeh et al., Racial disparities in the prevalence of monoclonal gammopathies: a population-based study of 12 482 persons from the National Health and Nutritional Examination Survey, Leukemia. juill, vol.28, issue.7, pp.1537-1579, 2014.

A. Smith, F. Wisloff, D. Samson, and . Forum, Nordic Myeloma Study Group and British Committee for Standards in Haematology. Guidelines on the diagnosis and management of multiple myeloma 2005, British Journal of Haematology. févr, vol.132, issue.4, pp.410-51, 2006.

R. A. Kyle and S. V. Rajkumar, Criteria for diagnosis, staging, risk stratification and response assessment of multiple myeloma, Leukemia. janv, vol.23, issue.1, pp.3-9, 2009.

A. Dispenzieri, J. A. Katzmann, R. A. Kyle, D. R. Larson, L. J. Melton et al., Prevalence and risk of progression of light-chain monoclonal gammopathy of undetermined significance: a retrospective population-based cohort study, The Lancet. mai, vol.375, issue.9727, pp.1721-1729, 2010.

T. Jelinek, F. Kryukov, L. Rihova, and R. Hajek, Plasma cell leukemia: from biology to treatment, European Journal of Haematology. juill, vol.95, issue.1, pp.16-26, 2015.

R. Mina, D. 'agostino, M. Cerrato, C. Gay, F. Palumbo et al., Plasma cell leukemia: update on biology and therapy, Leukemia & Lymphoma. 3 juill, vol.58, issue.7, pp.1538-1585, 2017.

A. Dispenzieri and . Syndrome, Update on diagnosis, risk stratification, and management: DISPENZIERI, American Journal of Hematology. août, vol.92, issue.8, pp.814-843, 2017.

B. Paiva, M. Chandia, M. Vidriales, E. Colado, T. Caballero-velazquez et al., Multiparameter flow cytometry for staging of solitary bone plasmacytoma: new criteria for risk of progression to myeloma, Blood. 21 août, vol.124, issue.8, pp.1300-1303, 2014.

M. Sant, P. Minicozzi, M. Mounier, L. A. Anderson, H. Brenner et al., Survival for haematological malignancies in Europe between 1997 and 2008 by region and age: results of EUROCARE-5, a population-based study, The Lancet Oncology. août, vol.15, issue.9, pp.931-973, 2014.

R. A. Kyle, S. V. Rajkumar, O. Landgren, J. Blade, G. Merlini et al., Monoclonal gammopathy of undetermined significance (MGUS) and smoldering (asymptomatic) multiple myeloma: IMWG consensus perspectives risk factors for progression and guidelines for monitoring and management, Leukemia. juin, vol.24, issue.6, pp.1121-1128, 2010.

A. Wood, R. Alexanian, and M. Dimopoulos, The Treatment of Multiple Myeloma, New England Journal of Medicine. 17 févr, vol.330, issue.7, pp.484-493, 1994.

W. Pönisch, P. S. Mitrou, K. Merkle, M. Herold, M. Assmann et al., Treatment of Bendamustine and Prednisone in patients with newly diagnosed multiple myeloma results in superior complete response rate, prolonged time to treatment failure and improved quality of life compared to treatment with Melphalan and Prednisone-a randomized phase III study of the East German Study Group of Hematology and Oncology (OSHO), Journal of Cancer Research and Clinical Oncology. avr, vol.132, issue.4, pp.205-217, 2006.

R. Alexanian, M. A. Dimopoulos, K. Delasalle, and B. Barlogie, Primary Dexamethasone Treatment of Multiple Myeloma, vol.5, 1992.

M. Attal, J. Harousseau, A. Stoppa, J. Sotto, J. Fuzibet et al., Randomized Trial of Autologous Bone Marrow Transplantation and Chemotherapy in Multiple Myeloma, N Engl J Med. 11 juill, vol.335, issue.2, pp.91-98, 1996.

V. Kotla, S. Goel, S. Nischal, C. Heuck, K. Vivek et al., Mechanism of action of lenalidomide in hematological malignancies, J Hematol Oncol, vol.2, issue.1, p.36, 2009.

G. Köhler and C. Milstein, Continuous cultures of fused cells secreting antibody of predefined specificity, Nature. août, vol.256, issue.5517, pp.495-502, 1975.

A. Palumbo, A. Chanan-khan, K. Weisel, A. K. Nooka, T. Masszi et al., Disponible sur, 2019.

B. Daratumumab, D. For-multiple, and . Myeloma, New England Journal of Medicine. 25 août, vol.375, issue.8, pp.754-66, 2016.

M. A. Dimopoulos, A. Oriol, H. Nahi, J. San-miguel, N. J. Bahlis et al.,

L. Daratumumab, D. For-multiple, and . Myeloma, New England Journal of Medicine, vol.375, issue.14, pp.1319-1350, 2016.

P. G. Richardson, M. Attal, F. Campana, S. Le-guennec, A. Hui et al., Isatuximab plus pomalidomide/dexamethasone versus pomalidomide/dexamethasone in relapsed/refractory multiple myeloma: ICARIA Phase III study design, Future Oncology. mai, vol.14, issue.11, pp.1035-1082, 2018.

A. Jakubowiak, M. Offidani, B. Pegourie, J. De-la-rubia, L. Garderet et al., Randomized phase 2 study: elotuzumab plus bortezomib/dexamethasone vs bortezomib/dexamethasone for relapsed/refractory MM, Blood. 9 juin, vol.127, issue.23, pp.2833-2873, 2016.

M. Sadelain, R. Brentjens, and I. Rivière, The Basic Principles of Chimeric Antigen Receptor Design, Cancer Discovery. avr, vol.3, issue.4, pp.388-98, 2013.

J. N. Kochenderfer and S. A. Rosenberg, Treating B-cell cancer with T cells expressing anti-CD19 chimeric antigen receptors, Nat Rev Clin Oncol. mai, vol.10, issue.5, pp.267-76, 2013.

A. D. Cohen, A. L. Garfall, E. A. Stadtmauer, J. J. Melenhorst, S. F. Lacey et al.,

, B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma, Journal of Clinical Investigation. 29 avr, vol.129, issue.6, pp.2210-2231, 2019.

X. Leleu, J. B. Micol, R. Guieze, C. Berthon, F. Kuhnovsky et al.,

, Thalidomide : mécanismes d'action et indications en hématologie. La Revue de Médecine Interne, févr, vol.26, issue.2, pp.119-146, 2005.

G. Fouquet, M. Macro, O. Decaux, C. Fohrer, S. Guidez et al.,

, Rev Med Interne. sept, vol.36, issue.9, pp.613-621, 2015.

M. H. Ma, H. H. Yang, K. Parker, S. Manyak, J. M. Friedman et al., The Proteasome Inhibitor PS-341 Markedly Enhances Sensitivity of Multiple Myeloma Tumor Cells to Chemotherapeutic Agents, vol.10, 2003.

P. G. Richardson, D. Irwin, J. Harousseau, H. Goldschmidt, J. Bladé et al., Bortezomib or High-Dose Dexamethasone for Relapsed Multiple Myeloma, The New England Journal of Medicine, vol.12, 2005.

R. Z. Orlowski and E. L. Zeger, Targeting the proteasome as a therapeutic strategy against haematological malignancies, Expert Opinion on Investigational Drugs. févr, vol.15, issue.2, pp.117-147, 2006.

, Commission de la Transparence, KYPROLIS [Internet], HAS, 2018.

A. K. Stewart, S. V. Rajkumar, M. A. Dimopoulos, T. Masszi, I. ?pi?ka et al.,

L. Carfilzomib, D. For-relapsed-multiple, and . Myeloma, New England Journal of Medicine. 8 janv, vol.372, issue.2, pp.142-52, 2015.

P. Moreau, T. Masszi, N. Grzasko, N. J. Bahlis, M. Hansson et al., Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma, New England Journal of Medicine. 28 avr, vol.374, issue.17, pp.1621-1655, 2016.

V. M. Richon, Cancer biology: mechanism of antitumour action of vorinostat (suberoylanilide hydroxamic acid), a novel histone deacetylase inhibitor, British Journal of Cancer. déc, vol.95, issue.S1, pp.2-6, 2006.

M. Dimopoulos, D. S. Siegel, S. Lonial, J. Qi, R. Hajek et al., Vorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, double-blind study. The Lancet Oncology, vol.14, pp.1129-1169, 2013.

J. F. San-miguel, V. Hungria, S. Yoon, M. Beksac, M. A. Dimopoulos et al., Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. The Lancet Oncology, vol.15, pp.1195-206, 2014.

P. G. Richardson, R. L. Schlossman, M. Alsina, D. M. Weber, S. E. Coutre et al., PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma, Blood, vol.122, issue.14, pp.2331-2338, 2013.

I. M. Ghobrial, E. Weller, R. Vij, N. C. Munshi, R. Banwait et al., Weekly bortezomib in combination with temsirolimus in relapsed or relapsed and refractory multiple myeloma: a multicentre, phase 1/2, open-label, dose-escalation study, The Lancet Oncology. mars, vol.12, issue.3, pp.263-72, 2011.

G. J. Morgan, F. E. Davies, W. M. Gregory, K. Cocks, S. E. Bell et al., First-line treatment with zoledronic acid as compared with clodronic acid in multiple myeloma (MRC Myeloma IX): a randomised controlled trial, The Lancet. déc, vol.376, issue.9757, pp.1989-99, 2010.

D. H. Henry, L. Costa, F. Goldwasser, V. Hirsh, V. Hungria et al.,

. 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, JCO. 20 mars, vol.29, issue.9, p.26, 2011.

M. Mateos, J. M. Hernandez, M. T. Hernandez, N. C. Gutierrez, L. Palomera et al., Bortezomib plus melphalan and prednisone in elderly untreated patients with multiple myeloma: updated time-to-events results and prognostic factors for time to progression, Haematologica. 1 avr, vol.93, issue.4, pp.560-565, 2008.

B. Durie, J. Harousseau, J. S. Miguel, J. Bladé, B. Barlogie et al., International uniform response criteria for multiple myeloma, Leukemia. sept, vol.20, issue.9, pp.1467-73, 2006.

S. Kumar, B. Paiva, K. C. Anderson, B. Durie, O. Landgren et al., International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma, The Lancet Oncology. août, vol.17, issue.8, pp.328-374, 2016.

B. Paiva, M. Vidriales, J. Cerveró, G. Mateo, J. J. Pérez et al.,

, Multiparameter flow cytometric remission is the most relevant prognostic factor for multiple myeloma patients who undergo autologous stem cell transplantation, Blood, vol.15, issue.10, pp.4017-4040, 2008.

A. Perrot, V. Lauwers-cances, J. Corre, N. Robillard, C. Hulin et al., Minimal residual disease negativity using deep sequencing is a major prognostic factor in multiple myeloma, Blood. 6 déc, vol.132, issue.23, pp.2456-64, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02006039

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell. mars, vol.144, issue.5, pp.646-74, 2011.

A. J. Friedenstein, I. Piatetzky-shapiro, and K. V. Petrakova, Osteogenesis in transplants of bone marrow cells, p.15

A. J. Friedenstein, R. K. Chailakhyan, N. V. Latsinik, and A. F. Panasyuk,

, Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues: Cloning In Vitro and Retransplantation In Vivo, Transplantation. avr, vol.17, issue.4, pp.641-50, 1974.

M. Dominici, L. Blanc, K. Mueller, I. Slaper-cortenbach, I. Marini et al., Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement, Cytotherapy, vol.8, issue.4, pp.315-322, 2006.

J. K. Fraser, I. Wulur, Z. Alfonso, and M. H. Hedrick, Fat tissue: an underappreciated source of stem cells for biotechnology, Trends in Biotechnology. avr, vol.24, issue.4, pp.150-154, 2006.

S. Li, K. Huang, J. Wu, M. S. Hu, M. Sanyal et al., Peripheral Blood-Derived Mesenchymal Stem Cells: Candidate Cells Responsible for Healing Critical-Sized Calvarial Bone Defects: Blood-Derived MSCs in Calvarial Bone Healing, STEM CELLS Translational Medicine. avr, vol.4, issue.4, pp.359-68, 2015.

R. Hass, C. Kasper, S. Böhm, and R. Jacobs, Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC, Cell Commun Signal. déc, vol.9, issue.1, p.12, 2011.

M. Wu, R. Zhang, Q. Zou, Y. Chen, M. Zhou et al., Comparison of the Biological Characteristics of Mesenchymal Stem Cells Derived from the Human Placenta and Umbilical Cord, Sci Rep. déc, vol.8, issue.1, p.5014, 2018.

A. Wilson and A. Trumpp, Bone-marrow haematopoietic-stem-cell niches, Nat Rev Immunol. févr, vol.6, issue.2, pp.93-106, 2006.

G. M. Spaggiari, H. Abdelrazik, F. Becchetti, and L. Moretta, MSCs inhibit monocyte-derived DC maturation and function by selectively interfering with the generation of immature DCs: central role of MSC-derived prostaglandin E2, Blood. 25 juin, vol.113, issue.26, pp.392-402, 2009.

G. Ren, L. Zhang, X. Zhao, G. Xu, Y. Zhang et al., Mesenchymal Stem Cell-Mediated Immunosuppression Occurs via Concerted Action of Chemokines and Nitric Oxide, Cell Stem Cell. févr, vol.2, issue.2, pp.141-50, 2008.

M. Castells, B. Thibault, E. Mery, M. Golzio, M. Pasquet et al., Ovarian ascites-derived Hospicells promote angiogenesis via activation of macrophages, Cancer Letters. déc, vol.326, issue.1, pp.59-68, 2012.

A. L. Naour, M. Prat, B. Thibault, R. Mével, L. Lemaitre et al., Tumor cells educate mesenchymal stromal cells to release chemoprotective and immunomodulatory factors, Journal of Molecular Cell Biology. 3 sept, p.90, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02390801

N. Espagnolle, A. Balguerie, E. Arnaud, L. Sensebé, and A. Varin, CD54-Mediated Interaction with Pro-inflammatory Macrophages Increases the Immunosuppressive Function of Human Mesenchymal Stromal Cells, Stem Cell Reports. avr, vol.8, issue.4, pp.961-76, 2017.

J. Centeno, C. , R. Schultz, J. Cheever, M. Freeman et al., Safety and Complications Reporting Update on the Re-Implantation of Culture-Expanded Mesenchymal Stem Cells Using Autologous Platelet Lysate Technique, CSCR. 1 déc, vol.6, issue.4, pp.368-78, 2011.

V. Gangji, J. Hauzeur, C. Matos, D. Maertelaer, V. Toungouz et al., Treatment of Osteonecrosis of the Femoral Head with Implantation of Autologous Bone-Marrow Cells: A Pilot Study, The Journal of Bone and Joint Surgery-American, vol.86, issue.6, pp.1153-60, 2004.

C. Peeters, M. Leijs, M. Reijman, G. Van-osch, and P. K. Bos, Safety of intraarticular cell-therapy with culture-expanded stem cells in humans: a systematic literature review. Osteoarthritis and Cartilage, vol.21, pp.1465-73, 2013.

A. Vega, M. A. Martín-ferrero, D. Canto, F. Alberca, M. García et al., Treatment of Knee Osteoarthritis With Allogeneic Bone Marrow Mesenchymal Stem Cells: A Randomized Controlled Trial, Transplantation. août, vol.99, issue.8, pp.1681-90, 2015.

J. M. Hare, J. E. Fishman, G. Gerstenblith, D. Velazquez, D. L. Zambrano et al., Comparison of Allogeneic vs Autologous Bone Marrow-Derived Mesenchymal Stem Cells Delivered by Transendocardial Injection in Patients With Ischemic Cardiomyopathy: The POSEIDON Randomized Trial, JAMA. 12 déc, vol.308, issue.22, p.2369, 2012.

A. Shpichka, D. Butnaru, E. A. Bezrukov, R. B. Sukhanov, A. Atala et al., Bilateral Transplantation of Allogenic Adult Human Bone Marrow-Derived Mesenchymal Stem Cells into the Subventricular Zone of Parkinson's Disease: A Pilot Clinical Study, Stem Cells International, vol.10, issue.1, pp.1-12, 2012.

P. Kebriaei, L. Isola, E. Bahceci, K. Holland, S. Rowley et al., Adult Human Mesenchymal Stem Cells Added to Corticosteroid Therapy for the Treatment of Acute Graftversus-Host Disease, Biology of Blood and Marrow Transplantation. juill, vol.15, issue.7, pp.804-815, 2009.

S. Ciavarella, A. Caselli, A. V. Tamma, A. Savonarola, G. Loverro et al., A Peculiar Molecular Profile of Umbilical Cord-Mesenchymal Stromal Cells Drives Their Inhibitory Effects on Multiple Myeloma Cell Growth and Tumor Progression, Stem Cells and Development. 15 juin, vol.24, issue.12, pp.1457-70, 2015.

S. Kidd, E. Spaeth, J. L. Dembinski, M. Dietrich, K. Watson et al., Direct Evidence of Mesenchymal Stem Cell Tropism for Tumor and Wounding Microenvironments Using In Vivo Bioluminescent Imaging. Stem Cells, vol.27, pp.2614-2637, 2009.

G. Ren, X. Zhao, Y. Wang, X. Zhang, X. Chen et al., CCR2-Dependent Recruitment of Macrophages by Tumor-Educated Mesenchymal Stromal Cells Promotes Tumor Development and Is Mimicked by TNF?, Cell Stem Cell. déc, vol.11, issue.6, pp.812-836, 2012.

E. Spaeth, A. Klopp, J. Dembinski, M. Andreeff, and F. Marini, Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells, Gene Ther. mai, vol.15, issue.10, pp.730-738, 2008.

K. Mandel, Y. Yang, A. Schambach, S. Glage, A. Otte et al., Mesenchymal Stem Cells Directly Interact with Breast Cancer Cells and Promote Tumor Cell Growth In Vitro and In Vivo, Stem Cells and Development. déc, vol.22, issue.23, pp.3114-3141, 2013.

K. A. Waite and C. Eng, From developmental disorder to heritable cancer: it's all in the BMP/TGF-? family, Nat Rev Genet, vol.4, issue.10, pp.763-73, 2003.

S. R. Wallace, M. M. Oken, K. L. Lunetta, A. Panoskaltsis-mortari, and A. M. Masellis, Abnormalities of bone marrow mesenchymal cells in multiple myeloma patients, p.12

B. Arnulf, S. Lecourt, J. Soulier, B. Ternaux, M. Lacassagne et al., Phenotypic and functional characterization of bone marrow mesenchymal stem cells derived from patients with multiple myeloma, Leukemia. janv, vol.21, issue.1, pp.158-63, 2007.

J. Corre, K. Mahtouk, M. Attal, M. Gadelorge, A. Huynh et al., Bone marrow mesenchymal stem cells are abnormal in multiple myeloma, Leukemia. mai, vol.21, issue.5, pp.1079-88, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00270565

L. Garderet, C. Mazurier, A. Chapel, I. Ernou, L. Boutin et al., Mesenchymal stem cell abnormalities in patients with multiple myeloma, Leukemia & Lymphoma. janv, vol.48, issue.10, pp.2032-2073, 2007.

W. G. Gunn, A. Conley, L. Deininger, S. D. Olson, D. J. Prockop et al., A Crosstalk Between Myeloma Cells and Marrow Stromal Cells Stimulates Production of DKK1 and Interleukin-6: A Potential Role in the Development of Lytic Bone Disease and Tumor Progression in Multiple Myeloma, STEM CELLS. avr, vol.24, issue.4, pp.986-91, 2006.

A. Garcia-gomez, J. Rivas, E. M. Ocio, E. Díaz-rodríguez, J. C. Montero et al., Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: implications in myeloma progression and myeloma bone disease. Oncotarget [Internet]. 30 sept, vol.5, 2014.

J. Corre, E. Labat, N. Espagnolle, B. Hebraud, H. Avet-loiseau et al., Bioactivity and Prognostic Significance of Growth Differentiation Factor GDF15 Secreted by Bone Marrow Mesenchymal Stem Cells in Multiple Myeloma, Cancer Research. 15 mars, vol.72, issue.6, pp.1395-406, 2012.

M. Garayoa, J. L. Garcia, C. Santamaria, A. Garcia-gomez, J. F. Blanco et al., Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors, Leukemia. août, vol.23, issue.8, pp.1515-1542, 2009.

T. André, N. Meuleman, B. Stamatopoulos, C. De-bruyn, K. Pieters et al., Evidences of Early Senescence in Multiple Myeloma Bone Marrow Mesenchymal Stromal Cells. Covas DT, éditeur, PLoS ONE. 21 mars, vol.8, issue.3, p.59756, 2013.

C. Giallongo, D. Tibullo, N. L. Parrinello, L. Cava, P. et al.,

, Granulocyte-like myeloid derived suppressor cells (G-MDSC) are increased in multiple myeloma and are driven by dysfunctional mesenchymal stem cells (MSC), Oncotarget, vol.27, issue.2016

, Disponible sur

B. Zdzisi?ska, A. Bojarska-junak, A. Dmoszy?ska, and M. Kandefer-szersze?, Abnormal cytokine production by bone marrow stromal cells of multiple myeloma patients in response to RPMI8226 myeloma cells, Arch Immunol Ther Exp. juin, vol.56, issue.3, pp.207-228, 2008.

G. Mcnee, K. L. Eales, W. Wei, D. S. Williams, A. Barkhuizen et al., Citrullination of histone H3 drives IL-6 production by bone marrow mesenchymal stem cells in MGUS and multiple myeloma, Leukemia. févr, vol.31, issue.2, pp.373-81, 2017.

X. Wang, Z. Zhang, and C. Yao, Survivin is upregulated in myeloma cell lines cocultured with mesenchymal stem cells, Leukemia Research, vol.34, issue.10, pp.1325-1334, 2010.

A. M. Roccaro, A. Sacco, P. Maiso, A. K. Azab, Y. Tai et al., BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression, J Clin Invest. 1 avr, vol.123, issue.4, pp.1542-55, 2013.

Y. Mori, N. Shimizu, M. Dallas, M. Niewolna, B. Story et al., Anti-?4 integrin antibody suppresses the development of multiple myeloma and associated osteoclastic osteolysis, Blood, vol.104, issue.7, pp.2149-54, 2004.

B. Li, J. Fu, P. Chen, and W. Zhuang, Impairment in Immunomodulatory Function of Mesenchymal Stem Cells from Multiple Myeloma Patients. Archives of Medical Research, vol.41, pp.623-656, 2010.

Y. Kawano, S. Fujiwara, N. Wada, M. Izaki, Y. H. Okuno et al., Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide, International Journal of Oncology. sept, vol.41, issue.3, pp.876-84, 2012.

M. Kaiser, M. Mieth, P. Liebisch, R. Oberländer, J. Rademacher et al., Serum concentrations of DKK-1 correlate with the extent of bone disease in patients with multiple myeloma: Serum DKK-1 correlates with myeloma bone disease, European Journal of Haematology. juin, vol.80, issue.6, pp.490-494, 2008.

G. A. Clines, K. S. Mohammad, Y. Bao, O. W. Stephens, L. J. Suva et al.,

, Dickkopf Homolog 1 Mediates Endothelin-1-Stimulated New Bone Formation, Molecular Endocrinology. 1 févr, vol.21, issue.2, pp.486-98, 2007.

E. Tian, F. Zhan, R. Walker, E. Rasmussen, Y. Ma et al., The Role of the Wnt-Signaling Antagonist DKK1 in the Development of Osteolytic Lesions in Multiple Myeloma, N Engl J Med. 25 déc, vol.349, issue.26, pp.2483-94, 2003.

J. J. Pinzone, B. M. Hall, N. K. Thudi, M. Vonau, Y. Qiang et al., The role of Dickkopf-1 in bone development, homeostasis, and disease, Blood. 15 janv, vol.113, issue.3, pp.517-542, 2009.

C. Christodoulides, The Wnt antagonist Dickkopf-1 and its receptors are coordinately regulated during early human adipogenesis, Journal of Cell Science. 15 juin, vol.119, issue.12, pp.2613-2633, 2006.

T. Gaur, C. J. Lengner, H. Hovhannisyan, R. A. Bhat, P. Bodine et al.,

W. Canonical and . Signaling, Promotes Osteogenesis by Directly Stimulating Runx2 Gene Expression, J Biol Chem. 30 sept, vol.280, issue.39, pp.33132-33172, 2005.

S. A. Siefert, C. Chabasse, S. Mukhopadhyay, M. H. Hoofnagle, D. K. Strickland et al., Enhanced venous thrombus resolution in plasminogen activator inhibitor type-2 deficient mice, J Thromb Haemost, vol.12, issue.10, pp.1706-1722, 2014.

D. R. Croucher, D. N. Saunders, S. Lobov, and M. Ranson, Revisiting the biological roles of PAI2 (SERPINB2) in cancer, Nature Reviews Cancer. juill, vol.8, issue.7, pp.535-580, 2008.

M. Yagci, G. T. Sucak, and R. Haznedar, Fibrinolytic activity in multiple myeloma, Am J Hematol. déc, vol.74, issue.4, pp.231-238, 2003.