689
Views
11
CrossRef citations to date
0
Altmetric
Review

Emerging treatment approaches for myeloma-related bone disease

, , &
Pages 217-228 | Received 18 Oct 2016, Accepted 13 Jan 2017, Published online: 29 Jan 2017

References

  • Ludwig H, Miguel JS, Dimopoulos MA, et al. International Myeloma Working Group recommendations for global myeloma care. Leukemia. 2013;28:981–992.
  • Kyle RA, Gertz MA, Witzig TE, et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin Proc. 2003;78:21–33.
  • Christoulas D, Terpos E, Dimopoulos MA. Pathogenesis and management of myeloma bone disease. Expert Rev Hematol. 2009;2:385–398.
  • Berenson JR, Lichtenstein A, Porter L, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. Myeloma Aredia Study Group. N Engl J Med. 1996;334:488–493.
  • Melton LJ 3rd, Kyle RA, Achenbach SJ, et al. Fracture risk with multiple myeloma: a population-based study. J Bone Miner Res. 2005;20:487–493.
  • Lecouvet FE, vande Berg BC, Maldague BE, et al. Vertebral compression fractures in multiple myeloma. Part I. Distribution and appearance at MR imaging. Radiology. 1997;204:195–199.
  • Terpos E, Dimopoulos MA. Myeloma bone disease: pathophysiology and management. Ann Oncol. 2005;16(8):1223–1231.
  • Terpos E, Berenson J, Raje N, et al. Management of bone disease in multiple myeloma. Expert Rev Hematol. 2014;7:113–125.
  • Fowler JA, Edwards CM, Croucher PI. Tumor-host cell interactions in the bone disease of myeloma. Bone. 2011;48:121–128. DOI:10.1016/j.bone.2010.06.029
  • Kovacic N, Croucher PI, McDonald MM. Signaling between tumor cells and the host bone marrow microenvironment. Calcif Tissue Int. 2013;94:125–139. DOI:10.1007/s00223-013-9794-7
  • Terpos E, Szydlo R, Apperley JF, et al. Soluble receptor activator of nuclear factor kappaB ligand-osteoprotegerin ratio predicts survival in multiple myeloma: proposal for a novel prognostic index. Blood. 2003;102:1064–1069.
  • Tian E, Zhan F, Walker R, et al. The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med. 2003;349:2483–2494.
  • Terpos E, Morgan G, Dimopoulos MA, et al. International Myeloma Working Group recommendations for the treatment of multiple myeloma-related bone disease. J Clin Oncol. 2013;31:2347–2357.
  • Rogers MJ, Crockett JC, Coxon FP, et al. Biochemical and molecular mechanisms of action of bisphosphonates. Bone. 2011;49:34–41.
  • Terpos E, Kleber M, Engelhardt M, et al. European Myeloma Network guidelines for the management of multiple myeloma-related complications. Haematologica. 2016;101:115–208.
  • Morgan GJ, Davies FE, Gregory WM, et al. Effects of induction and maintenance plus long-term bisphosphonates on bone disease in patients with multiple myeloma: the Medical Research Council Myeloma IX Trial. Blood. 2012;119:5374–5383.
  • Terpos E, Confavreux CB, Clézardin P. Bone antiresorptive agents in the treatment of bone metastases associated with solid tumours or multiple myeloma. Bonekey Rep. 2015;4:744.
  • Terpos E, Dimopoulos MA, Berenson J. Effects of bone-targeted agents on cancer progression and mortality. Established role of bisphosphonate therapy for prevention of skeletal complications from myeloma bone disease. Crit Rev Oncol Hematol. 2011;77(Suppl 1):S13–23.
  • Terpos E, Roodman GD, Dimopoulos MA. Optimal use of bisphosphonates in patients with multiple myeloma. Blood. 2013;121:3325–3328.
  • A double-blind, placebo-controlled, randomized phase 2 study of BHQ880, an Anti-Dickkopf1 (DKK1) Monoclonal Antibody (mAb), in patients with untreated multiple myeloma and renal insufficiency. n d [cited 2015 Dec 18]. Available from: https://clinicaltrials.gov/ct2/show/record/NCT01337752
  • A phase Ib/II multicenter dose-determination study, with an adaptive, randomized, placebo-controlled, double-blind phase II, using various repeated IV doses of BHQ880 in combination with zoledronic acid in relapsed or refractory myeloma patients with prior n d. [cited 2015 Jan 18]. Available from: https://clinicaltrials.gov/ct2/show/NCT00741377
  • Morgan GJ, Child JA, Gregory WM, et al. Effects of zoledronic acid versus clodronic acid on skeletal morbidity in patients with newly diagnosed multiple myeloma (MRC Myeloma IX): secondary outcomes from a randomised controlled trial. Lancet Oncol. 2011;12(8):743–752.
  • Ria R, Reale A, Moschetta M, et al. A retrospective study of skeletal and disease-free survival benefits of zoledronic acid therapy in patients with multiple myeloma treated with novel agents. Int J Clin Exp Med. 2013;6(1):30–38.
  • Terpos E, Christoulas D, Kokkoris P, et al. Increased bone mineral density in a subset of patients with relapsed multiple myeloma who received the combination of bortezomib, dexamethasone and zoledronic acid. Ann Oncol. 2010;21:1561–1562.
  • Moschetta M, Di Pietro G, Ria R, et al. Bortezomib and zoledronic acid onangiogenic and vasculogenic activities of bonemarrow macrophages in patients with multiple myeloma. Eur J Cancer. 2010;46:420–429.
  • Lacey DL, Timms E, Tan HL, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998;93:165–176.
  • Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397:315–323.
  • Simonet WS, Lacey DL, Dunstan CR, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89:309–319.
  • Alexander SPH, Benson HE, Faccenda E, et al. CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: catalytic receptors. Br J Pharmacol. 2013a;170:1676–1705.
  • Terpos E, Efstathiou E, Christoulas D, et al. RANKL inhibition: clinical implications for the management of patients with multiple myeloma and solid tumors with bone metastases. Expert Opin Biol Ther. 2009;9:465–479.
  • Jakob C, Goerke A, Terpos E, et al. Serum levels of total-RANKL in multiple myeloma. Clin Lymphoma Myeloma. 2009;9:430–435.
  • Roodman GD, Dougall WC. RANK ligand as a therapeutic target for bone metastases and multiple myeloma. Cancer Treat Rev. 2008;34(1):92–101.
  • Giuliani N, Bataille R, Mancini C, et al. Myeloma cells induce imbalance in the osteoprotegerin/osteoprotegerin ligand system in the human bone marrow environment. Blood. 2001;98:3527–3533.
  • Croucher PI, Shipman CM, Lippitt JM, et al. Osteoprotegerin inhibits the development of osteolytic bone disease in multiple myeloma. Blood. 2001;98:3534–3540.
  • Seidel C, Hjertner O, Abildgaard N, et al. Serum osteoprotegerin levels are reduced in patients with multiple myeloma with lytic bone disease. Blood. 2001;98:2269–2271.
  • Lipton ASG, Figueroa J, Alvarado C, et al. Randomized active-controlled phase II study of denosumab efficacy and safety in patients with breast cancer-related bone metastases. J Clin Oncol. 2007;25:4431–4437.
  • Lipton ASG, Figueroa J, Alvarado C, et al. Extended efficacy and safety of denosumab in breast cancer patients with bone metastases not receiving prior bisphosphonate therapy. Clin Cancer Res. 2008;14:6690–6696.
  • Fizazi KLA, Mariette X, Body JJ, et al. Randomized phase II trial of denosumab in patients with bone metastases from prostate cancer, breast cancer, or other neoplasms after intravenous bisphosphonates. J Clin Oncol. 2009;27:1564–1571.
  • Raje N, Vadhan-Raj S, Willenbacher W, et al. Evaluating results from the multiple myeloma patient subset treated with denosumab or zoledronic acid in a randomized phase 3 trial. Blood Cancer J. 2016;6:e378.
  • Stopeck ATLA, Body JJ, Steger GG, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol. 2010;28:5132–5139.
  • Henry DHCL, Goldwasser F, Hirsh V, 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. J Clin Oncol. 2011;29:1125–1132.
  • Bafico A, Liu G, Yaniv A, et al. Novel mechanism of Wnt signalling inhibition mediated by Dickkopf-1 interaction with LRP6/Arrow. Nat Cell Biol. 2001;3:683–686.
  • Mao B, Wu W, Li Y, et al. LDL-receptor-related protein 6 is a receptor for Dickkopf proteins. Nature. 2001;411:321–325.
  • Gunn WG, Conley A, Deininger L, 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. 2006;24:986–991.
  • Gavriatopoulou M, Dimopoulos MA, Christoulas D, et al. Dickkopf-1: a suitable target for the management of myeloma bone disease. Expert Opin Ther Targets. 2009;13:839–848.
  • Politou MC, Heath DJ, Rahemtulla A, et al. Serum concentrations of Dickkopf-1 protein are increased in patients with multiple myeloma and reduced after autologous stem cell transplantation. Int J Cancer. 2006;119:1728–1731.
  • Qian J, Zheng Y, Zheng C, et al. Active vaccination with Dickkopf-1 induces protective and therapeutic antitumor immunity in murine multiple myeloma. Blood. 2012;119:161–169.
  • Fulciniti M, Tassone P, Hideshima T, et al. Anti-DKK1 mAb (BHQ880) as a potential therapeutic agent for multiple myeloma. Blood. 2009;114:371–379.
  • Pozzi S, Fulciniti M, Yan H, et al. In vivo and in vitro effects of a novel anti-Dkk1 neutralizing antibody in multiple myeloma. Bone. 2013;53:487–496.
  • Munshi NC, Beck JT, Bensinger W, et al. Early evidence of anabolic bone activity of BHQ880, a fully human anti-dkk1 neutralizing antibody: results of a phase 2 study in previously untreated patients with smoldering multiple myeloma at risk for progression. Blood. 2012;120:Abstract 331. ASH, Atlanta, Georgia
  • Qiang YW, Walsh K, Yao L, et al. Wnts induce migration and invasion of myeloma plasma cells. Blood. 2005;106:1786–1793.
  • Edwards CM, Edwards JR, Lwin ST, et al. Increasing Wnt signaling in the bone marrow microenvironment inhibits the development of myeloma bone disease and reduces tumor burden in bone in vivo. Blood. 2008;111:2833–2842.
  • Balemans W, Ebeling M, Patel N, et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum Mol Genet. 2001;10:537–543.
  • Balemans W, Patel N, Ebeling M, et al. Identification of a 52-kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002;39:91–97.
  • Brunetti G, Oranger A, Mori G, et al. Sclerostin is overexpressed by plasma cells from multiple myeloma patients. Ann N Y Acad Sci. 2011;1237:19–23.
  • Terpos E, Christoulas D, Katodritou E, et al. Elevated circulating sclerostin correlates with advanced disease features and abnormal bone remodeling in symptomatic myeloma: reduction post-bortezomib monotherapy. Int J Cancer. 2012;131:1466–1471.
  • Colucci S, Brunetti G, Oranger A, et al. Myeloma cells suppress osteoblasts through sclerostin secretion. Blood Cancer J. 2011;1:e27.
  • Gkotzamanidou M, Dimopoulos MA, Kastritis E, et al. Sclerostin: a possible target for the management of cancer-induced bone disease. Expert Opin Ther Targets. 2012;16:761–769.
  • van Bezooijen RL, Ten Dijke P, Papapoulos SE, et al. SOST/sclerostin, an osteocyte-derived negative regulator of bone formation. Cytokine Growth Factor Rev. 2005;16:319–327.
  • Sutherland MK, Geoghegan JC, Yu C, et al. Sclerostin promotes the apoptosis of human osteoblastic cells: a novel regulation of bone formation. Bone. 2004;35:828–835.
  • Winkler DG, Sutherland MK, Geoghegan JC, et al. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. Embo J. 2003;22:6267–6276.
  • Li X, Ominsky MS, Warmington KS, et al. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Miner Res. 2009;24:578–588.
  • Li X, Warmington KS, Niu QT, et al. Inhibition of sclerostin by monoclonal antibody increases bone formation, bone mass, and bone strength in aged male rats. J Bone Miner Res. 2010;25:2647–2656.
  • Agholme F, Li X, Isaksson H, et al. Sclerostin antibody treatment enhances metaphyseal bone healing in rats. J Bone Miner Res. 2010;25:2412–2418.
  • Ominsky MS, Li C, Li X, et al. Inhibition of sclerostin by monoclonal antibody enhances bone healing and improves bone density and strength of nonfractured bones. J Bone Miner Res. 2011;26:1012–1021.
  • Chen XX, Baum W, Dwyer D, et al. Sclerostin inhibition reverses systemic, periarticular and local bone loss in arthritis. Ann Rheum Dis. 2013;72:1732–1736.
  • McColm J, Hu L, Womack T, et al. Single- and multiple-dose randomized studies of blosozumab, a monoclonal antibody against sclerostin, in healthy postmenopausal women. J Bone Miner Res. 2013;29:935–943.
  • Padhi D, Jang G, Stouch B, et al. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J Bone Miner Res. 2011;26:19–26.
  • McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412–420.
  • Norris RA, Moreno-Rodriguez R, Hoffman S, et al. The many facets of the matricellular protein periostin during cardiac development, remodeling, and pathophysiology. J Cell Commun Signal. 2009;3:275–286.
  • Cobo T, Viloria CG, Solares L, et al. Role of periostin in adhesion and migration of bone remodeling cells. PLoS One. 2016;11:e0147837.
  • Merle B, Bouet G, Rousseau JC, et al. Periostin and transforming growth factor β-induced protein (TGFβIp) are both expressed by osteoblasts and osteoclasts. Cell Biol Int. 2014;38:398–404.
  • Bonnet N, Garnero P, Ferrari S. Periostin action in bone. Mol Cell Endocrinol. 2015;432:75–82.
  • Rousseau JC, Sornay-Rendu E, Bertholon C, et al. Serum periostin is associated with fracture risk in postmenopausal women: a 7-year prospective analysis of the OFELY study. J Clin Endocrinol Metab. 2014;99:2533–2539.
  • Kim BJ, Rhee Y, Kim CH, et al. Plasma periostin associates significantly with non-vertebral but not vertebral fractures in postmenopausal women: clinical evidence for the different effects of periostin depending on the skeletal site. Bone. 2015;81:435–441.
  • Sakellariou GT, Anastasilakis AD, Bisbinas I, et al. Circulating periostin levels in patients with AS: association with clinical and radiographic variables, inflammatory markers and molecules involved in bone formation. Rheumatology (Oxford). 2015;54:908–914.
  • Terpos E, Kastritis E, Christoulas D, et al. Circulating activin-A is elevated in patients with advanced multiple myeloma and correlates with extensive bone involvement and inferior survival; no alterations post-lenalidomide and dexamethasone therapy. Ann Oncol. 2012;23:2681–2686.
  • Kapoor S. Periostin and its emerging role in systemic carcinogenesis. Osteoporos Int. 2014;25:1423–1424.
  • Underwood TJ, Hayden AL, Derouet M, et al. Cancer associated fibroblasts predict for poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma. J Pathol. 2015;235:466–477.
  • Hu F, Shang XF, Wang W, et al. High-level expression of periostin is significantly correlated with tumour angiogenesis and poor prognosis in osteosarcoma. Int J Exp Pathol. 2016;97:86–92.
  • Contié S, Voorzanger-Rousselot N, Litvin J, et al. Increased expression and serum levels of the stromal cell-secreted protein periostin in breast cancer bone metastases. Int J Cancer. 2011;128:352–360.
  • Terpos E, Christoulas D, Kastritis E, et al. High levels of periostin correlate with increased fracture rate, diffuse MRI pattern, abnormal bone remodeling and advanced disease stage in patients with newly diagnosed symptomatic multiple myeloma. Blood Cancer J. 2016;6:e482.
  • Kyutoku M, Taniyama Y, Katsuragi N, et al. Role of periostin in cancer progression and metastasis: inhibition of breast cancer progression and metastasis by anti-periostin antibody in a murine model. Int J Mol Med. 2011;28:181–186.
  • Brosh N, Sternberg D, Honigwachs-Sha’anani J, et al. The plasmacytoma growth inhibitor restrictin-P is an antagonist of interleukin 6 and interleukin 11. Identification as a stroma-derived activin A. J Biol Chem. 1995;270:29594–29600.
  • Sternberg D, Honigwachs-Sha’anani J, Brosh N, et al. Restrictin-P/stromal activin A, kills its target cells via an apoptotic mechanism. Growth Factors. 1995;12:277–287.
  • Deli A, Kreidl E, Santifaller S, et al. Activins and activin antagonists in hepatocellular carcinoma. World J Gastroenterol. 2008;14:1699–1709.
  • Hashimoto M, Shoda A, Inoue S, et al. Functional regulation of osteoblastic cells by the interaction of activin-A with follistatin. J Biol Chem. 1992;267:4999–5004.
  • Futakuchi M, Nannuru KC, Varney ML, et al. Transforming growth factor-beta signaling at the tumor-bone interface promotes mammary tumor growth and osteoclast activation. Cancer Sci. 2009;100:71–81.
  • Butler CM, Gold EJ, Risbridger GP. Should activin betaC be more than a fading snapshot in the activin/TGFbeta family album? Cytokine Growth Factor Rev. 2005;16:377–385.
  • Sugatani T, Alvarez UM, Hruska KA. Activin A stimulates IkappaB-alpha/NFkappaB and RANK expression for osteoclast differentiation, but not AKT survival pathway in osteoclast precursors. J Cell Biochem. 2003;90:59–67.
  • Masi L, Malentacchi C, Campanacci D, et al. Transforming growth factor-beta isoform and receptor expression in chondrosarcoma of bone. Virchows Arch. 2002;440:491–497.
  • Gobbi G, Sangiorgi L, Lenzi L, et al. Seven BMPs and all their receptors are simultaneously expressed in osteosarcoma cells. Int J Oncol. 2002;20:143–147.
  • Dowling CR, Risbridger GP. The role of inhibins and activins in prostate cancer pathogenesis. Endocr Relat Cancer. 2000;7:243–256.
  • Risbridger GP, Mellor SL, McPherson SJ, et al. The contribution of inhibins and activins to malignant prostate disease. Mol Cell Endocrinol. 2001;180:149–153.
  • Vallet S, Mukherjee S, Vaghela N, et al. Activin A promotes multiple myeloma-induced osteolysis and is a promising target for myeloma bone disease. Proc Natl Acad Sci U S A. 2010;107:5124–5129.
  • Lee JW, Chung HY, Ehrlich LA, et al. IL-3 expression by myeloma cells increases both osteoclast formation and growth of myeloma cells. Blood. 2004;103:2308–2315.
  • Fields SZ, Parshad S, Anne M, et al. Activin receptor antagonists for cancer-related anemia and bone disease. Expert Opin Investig Drugs. 2013;22:87–101.
  • Chantry AD, Heath D, Mulivor AW, et al. Inhibiting activin-A signaling stimulates bone formation and prevents cancer-induced bone destruction in vivo. J Bone Miner Res. 2010;25:2633–2646.
  • Scullen T, Santo L, Vallet S, et al. Lenalidomide in combination with an activin A-neutralizing antibody: preclinical rationale for a novel anti-myeloma strategy. Leukemia. 2013 Aug;27(8):1715–1721.
  • Abdulkadyrov KM, Salogub GN, Khuazheva NK, et al. Sotatercept in patients with osteolytic lesions of multiple myeloma. Br J Haematol. 2014;165:814–823.
  • Ruckle J, Jacobs M, Kramer W, et al. Single-dose, randomized, double-blind, placebo-controlled study of ACE-011 (ActRIIA-IgG1) in postmenopausal women. J Bone Miner Res. 2009;24:744–752.
  • Hirai H, Maru Y, Hagiwara K, et al. A novel putative tyrosine kinase receptor encoded by the eph gene. Science. 1987;238:1717–1720.
  • Mundy GR, Elefteriou F. Boning up on ephrin signaling. Cell. 2006;126:441–443.
  • Zhao C, Irie N, Takada Y, et al. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab. 2006;4:111–121.
  • Allan EH, Hausler KD, Wei T, et al. EphrinB2 regulation by PTH and PTHrP revealed by molecular profiling in differentiating osteoblasts. J Bone Miner Res. 2008;23:1170–1181.
  • Martin TJ, Allan EH, Ho PW, et al. Communication between ephrinB2 and EphB4 within the osteoblast lineage. Adv Exp Med Biol. 2010;658:51–60.
  • Takyar FM, Tonna S, Ho PW, et al. EphrinB2/EphB4 inhibition in the osteoblast lineage modifies the anabolic response to parathyroid hormone. J Bone Miner Res. 2013;28:912–925.
  • Bates AL, Mundy GR, Edwards CM. Myeloma cells decrease ephb4 expression in osteoblasts: a novel mechanism for regulation of bone formation in multiple myeloma. J Bone Miner Res. 2007;22:(S309)(abstract).
  • DiMascio L, Voermans C, Uqoezwa M, et al. Identification of adiponectin as a novel hemopoietic stem cell growth factor. J Immunol. 2007;178:3511–3520.
  • Swarbrick MM, Havel PJ. Physiological, pharmacological, and nutritional regulation of circulating adiponectin concentrations in humans. Metab Syndr Relat Disord. 2008;6:87–102.
  • Berner HS, Lyngstadaas SP, Spahr A, et al. Adiponectin and its receptors are expressed in bone-forming cells. Bone. 2004;35:842–849.
  • Shinoda Y, Yamaguchi M, Ogata N, et al. Regulation of bone formation by adiponectin through autocrine/paracrine and endocrine pathways. J Cell Biochem. 2006;99:196–208.
  • Oshima K, Nampei A, Matsuda M, et al. Adiponectin increases bone mass by suppressing osteoclast and activating osteoblast. Biochem Biophys Res Commun. 2005;331:520–526.
  • Yamaguchi N, Kukita T, Li YJ, et al. Adiponectin inhibits osteoclast formation stimulated by lipopolysaccharide from Actinobacillus actinomycetemcomitans. FEMS Immunol Med Microbiol. 2007;49:28–34.
  • Lenchik L, Register TC, Hsu FC, et al. Adiponectin as a novel determinant of bone mineral density and visceral fat. Bone. 2003;33:646–651.
  • Jurimae J, Rembel K, Jurimae T, et al. Adiponectin is associated with bone mineral density in perimenopausal women. Horm Metab Res. 2005;37:297–302.
  • Richards JB, Valdes AM, Burling K, et al. Serum adiponectin and bone mineral density in women. J Clin Endocrinol Metab. 2007;92:1517–1523.
  • Fowler JA, Lwin ST, Drake MT, et al. Host-derived adiponectin is tumor-suppressive and a novel therapeutic target for multiple myeloma and the associated bone disease. Blood. 2011;118:5872–5882.
  • de Weers M, Mensink RG, Kraakman ME, et al. Mutation analysis of the Bruton’s tyrosine kinase gene in X-linked agammaglobulinemia: identification of a mutation which affects the same codon as is altered in immunodeficient xid mice. Hum Mol Genet. 1994;3:161–166.
  • Lee SH, Kim T, Jeong D, et al. The tec family tyrosine kinase Btk Regulates RANKL-induced osteoclast maturation. J Biol Chem. 2008;283:11526–11534.
  • Shinohara M, Koga T, Okamoto K, et al. Tyrosine kinases Btk and Tec regulate osteoclast differentiation by linking RANK and ITAM signals. Cell. 2008;132:794–806.
  • Chauhan D, Auclair D, Robinson EK, et al. Identification of genes regulated by dexamethasone in multiple myeloma cells using oligonucleotide arrays. Oncogene. 2002;21:1346–1358.
  • Rushworth SA, Bowles KM, Barrera LN, et al. BTK inhibitor ibrutinib is cytotoxic to myeloma and potently enhances bortezomib and lenalidomide activities through NF-kappaB. Cell Signal. 2013;25:106–112.
  • Tai YT, Chang BY, Kong SY, et al. Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma. Blood. 2012;120:1877–1887.
  • Gilks CB, Bear SE, Grimes HL, et al. Progression of interleukin-2 (IL-2)-dependent rat T cell lymphoma lines to IL-2-independent growth following activation of a gene (Gfi-1) encoding a novel zinc finger protein. Mol Cell Biol. 1993;13:1759–1768.
  • D’Souza S, del Prete D, Jin S, et al. Gfi1 expressed in bone marrow stromal cells is a novel osteoblast suppressor in patients with multiple myeloma bone disease. Blood. 2011;118:6871–6880.
  • Jin SDS, Sun Q, Sammut B, et al. The transcription repressor Gfi1 directly interacts with the Runx2 gene in osteoblast precursors to mediate repression by multiple myeloma cells via TNFalpha, leading to blocked osteoblast differentiation. J Bone Miner Res. 2011;26(S1):S180.
  • Choi SJ, Cruz JC, Craig F, et al. Macrophage inflammatory protein 1-alpha is a potential osteoclast stimulatory factor in multiple myeloma. Blood. 2000;96:671–675.
  • Lentzsch S, Gries M, Janz M, et al. Macrophage inflammatory protein 1-alpha (MIP-1 alpha) triggers migration and signaling cascades mediating survival and proliferation in multiple myeloma (MM) cells. Blood. 2003;101:3568–3573.
  • Terpos E, Politou M, Szydlo R, et al. Serum levels of macrophage inflammatory protein-1 alpha (MIP-1alpha) correlate with the extent of bone disease and survival in patients with multiple myeloma. Br J Haematol. 2003;123:106–109.
  • Alexander SPH, Benson HE, Faccenda E, et al.; CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013b;170:1459–1581.
  • Terpos E, Politou M, Viniou N, et al. Significance of macrophage inflammatory protein-1 alpha (MIP-1a) in multiple myeloma. Leuk Lymphoma. 2005;46:1699–1707.
  • Oba Y, Lee JW, Ehrlich LA, et al. MIP-1alpha utilizes both CCR1 and CCR5 to induce osteoclast formation and increase adhesion of myeloma cells to marrow stromal cells. Exp Hematol. 2005;33:272–278.
  • Oyayobi B, Williams PJ, Pulkrabek D, et al. In vivo osteoclastogenic effects of the CC chemokine, macrophage inflammatory protein (MIP)-1α. Bone. 2001;27:S81.
  • Menu E, De Leenheer E, de Raeve H, et al. Role of CCR1 and CCR5 in homing and growth of multiple myeloma and in the development of osteolytic lesions: a study in the 5TMM model. Clin Exp Metastasis. 2006;23:291–300.
  • Vallet S, Pozzi S, Patel K, et al. A novel role for CCL3 (MIP-1alpha) in myeloma-induced bone disease via osteocalcin downregulation and inhibition of osteoblast function. Leukemia. 2011;25:1174–1181.
  • Novak AJ, Darce JR, Arendt BK, et al. Expression of BCMA, TACI, and BAFF-R in multiple myeloma: a mechanism for growth and survival. Blood. 2004;103:689–694.
  • Abe M, Kido S, Hiasa M, et al. BAFF and APRIL as osteoclast-derived survival factors for myeloma cells: a rationale for TACI-Fc treatment in patients with multiple myeloma. Leukemia. 2006;20:1313–1315.
  • Neri P, Kumar S, Fulciniti MT, et al. Neutralizing B-cell activating factor antibody improves survival and inhibits osteoclastogenesis in a severe combined immunodeficient human multiple myeloma model. Clin Cancer Res. 2007;13:5903–5909.
  • Raje N, Faber EA, Richardson PG, et al. Phase 1 study of tabalumab, a human anti-B-cell activating factor antibody, and bortezomib in patients with relapsed/refractory multiple myeloma. Clin Cancer Res. 2016;22(23):5688–5695 .
  • Garrett IR, Chen D, Gutierrez G, et al. Selective inhibitors of the osteoblast proteasome stimulate bone formation in vivo and in vitro. J Clin Invest. 2003;111:1771–1782.
  • Zavrski I, Krebbel H, Wildemann B, et al. Proteasome inhibitors abrogate osteoclast differentiation and osteoclast function. Biochem Biophys Res Commun. 2005;333:200–205.
  • Darnay BG, Besse A, Poblenz AT, et al. TRAFs in RANK signaling. Adv Exp Med Biol. 2007;597:152–159.
  • Pennisi A, Li X, Ling W, et al. The proteasome inhibitor, bortezomib suppresses primary myeloma and stimulates bone formation in myelomatous and nonmyelomatous bones in vivo. Am J Hematol. 2009;84:6–14.
  • Qiang YW, Hu B, Chen Y, et al. Bortezomib induces osteoblast differentiation via Wnt-independent activation of beta-catenin/TCF signaling. Blood. 2009;113:4319–4330.
  • Terpos E, Heath DJ, Rahemtulla A, et al. Bortezomib reduces serum dickkopf-1 and receptor activator of nuclear factor-kappaB ligand concentrations and normalises indices of bone remodelling in patients with relapsed multiple myeloma. Br J Haematol. 2006;135:688–692.
  • Heider U, Kaiser M, Muller C, et al. Bortezomib increases osteoblast activity in myeloma patients irrespective of response to treatment. Eur J Haematol. 2006;77:233–238.
  • Oyajobi BO, Garrett IR, Gupta A, et al. Stimulation of new bone formation by the proteasome inhibitor, bortezomib: implications for myeloma bone disease. Br J Haematol. 2007;139:434–438.
  • Zangari M, Yaccoby S, Pappas L, et al. A prospective evaluation of the biochemical, metabolic, hormonal and structural bone changes associated with bortezomib response in multiple myeloma patients. Haematologica. 2011;96:333–336.
  • Mohty M, Malard F, Mohty B, et al. The effects of bortezomib on bone disease in patients with multiple myeloma. Cancer. 2014;120:618–623.
  • Delforge M, Terpos E, Richardson PG, et al. Fewer bone disease events, improvement in bone remodeling, and evidence of bone healing with bortezomib plus melphalan-prednisone vs. melphalan-prednisone in the phase III VISTA trial in multiple myeloma. Eur J Haematol. 2011 May;86(5):372–384.
  • Terpos E, Christoulas D, Kastritis E, et al. VTD consolidation, without bisphosphonates, reduces bone resorption and is associated with a very low incidence of skeletal-related events in myeloma patients post ASCT. Leukemia. 2014;28:928–934.
  • Anderson G, Gries M, Kurihara N, et al. Thalidomide derivative CC-4047 inhibits osteoclast formation by down-regulation of PU.1. Blood. 2006;107:3098–3105.
  • Munemasa S, Sakai A, Kuroda Y, et al. Osteoprogenitor differentiation is not affected by immunomodulatory thalidomide analogs but is promoted by low bortezomib concentration, while both agents suppress osteoclast differentiation. Int J Oncol. 2008;33:129–136.
  • Breitkreutz I, Raab MS, Vallet S, et al. Lenalidomide inhibits osteoclastogenesis, survival factors and bone-remodeling markers in multiple myeloma. Leukemia. 2008;22:1925–1932.
  • Terpos E, Mihou D, Szydlo R, et al. The combination of intermediate doses of thalidomide with dexamethasone is an effective treatment for patients with refractory/relapsed multiple myeloma and normalizes abnormal bone remodeling, through the reduction of sRANKL/osteoprotegerin ratio. Leukemia. 2005;19:1969–1976.
  • Terpos E, Christoulas D, Kastritis E, et al. The combination of lenalidomide and dexamethasone reduces bone resorption in responding patients with relapsed/refractory multiple myeloma but has no effect on bone formation: final results on 205 patients of the Greek myeloma study group. Am J Hematol. 2014;89:34–40.
  • Santo L, Hideshima T, Kung AL, et al. Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma. Blood. 2012 Mar 15;119(11):2579–2589.
  • Alsayed Y, Ngo H, Runnels J, et al. Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma. Blood. 2007;109:2708–2717.
  • Beider K, Begin M, Abraham M, et al. CXCR4 antagonist 4F-benzoyl-TN14003 inhibits leukemia and multiple myeloma tumor growth. Exp Hematol. 2011;39:282–292.
  • Taylor RM, Kashima TG, Knowles HJ, et al. VEGF, FLT3 ligand, PlGF and HGF can substitute for M-CSF to induce human osteoclast formation: implications for giant cell tumour pathobiology. Lab Investig. 2012;92:1398–1400.
  • Tanaka Y, Abe M, Hiasa M, et al. Myeloma cell-osteoclast interaction enhances angiogenesis together with bone resorption: a role for vascular endothelial cell growth factor and osteopontin. Clin Cancer Res. 2007;13:816e823.
  • Fulciniti M, Hideshima T, Vermot-Desroches C. A high-affinity fully human anti-IL-6 mAb, 1339, for the treatment of multiple myeloma. Clin Cancer Res. 2009;15:7144e7152.
  • Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17:1231e1234.
  • Brunetti G, Rizzi R, Oranger A, et al. LIGHT/TNFSF14 increases osteoclastogenesis and decreases osteoblastogenesis in multiple myeloma-bone disease. Oncotarget. 2014;5:12950e12967.
  • Gopalakrishnan N, Saravanakumar M, Madankumar P, et al. Colocalization of beta-catenin with Notch intracellular domain in colon cancer: a possible role of Notch1 signaling in activation of CyclinD1-mediated cell proliferation. Mol Cell Biochem. 2014;396:281e293.
  • Delgado-Calle J, Anderson J, Cregor MD, et al. Bidirectional notch signaling and osteocyte-derived factors in the bone marrow microenvironment promote tumor cell proliferation and bone destruction in multiple myeloma. Cancer Res. 2016;76:1089e1100.
  • Lim HJ, Crowe P, Yang JL, et al. Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J Cancer Res Clin Oncol. 2015;141:671–689.
  • Glauer J, Pletz N, Schon M, et al. A novel selective small-molecule PI3K inhibitor is effective against human multiple myeloma in vitro and in vivo. Blood Cancer J. 2013;3:e141.
  • Gan ZY, Fitter S, Vandyke K, et al. The effect of the dual PI3K and mTOR inhibitor BEZ235 on tumour growth and osteolytic bone disease in multiple myeloma. Eur J Haematol. 2015;94:343e354.
  • Iwakura Y, Ishigame H, Saijo S, et al. Functional specialization of IL-17 family members. Immunity. 2011;34(2):149–162.
  • Prabhala RH, Pelluru D, Fulciniti M, et al. Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 2010;115(26):5385–5392.
  • Chabaud M, Garnero P, Dayer JM, et al. Contribution of interleukin 17 to synovium matrix destruction in rheumatoid arthritis. Cytokine. 2000;12(7):1092–1099.
  • Xu C, Yu L, Zhan P, et al. Elevated pleural effusion IL-17 is a diagnostic marker and outcome predictor in lung cancer patients. Eur J Med Res. 2014 May 8;19(1):23.
  • Prabhala RH, Fulciniti M, Pelluru D, et al. Targeting IL-17A in multiple myeloma. A potential novel therapeutic approach in myeloma. Leukemia. 2016 Feb;30(2):379–389.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.