Treatment of Osteoporosis, with a Focus on 2 Monoclonal Antibodies

Biao Chang, Qi Quan, Yunqi Li, Haixia Qiu, Jiang Peng, Ying Gu, Biao Chang, Qi Quan, Yunqi Li, Haixia Qiu, Jiang Peng, Ying Gu

Abstract

Osteoporosis is a common skeletal disease characterized by bone loss and subsequent increased risk of fragility fractures. Recent advances in our mechanistic understanding of molecular communications among osteoblasts, osteoclasts, and osteocytes give insight into the important roles of the canonical Wnt/β-catenin pathway and the RANK/RANKL/OPG pathway in the process of bone remodeling. Due to the translation of the canonical Wnt/β-catenin pathway and the RANK/RANKL/OPG pathway in the regulation of osteoblasts and osteoclasts, new targets have been studied in recent years, such as sclerostin and receptor activator of NF-κB ligand (RANKL). In this review, we first introduce the signaling pathways involved in interactions among osteoblasts, osteoclasts, and osteocytes. Next, we describe clinical trials of denosumab and romosozumab, which are monoclonal antibodies that target RANKL and sclerostin, respectively. We analyze the efficacy of these drugs and provide a profile for the management of osteoporosis.

Conflict of interest statement

Conflict of interest

None.

Figures

Figure 1
Figure 1
The signaling pathways involved in bone remodeling. Dkk-1 – Dickkopf-related protein 1; OPG – osteoprotegerin; PTH – parathyroid hormone; RANK – receptor activator of NF-κB; RANKL – receptor activator of NF-κB ligand.

References

    1. Siris ES, Miller PD, Barrett-Connor E, et al. Identification and fracture outcomes of undiagnosed low bone mineral density in postmenopausal women: Results from the National Osteoporosis Risk Assessment. JAMA. 2001;286:2815–22.
    1. Kanis JA, McCloskey EV, Johansson H, et al. European guidance for the diagnosis and management of osteoporosis in postmenopausal women. Osteoporos Int. 2013;24:23–57.
    1. Qaseem A, Forciea M, McLean R, Denberg T. Treatment of low bone density or osteoporosis to prevent fractures in men and women: A clinical practice guideline update from the American College of Physicians. Ann Intern Med. 2017;166:818–39.
    1. Cotts K, Cifu AS. Treatment of osteoporosis. JAMA. 2018;319:1040–41.
    1. Ensrud KE, Crandall CJ. Osteoporosis. Ann Intern Med. 2017;167:ITC17–32.
    1. Rachner TD, Khosla S, Hofbauer LC. Osteoporosis: Now and the future. Lancet. 2011;377:1276–87.
    1. Cosman F, de Beur SJ, LeBoff MS, et al. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Osteoporos Int. 2014;25:2359–81.
    1. McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006;354:821–31.
    1. Leder BZ, Tsai JN, Uihlein AV, et al. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA-Switch study): Extension of a randomised controlled trial. Lancet. 2015;386:1147–55.
    1. Bone HG, Wagman RB, Brandi ML, et al. 10 years of denosumab treatment in postmenopausal women with osteoporosis: Results from the phase 3 randomised FREEDOM trial and open-label extension. Lancet Diabetes Endocrinol. 2017;5:513–23.
    1. McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412–20.
    1. Cosman F, Crittenden DB, Adachi JD, et al. Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med. 2016;375:1532–43.
    1. van Bezooijen RL, Roelen BA, Visser A, et al. Sclerostin is an osteocyte-expressed negative regulator of bone formation, but not a classical BMP antagonist. J Exp Med. 2004;199:805–14.
    1. Poole KE, van Bezooijen RL, Loveridge N, et al. Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation. FASEB J. 2005;19:1842–44.
    1. Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 2011;17:1231–34.
    1. Capulli M, Paone R, Rucci N. Osteoblast and osteocyte: Games without frontiers. Arch Biochem Biophys. 2014;561:3–12.
    1. Canalis E. Wnt signalling in osteoporosis: mechanisms and novel therapeutic approaches. Nat Rev Endocrinol. 2013;9:575–83.
    1. Nagy V, Penninger JM. The RANKL-RANK story. Gerontology. 2015;61:534–42.
    1. Baron R, Rawadi G. Targeting the Wnt/beta-catenin pathway to regulate bone formation in the adult skeleton. Endocrinology. 2007;148:2635–43.
    1. Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005;280:19883–87.
    1. Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling pathway. J Cell Sci. 2003;116:2627–34.
    1. Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, et al. Wnt/beta-catenin signaling is a normal physiological response to mechanical loading in bone. J Biol Chem. 2006;281:31720–28.
    1. Robling AG, Niziolek PJ, Baldridge LA, et al. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem. 2008;283:5866–75.
    1. Tu X, Rhee Y, Condon KW, et al. Sost downregulation and local Wnt signaling are required for the osteogenic response to mechanical loading. Bone. 2012;50:209–17.
    1. Brunkow ME, Gardner JC, Van Ness J, et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am J Hum Genet. 2001;68:577–89.
    1. Li X, Ominsky MS, Niu QT, et al. Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J Bone Miner Res. 2008;23:860–69.
    1. 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;24:578–88.
    1. Ominsky MS, Vlasseros F, Jolette J, et al. Two doses of sclerostin antibody in cynomolgus monkeys increases bone formation, bone mineral density, and bone strength. J Bone Miner Res. 2010;25:948–59.
    1. Glass DA, 2nd, Bialek P, Ahn JD, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005;8:751–64.
    1. Otero K, Shinohara M, Zhao H, et al. TREM2 and β-catenin regulate bone homeostasis by controlling the rate of osteoclastogenesis. J Immunol. 2012;188:2612–21.
    1. Yasuda H, Shima N, Nakagawa N, et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci USA. 1998;95:3597–602.
    1. Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397:315–23.
    1. Lacey DL, Timms E, Tan HL, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998;93:165–76.
    1. Lacey D, Tan H, Lu J, et al. Osteoprotegerin ligand modulates murine osteoclast survival in vitro and in vivo. Am J Pathol. 2000;157:435–48.
    1. Simonet W, Lacey D, Dunstan C, et al. Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell. 1997;89:309–19.
    1. Bucay N, Sarosi I, Dunstan C, et al. osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev. 1998;12:1260–68.
    1. Capparelli C, Morony S, Warmington K, et al. Sustained antiresorptive effects after a single treatment with human recombinant osteoprotegerin (OPG): A pharmacodynamic and pharmacokinetic analysis in rats. J Bone Miner Res. 2003;18:852–58.
    1. Kostenuik P, Bolon B, Morony S, et al. Gene therapy with human recombinant osteoprotegerin reverses established osteopenia in ovariectomized mice. Bone. 2004;34:656–64.
    1. Eghbali-Fatourechi G, Khosla S, Sanyal A, et al. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest. 2003;111:1221–30.
    1. Bekker P, Holloway D, Nakanishi A, et al. The effect of a single dose of osteoprotegerin in postmenopausal women. J Bone Miner Res. 2001;16:348–60.
    1. Fisher JE, Rodan GA, Reszka AA. In vivo effects of bisphosphonates on the osteoclast mevalonate pathway. Endocrinology. 2000;141:4793–96.
    1. Martins CA, Leyhausen G, Volk J, Geurtsen W. Effects of alendronate on osteoclast formation and activity in vitro. J Endodont. 2015;41:45–49.
    1. Black DM, Cummings SR, Karpf DB, et al. Randomised trial of effect of alendronate on risk of fracture in women with existing vertebral fractures. Fracture Intervention Trial Research Group. Lancet. 1996;348:1535–41.
    1. Rizzoli R, Greenspan SL, Bone G, 3rd, et al. Two-year results of once-weekly administration of alendronate 70 mg for the treatment of postmenopausal osteoporosis. J Bone Miner Res. 2002;17:1988–96.
    1. Yan Y, Wang W, Zhu H, et al. The efficacy and tolerability of once-weekly alendronate 70 mg on bone mineral density and bone turnover markers in postmenopausal Chinese women with osteoporosis. J Bone Miner Metab. 2009;27:471–78.
    1. Liberman UA, Weiss SR, Broll J, et al. Effect of oral alendronate on bone mineral density and the incidence of fractures in postmenopausal osteoporosis. The Alendronate Phase III Osteoporosis Treatment Study Group. N Engl J Med. 1995;333:1437–43.
    1. Bone HG, Hosking D, Devogelaer JP, et al. Ten years’ experience with alendronate for osteoporosis in postmenopausal women. N Engl J Med. 2004;350:1189–99.
    1. Chesnut CH, 3rd, Skag A, Christiansen C, et al. Effects of oral ibandronate administered daily or intermittently on fracture risk in postmenopausal osteoporosis. J Bone Miner Res. 2004;19:1241–49.
    1. Reginster J, Minne HW, Sorensen OH, et al. Randomized trial of the effects of risedronate on vertebral fractures in women with established postmenopausal osteoporosis. Vertebral Efficacy with Risedronate Therapy (VERT) Study Group. Osteoporos Int. 2000;11:83–91.
    1. Harris ST, Watts NB, Genant HK, et al. Effects of risedronate treatment on vertebral and nonvertebral fractures in women with postmenopausal osteoporosis: A randomized controlled trial. Vertebral Efficacy With Risedronate Therapy (VERT) Study Group. JAMA. 1999;282:1344–52.
    1. Black DM, Schwartz AV, Ensrud KE, et al. Effects of continuing or stopping alendronate after 5 years of treatment: the Fracture Intervention Trial Long-term Extension (FLEX): A randomized trial. JAMA. 2006;296:2927–38.
    1. Bauer DC, Schwartz A, Palermo L, et al. Fracture prediction after discontinuation of 4 to 5 years of alendronate therapy: The FLEX study. JAMA Intern Med. 2014;174:1126–34.
    1. Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med. 2007;356:1809–22.
    1. Eriksen EF, Diez-Perez A, Boonen S. Update on long-term treatment with bisphosphonates for postmenopausal osteoporosis: A systematic review. Bone. 2014;58:126–35.
    1. Boonen S, Reginster JY, Kaufman JM, et al. Fracture risk and zoledronic acid therapy in men with osteoporosis. N Engl J Med. 2012;367:1714–23.
    1. Greenspan SL, Perera S, Ferchak MA, et al. Efficacy and safety of single-dose zoledronic acid for osteoporosis in frail elderly women: A randomized clinical trial. JAMA Intern Med. 2015;175:913–21.
    1. Reid IR, Brown JP, Burckhardt P, et al. Intravenous zoledronic acid in postmenopausal women with low bone mineral density. N Engl J Med. 2002;346:653–61.
    1. Hue TF, Cummings SR, Cauley JA, et al. Effect of bisphosphonate use on risk of postmenopausal breast cancer: results from the randomized clinical trials of alendronate and zoledronic acid. JAMA Intern Med. 2014;174:1550–57.
    1. Lindsay R, Nieves J, Formica C, et al. Randomised controlled study of effect of parathyroid hormone on vertebral-bone mass and fracture incidence among postmenopausal women on oestrogen with osteoporosis. Lancet. 1997;350:550–55.
    1. Neer R, Arnaud C, Zanchetta J, et al. Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med. 2001;344:1434–41.
    1. Orwoll E, Scheele W, Paul S, et al. The effect of teriparatide human parathyroid hormone (1–34) therapy on bone density in men with osteoporosis. J Bone Miner Res. 2003;18:9–17.
    1. Boonen S, Marin F, Obermayer-Pietsch B, et al. Effects of previous antiresorptive therapy on the bone mineral density response to two years of teriparatide treatment in postmenopausal women with osteoporosis. J Clin Endocrinol Metab. 2008;93:852–60.
    1. Obermayer-Pietsch BM, Marin F, McCloskey EV, et al. Effects of two years of daily teriparatide treatment on BMD in postmenopausal women with severe osteoporosis with and without prior antiresorptive treatment. J Bone Miner Res. 2008;23:1591–600.
    1. Hodsman AB, Bauer DC, Dempster DW, et al. Parathyroid hormone and teriparatide for the treatment of osteoporosis: A review of the evidence and suggested guidelines for its use. Endocr Rev. 2005;26:688–703.
    1. Rittmaster R, Bolognese M, Ettinger M, et al. Enhancement of bone mass in osteoporotic women with parathyroid hormone followed by alendronate. J Clin Endocrinol Metab. 2000;85:2129–34.
    1. Black DM, Greenspan SL, Ensrud KE, et al. The effects of parathyroid hormone and alendronate alone or in combination in postmenopausal osteoporosis. N Engl J Med. 2003;349:1207–15.
    1. Finkelstein JS, Hayes A, Hunzelman JL, et al. The effects of parathyroid hormone, alendronate, or both in men with osteoporosis. N Engl J Med. 2003;349:1216–26.
    1. Black D, Bilezikian J, Ensrud K, et al. One year of alendronate after one year of parathyroid hormone (1–84) for osteoporosis. N Engl J Med. 2005;353:555–65.
    1. Tsai JN, Uihlein AV, Lee H, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet. 2013;382:50–56.
    1. Leder BZ, Tsai JN, Uihlein AV, et al. Two years of Denosumab and teriparatide administration in postmenopausal women with osteoporosis (The DATA Extension Study): A randomized controlled trial. J Clin Endocrinol Metab. 2014;99:1694–700.
    1. Genant HK, Engelke K, Bolognese MA, et al. Effects of romosozumab compared with teriparatide on bone density and mass at the spine and hip in postmenopausal women with low bone mass. J Bone Miner Res. 2017;32:181–87.
    1. Langdahl B, Libanati C, Crittenden D, et al. Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial. Lancet. 2017;390:1585–94.
    1. Kendler D, Marin F, Zerbini C, et al. Effects of teriparatide and risedronate on new fractures in post-menopausal women with severe osteoporosis (VERO): A multicentre, double-blind, double-dummy, randomised controlled trial. Lancet. 2017 [Epub ahead of print]
    1. Prince R, Sipos A, Hossain A, et al. Sustained nonvertebral fragility fracture risk reduction after discontinuation of teriparatide treatment. J Bone Miner Res. 2005;20:1507–13.
    1. Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results From the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321–33.
    1. Komm BS, Morgenstern D, A Yamamoto L, Jenkins SN. The safety and tolerability profile of therapies for the prevention and treatment of osteoporosis in postmenopausal women. Expert Rev Clin Pharmacol. 2015;8:769–84.
    1. Langer R, Manson J, Allison M. Have we come full circle – or moved forward? The Women’s Health Initiative 10 years on. Climacteric. 2012;15:206–12.
    1. Bekker PJ, Holloway DL, Rasmussen AS, et al. A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res. 2004;19:1059–66.
    1. Lewiecki EM, Miller PD, McClung MR, et al. Two-year treatment with denosumab (AMG 162) in a randomized phase 2 study of postmenopausal women with low BMD. J Bone Miner Res. 2007;22:1832–41.
    1. Papapoulos S, Chapurlat R, Libanati C, et al. Five years of denosumab exposure in women with postmenopausal osteoporosis: results from the first two years of the FREEDOM extension. J Bone Miner Res. 2012;27:694–701.
    1. Cummings S, San Martin J, McClung M, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361:756–65.
    1. Bone HG, Chapurlat R, Brandi ML, et al. The effect of three or six years of denosumab exposure in women with postmenopausal osteoporosis: Results from the FREEDOM extension. J Clin Endocrinol Metab. 2013;98:4483–92.
    1. Papapoulos S, Lippuner K, Roux C, et al. The effect of 8 or 5 years of denosumab treatment in postmenopausal women with osteoporosis: Results from the FREEDOM Extension study. Osteoporos Int. 2015;26:2773–83.
    1. 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.
    1. Saag K, Petersen J, Brandi M, et al. Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med. 2017;377:1417–27.
    1. Evenepoel P, D’Haese P, Brandenburg V. Romosozumab in postmenopausal women with osteopenia. N Eng J Med. 2014;370:1664.
    1. Ma YL, Zeng QQ, Chiang AY, et al. Effects of teriparatide on cortical histomorphometric variables in postmenopausal women with or without prior alendronate treatment. Bone. 2014;59:139–47.
    1. Tsai J, Uihlein A, Burnett-Bowie S, et al. Comparative effects of teriparatide, denosumab, and combination therapy on peripheral compartmental bone density, microarchitecture, and estimated strength: the DATA-HRpQCT Study. J Bone Miner Res. 2015;30:39–45.
    1. Macdonald H, Nishiyama K, Hanley D, Boyd S. Changes in trabecular and cortical bone microarchitecture at peripheral sites associated with 18 months of teriparatide therapy in postmenopausal women with osteoporosis. Osteoporos Int. 2011;22:357–62.
    1. Lewis J, Schousboe J, Prince R. Romosozumab versus alendronate and fracture risk in women with osteoporosis. N Engl J Med. 2018;378:194–95.
    1. Saag K, Petersen J, Grauer A. Romosozumab versus alendronate and fracture risk in women with osteoporosis. N Engl J Med. 2018;378:195–96.
    1. Tsourdi E, Rachner T, Hofbauer L. Romosozumab versus alendronate and fracture risk in women with osteoporosis. N Engl J Med. 2018;378:195.
    1. He X, Wang E, Bao Y, et al. High serum levels of sclerostin and Dickkopf-1 are associated with acute ischaemic stroke. Atherosclerosis. 2016;253:22–28.
    1. Song G, Lee Y. Romosozumab versus alendronate and fracture risk in women with osteoporosis. N Engl J Med. 2018;378:194.
    1. Hirsch C. In postmenopausal women with osteoporosis, romosozumab followed by alendronate reduced fractures vs. alendronate alone. Ann Intern Med. 2018;168:JC3.
    1. Eastell R, Nickelsen T, Marin F, et al. Sequential treatment of severe postmenopausal osteoporosis after teriparatide: final results of the randomized, controlled European Study of Forsteo (EUROFORS) J Bone Miner Res. 2009;24:726–36.
    1. Cosman F, Eriksen E, Recknor C, et al. Effects of intravenous zoledronic acid plus subcutaneous teriparatide [rhPTH(1–34)] in postmenopausal osteoporosis. J Bone Miner Res. 2011;26:503–11.
    1. Finkelstein JS, Wyland JJ, Lee H, Neer RM. Effects of teriparatide, alendronate, or both in women with postmenopausal osteoporosis. J Clin Endocrinol Metab. 2010;95:1838–45.
    1. Hofbauer LC, Rachner TD. More DATA to guide sequential osteoporosis therapy. Lancet. 2015;386:1116–18.

Source: PubMed

3
订阅