REVIEW PAPER
A view at monoclonal antibodies in therapy of osteoporosis
 
More details
Hide details
1
Department of Pharmacodynamics, Medical University of Bialystok, Poland
 
2
Department of Monitored Pharmacotherapy, Medical University of Bialystok, Poland
 
 
Submission date: 2015-07-19
 
 
Acceptance date: 2015-07-31
 
 
Publication date: 2020-03-26
 
 
Corresponding author
Dariusz Pawlak   

Department of Pharmacodynamics, Medical University in Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland. Tel.: +48 857485601; fax: +48 857485601.
 
 
Pol. Ann. Med. 2015;22(2):149-154
 
KEYWORDS
ABSTRACT
Introduction:
Osteoporosis is a bone disease, which leads to increased fracture risk and weakens bone strength. Drugs used in current therapies of this disease are far from perfect thus the search for new effective compounds is an ongoing process, and some researchers put great hopes in monoclonal antibodies in this field.

Aim:
The purpose of this paper is to discuss monoclonal antibodies as potentially beneficial therapy of osteoporosis.

Material and methods:
It was based upon the available literature and publications.

Results and discussion:
Sclerostin is a glycoprotein that belongs to Wnt inhibitors. Wnt/β-catenin signaling pathway is essential for normal physiological cell functions such as differentiation or proliferation. Inhibition of sclerostin activity can result in increased bone mineral density, and can be achieved by using antibodies against this factor, i.e. romosozumab and blosozumab. Another compound that has an influence on Wnt/β-catenin signaling pathway is Dickkopf-1. Monoclonal antibodies against this factor have been tested in bone diseases and found to contribute to increased bone mineral density. Other antiresorptive agent indicated for the treatment of osteoporosis is a receptor activator of nuclear factor-kβ ligand (RANKL) inhibitor. Denosumab is a human antibody to RANKL, and it decreases osteoclastogenesis and osteoclast activity, leading to reduced bone resorption. It is currently used in treatment of postmenopausal osteoporosis.

Conclusions:
The essential goal for the management of osteoporosis is to increase bone mass and reduce fracture risk by slowing or stopping bone loss. Monoclonal antibodies that have been recently developed are becoming an important option in the pharmacotherapy of osteoporosis.

CONFLICT OF INTEREST
None declared.
 
REFERENCES (47)
1.
Powroznik B, Kubowicz P, Pekala E. Monoclonal antibodies in targeted therapy. Postepy Hig Med Dosw (Online). 2012;66:663–673 [in Polish].
 
2.
Cairoli E, Zhukouskaya VV, Eller-Vainicher C, Chiodini I. Perspectives on osteoporosis therapies. J Endocrinol Invest. 2015. http://dx.doi.org/10.1007/s406... [in press].
 
3.
Costa AG, Bilezikian JP. Sclerostin: therapeutic horizons based upon its actions. Curr Osteoporos Rep. 2012;10(1):64–72. http://dx.doi.org/10.1007/s119....
 
4.
Hernandez P, Whitty C, John Wardale R, Henson FM. New insights into the location and form of sclerostin. Biochem Biophys Res Commun. 2014;446(4):1108–1113.
 
5.
Ke HZ, Richards WG, Li X, Ominsky MS. Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocr Rev. 2012;33(5):747–783. http://dx.doi.org/10.1210/er.2....
 
6.
Compton JT, Lee FY. A review of osteocyte function and the emerging importance of sclerostin. J Bone Joint Surg Am. 2014;96(19):1659–1668.
 
7.
Ott SM. Bone cells, sclerostin, and FGF23: what's bred in the bone will come out in the flesh. Kidney Int. 2015;87(3):499–501. http://dx.doi.org/10.1038/ki.2....
 
8.
Weivoda MM, Oursler MJ. Developments in sclerostin biology: regulation of gene expression, mechanisms of action, and physiological functions. Curr Osteoporos Rep. 2014;12(1):107–114. http://dx.doi.org/10.1007/s119....
 
9.
Clarke BL. Anti-sclerostin antibodies: utility in treatment of osteoporosis. Maturitas. 2014;78(3):199–204. http://dx.doi.org/10.1016/j.ma....
 
10.
Garnero P. New developments in biological markers of bone metabolism in osteoporosis. Bone. 2014;66:46–55. http://dx.doi.org/10.1016/j.bo....
 
11.
Yavropoulou MP, Xygonakis C, Lolou M, Karadimou F, Yovos JG. The sclerostin story: from human genetics to the development of novel anabolic treatment for osteoporosis. Hormones (Athens). 2014;13(4):323–337. http://dx.doi.org/10.14310/hor....
 
12.
Brandenburg VM, D'Haese P, Deck A, et al. From skeletal to cardiovascular disease in 12 steps—the evolution of sclerostin as a major player in CKD-MBD. Pediatr Nephrol. 2015. http://dx.doi.org/10.1007/s004... [in press].
 
13.
Rossini M, Gatti D, Adami S. Involvement of WNT/b-catenin signaling in the treatment of osteoporosis. Calcif Tissue Int. 2013;93(2):121–132. http://dx.doi.org/10.1007/s002....
 
14.
Reichert JM. Antibodies to watch in 2015. MAbs. 2015;7(1):1–8. http://dx.doi.org/10.4161/1942....
 
15.
Jean G, Chazot C. Sclerostin in CKD-MBD: one more paradoxical bone protein? Nephrol Dial Transplant. 2013;28(12):2932–2935. http://dx.doi.org/10.1093/ndt/....
 
16.
Recker RR, Benson CT, Matsumoto T, et al. A randomized, double-blind phase 2 clinical trial of blosozumab, a sclerostin antibody, in postmenopausal women with low bone mineral density. J Bone Miner Res. 2015;30(2):216–224. http://dx.doi.org/10.1002/jbmr....
 
17.
Bhutani G, Gupta MC. Emerging therapies for the treatment of osteoporosis. J Midlife Health. 2013;4(3):147–152. http://dx.doi.org/10.4103/0976....
 
18.
Ng KW, Martin TJ. New therapeutics for osteoporosis. Curr Opin Pharmacol. 2014;16:58–63. http://dx.doi.org/10.1016/j.co....
 
19.
McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370(5):412–420.
 
20.
Padhi D, Allison M, Kivitz AJ, et al. Multiple doses of sclerostin antibody romosozumab in healthy men and postmenopausal women with low bone mass: a randomized, double-blind, placebo-controlled study. J Clin Pharmacol. 2014;54(2):168–178.
 
21.
Lim V, Clarke BL. New therapeutic targets for osteoporosis: beyond denosumab. Maturitas. 2012;73(3):269–272. http://dx.doi.org/10.1016/j.ma....
 
22.
O'Brien CA. Control of RANKL gene expression. Bone. 2010;46(4):911–919. http://dx.doi.org/10.1016/j.bo....
 
23.
Wong BR, Josien R, Lee SY, et al. TRANCE (tumor necrosis factor [TNF]-related activation-induced cytokine), a new TNF family member predominantly expressed in T cells, is a dendritic cell-specific survival factor. J Exp Med. 1997;186(12):2075–2080. http://dx.doi.org/10.1084/jem.....
 
24.
Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG system in immunity, bone, and beyond. Front Immunol. 2014;5:511. http://dx.doi.org/10.3389/fimm....
 
25.
Lum L, Wong BR, Josien R, et al. Evidence for a role of a tumor necrosis factor-alpha (TNF-alpha)-converting enzyme-like protease in shedding of TRANCE, a TNF family member involved in osteoclastogenesis and dendritic cell survival. J Biol Chem. 1999;274(19):13613–21368. http://dx.doi.org/10.1074/jbc.....
 
26.
Hikita A, Yana I, Wakeyama H, et al. Negative regulation of osteoclastogenesis by ectodomain shedding of receptor activator of NF-kappaB ligand. J Biol Chem. 2006;281(48):36846–43655. http://dx.doi.org/10.1074/jbc.....
 
27.
Li J, Sarosi I, Yan XQ, et al. RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci U S A. 2000;97(4):1566–1571. http://dx.doi.org/10.1073/pnas....
 
28.
Tat SK, Pelletier JP, Velasco CR, Padrines M, Martel-Pelletier J. New perspective in osteoarthritis: the OPG and RANKL system as a potential therapeutic target? Keio J Med. 2009;58(1):29–40. http://dx.doi.org/10.2302/kjm.....
 
29.
Romas E. Clinical application of RANK-ligand. Intern Med J. 2009;39(2):110–116. http://dx.doi.org/10.1111/j.14....
 
30.
Miller PD, Bolognese MA, Lewiecki EM, et al. Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after longterm continued, discontinued, and restarting of therapy: a randomized blinded phase 2 clinical trial. Bone. 2008;43(2):222–229. http://dx.doi.org/10.1016/j.bo....
 
31.
Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999;397(6717):315–323. http://dx.doi.org/10.1038/1685....
 
32.
Bucay N, Sarosi I, Dunstan CR, et al. Osteoprotegerin-deficient mice develop early inset osteoporosis and arterial calcification. Genes Dev. 1998;12(9):1260–1268. http://dx.doi.org/10.1101/gad.....
 
33.
Mizuno A, Amizuka N, Irie K, et al. Severe osteoporosis in mice lacking osteoclastogenesis inhibitory factor/osteoprotegerin. Biochem Biophys Res Commun. 1998;247(3):610–615. http://dx.doi.org/10.1006/bbrc....
 
34.
Suresh E, Abrahamsen B. Denosumab: a novel antiresorptive drug for osteoporosis. Cleve Clin J Med. 2015;82(2):105–114. http://dx.doi.org/10.3949/ccjm....
 
35.
Diab DL, Watts NB. Denosumab in osteoporosis. Expert Opin Drug Saf. 2014;13(2):247–253. http://dx.doi.org/10.1517/1474....
 
36.
Tella SH, Gallagher JC. Biological agents in management of osteoporosis. Eur J Clin Pharmacol. 2014;70(11):1291–1301. http://dx.doi.org/10.1007/s002....
 
37.
Amgen. Prolia® (denosumab). Thousand Oaks, CA: Amgen Inc.; 2010. http://pi.amgen.com/united_sta.... Accessed 16.2.2015.
 
38.
Choi HJ. New antiresorptive therapies for postmenopausal osteoporosis. J Menopausal Med. 2015;21(1):1–11. http://dx.doi.org/10.6118/jmm.....
 
39.
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(7):1059–1066. http://dx.doi.org/10.1359/JBMR....
 
40.
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(12):1832–1841. http://dx.doi.org/10.1359/jbmr....
 
41.
McClung MR, Lewiecki EM, Geller ML, et al. Effect of denosumab on bone mineral density and biochemical markers of bone turnover: 8-year results of phase 2 clinical trials. Osteoporos Int. 2013;24(1):227–235. http://dx.doi.org/10.1007/s001....
 
42.
Miller PD, Wagman RB, Peacock M, et al. Effect of denosumab on bone mineral density and biochemical markers of bone turnover: six-year results of a phase 2 clinical trial. J Clin Endocrinol Metab. 2011;96(2):394–402. http://dx.doi.org/10.1210/jc.2....
 
43.
McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006;354(8):821–831. http://dx.doi.org/10.1056/NEJM....
 
44.
Cummings SR, San Martin J, McClung MR, et al. FREEDOM Trial. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361(8):756–765. http://dx.doi.org/10.1056/NEJM....
 
45.
Brown JP, Prince RL, Deal C, et al. Comparison of the effect of denosumab and alendronate on BMD and biochemical markers of bone turnover in postmenopausal women with low bone mass: a randomized, blinded, phase 3 trial. J Bone Miner Res. 2009;24(1):153–161. http://dx.doi.org/10.1359/jbmr....
 
46.
Kendler DL, Roux C, Benhamou CL, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res. 2010;25(1):72–81. http://dx.doi.org/10.1359/jbmr....
 
47.
Reid IR, Miller PD, Brown JP, et al. Effects of denosumab on bone histomorphometry: the FREEDOM and STAND studies. J Bone Miner Res. 2010;25(10):2256–2265. http://dx.doi.org/10.1002/jbmr....
 
Journals System - logo
Scroll to top