Korean J Pain.  2017 Apr;30(2):86-92. 10.3344/kjp.2017.30.2.86.

Can denosumab be a substitute, competitor, or complement to bisphosphonates?

Affiliations
  • 1Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea. pain@pusan.ac.kr
  • 2Department of Orthopedics, Ludwig-Maximilian-University Munich, Grosshadern Campus, Munich, Germany.

Abstract

Osteoblasts, originating from mesenchymal cells, make the receptor activator of the nuclear factor kappa B ligand (RANKL) and osteoprotegerin (OPG) in order to control differentiation of activated osteoclasts, originating from hematopoietic stem cells. When the RANKL binds to the RANK of the pre-osteoclasts or mature osteoclasts, bone resorption increases. On the contrary, when OPG binds to the RANK, bone resorption decreases. Denosumab (AMG 162), like OPG (a decoy receptor), binds to the RANKL, and reduces binding between the RANK and the RANKL resulting in inhibition of osteoclastogenesis and reduction of bone resorption. Bisphosphonates (BPs), which bind to the bone mineral and occupy the site of resorption performed by activated osteoclasts, are still the drugs of choice to prevent and treat osteoporosis. The merits of denosumab are reversibility targeting the RANKL, lack of adverse gastrointestinal events, improved adherence due to convenient biannual subcutaneous administration, and potential use with impaired renal function. The known adverse reactions are musculoskeletal pain, increased infections with adverse dermatologic reactions, osteonecrosis of the jaw, hypersensitivity reaction, and hypocalcemia. Treatment with 60 mg of denosumab reduces the bone resorption marker, serum type 1 C-telopeptide, by 3 days, with maximum reduction occurring by 1 month. The mean time to maximum denosumab concentration is 10 days with a mean half-life of 25.4 days. In conclusion, the convenient biannual subcutaneous administration of 60 mg of denosumab can be considered as a first-line treatment for osteoporosis in cases of low compliance with BPs due to gastrointestinal trouble and impaired renal function.

Keyword

Bisphosphonates; Bone mineral density; Bone resorption; Denosumab; Hypocalcemia; Monoclonal antibodies; Osteoclast; Osteoporosis; Osteoprotegerin; RANK ligand

MeSH Terms

Antibodies, Monoclonal
Biomarkers
Bone Density
Bone Resorption
Compliance
Denosumab*
Diphosphonates*
Half-Life
Hematopoietic Stem Cells
Hypersensitivity
Hypocalcemia
Jaw
Miners
Musculoskeletal Pain
NF-kappa B
Osteoblasts
Osteoclasts
Osteonecrosis
Osteoporosis
Osteoprotegerin
RANK Ligand
Antibodies, Monoclonal
Biomarkers
Denosumab
Diphosphonates
NF-kappa B
Osteoprotegerin
RANK Ligand

Figure

  • Fig. 1 Bone remodeling cycle and medications for the treatment of osteoporosis. Bone remodeling cycle consists of at least 6 different phases: 1. resting, 2. activation, 3. resorption, 4. reversal, 5. formation, and 6. mineralization. Drugs for the treatment or prevention of osteoporosis act on different phases. In the activation phase, denosumab, like OPG (a decoy receptor produced by osteoblasts), binds the RANKL and blocks the RANKL from binding to the RANK, inhibits the differentiation steps from pre-osteoblasts via mature osteoclasts to activated osteoclasts, and finally reduces bone resorption. In the resorption phase, bisphosphonates bind to the bone mineral and take the space where activated osteoclasts attach at sites of bone resorption. Odanacatib, a cathepsin K inhibitor, inhibits the osteoclastic enzyme that degrades collagens. Saracatinib, a c-src inhibitor, inhibits osteoclastic activation. Selective estrogen receptor modulators (SERMs) and hormone (estrogen) replacement therapy interfere with various osteoblast-derived factors that stimulate osteoclasts. In the formation phase, strontium ranelate stimulates pre-osteoblasts to differentiate into osteoblasts, and stimulates osteoblasts to secrete OPG in order to prevent pre-osteoclasts from becoming activated osteoclasts via mature osteoclasts, as well. Parathyroid hormone (PTH) analogues and PTH-related protein (PTHrP) analogues increase the number and activity of osteoblasts. Romosozumab (AMG 785) and blosozumab, anti-sclerostin monoclonal antibodies, bind to the sclerostin (a glycoprotein inhibitor of osteoblast Wnt signaling produced by osteocytes) and inhibit its action. Cbl: Casitas B-lineage lymphoma, FAK: focal adhesion kinase, OPG: osteoprotegerin, PI3k: phosphoinositide 3-kinase, PTH: parathyroid hormone, PTHrP: PTH-related protein, RANK: the nuclear factor kappa B, RANKL: RANK ligand, SERMs: selective estrogen receptor modulators, Src: Src family kinase (a group of non-receptor tyrosine kinases). Modified from Connelly D. Osteoporosis: moving beyond bisphosphonates. Pharmaceutical Journal 2016 Nov [2016 Nov 23]. Available at http://www.pharmaceutical-journal.com/news-and-analysis/infographics/osteoporosis-moving-beyond-bisphosphonates/20201978.article


Cited by  1 articles

Medications for osteoporotic pain
Sang Wook Shin
Korean J Pain. 2017;30(2):85-85.    doi: 10.3344/kjp.2017.30.2.85.


Reference

1. Cosman F, de Beur SJ, LeBoff MS, Lewiecki EM, Tanner B, Randall S, et al. Clinician's guide to prevention and treatment of osteoporosis. Osteoporos Int. 2014; 25:2359–2381. PMID: 25182228.
Article
2. Drake MT, Clarke BL, Khosla S. Bisphosphonates: mechanism of action and role in clinical practice. Mayo Clin Proc. 2008; 83:1032–1045. PMID: 18775204.
Article
3. Baron R, Ferrari S, Russell RG. Denosumab and bisphosphonates: different mechanisms of action and effects. Bone. 2011; 48:677–692. PMID: 21145999.
Article
4. McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, et al. Denosumab in postmenopausal women with low bone mineral density. N Engl J Med. 2006; 354:821–831. PMID: 16495394.
Article
5. Cheung AM, Frame H, Ho M, Mackinnon ES, Brown JP. Bone strength and management of postmenopausal fracture risk with antiresorptive therapies: considerations for women's health practice. Int J Womens Health. 2016; 8:537–547. PMID: 27729815.
6. Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009; 361:756–765. PMID: 19671655.
Article
7. Khosla S. Increasing options for the treatment of osteoporosis. N Engl J Med. 2009; 361:818–820. PMID: 19671654.
Article
8. Rizzoli R, Yasothan U, Kirkpatrick P. Denosumab. Nat Rev Drug Discov. 2010; 9:591–592. PMID: 20671758.
Article
9. Zaheer S, LeBoff M, Lewiecki EM. Denosumab for the treatment of osteoporosis. Expert Opin Drug Metab Toxicol. 2015; 11:461–470. PMID: 25614274.
Article
10. Janjan N. Bone metastases: approaches to management. Semin Oncol. 2001; 28:28–34.
Article
11. Weinfurt KP, Li Y, Castel LD, Saad F, Timbie JW, Glendenning GA, et al. The significance of skeletal-related events for the health-related quality of life of patients with metastatic prostate cancer. Ann Oncol. 2005; 16:579–584. PMID: 15734776.
Article
12. Yong M, Jensen AÖ, Jacobsen JB, Nørgaard M, Fryzek JP, Sørensen HT. Survival in breast cancer patients with bone metastases and skeletal-related events: a population-based cohort study in Denmark (1999-2007). Breast Cancer Res Treat. 2011; 129:495–503. PMID: 21461730.
Article
13. Cassinello Espinosa J, González Del Alba Baamonde A, Rivera Herrero F, Holgado Martín E. SEOM (Spanish Society of Clinical Oncology). SEOM guidelines for the treatment of bone metastases from solid tumours. Clin Transl Oncol. 2012; 14:505–511. PMID: 22721794.
Article
14. von Moos R, Costa L, Ripamonti CI, Niepel D, Santini D. Improving quality of life in patients with advanced cancer: targeting metastatic bone pain. Eur J Cancer. 2017; 71:80–94. PMID: 27984770.
Article
15. Raggatt LJ, Partridge NC. Cellular and molecular mechanisms of bone remodeling. J Biol Chem. 2010; 285:25103–25108. PMID: 20501658.
Article
16. Proff P, Römer P. The molecular mechanism behind bone remodelling: a review. Clin Oral Investig. 2009; 13:355–362.
Article
17. Ghayor C, Weber FE. Epigenetic regulation of bone remodeling and its impacts in osteoporosis. Int J Mol Sci. 2016; 17:E1446. PMID: 27598138.
Article
18. Crockett JC, Rogers MJ, Coxon FP, Hocking LJ, Helfrich MH. Bone remodelling at a glance. J Cell Sci. 2011; 124:991–998. PMID: 21402872.
Article
19. Miller SC, de Saint-Georges L, Bowman BM, Jee WS. Bone lining cells: structure and function. Scanning Microsc. 1989; 3:953–960. PMID: 2694361.
20. Hanley DA, Adachi JD, Bell A, Brown V. Denosumab: mechanism of action and clinical outcomes. Int J Clin Pract. 2012; 66:1139–1146. PMID: 22967310.
Article
21. Xing L, Xiu Y, Boyce BF. Osteoclast fusion and regulation by RANKL-dependent and independent factors. World J Orthop. 2012; 3:212–222. PMID: 23362465.
Article
22. Martin TJ. Paracrine regulation of osteoclast formation and activity: milestones in discovery. J Musculoskelet Neuronal Interact. 2004; 4:243–253. PMID: 15615492.
23. Clarke BL. Anti-sclerostin antibodies: utility in treatment of osteoporosis. Maturitas. 2014; 78:199–204. PMID: 24842796.
Article
24. Boivin G, Farlay D, Bala Y, Doublier A, Meunier PJ, Delmas PD. Influence of remodeling on the mineralization of bone tissue. Osteoporos Int. 2009; 20:1023–1026. PMID: 19340504.
Article
25. Bone HG, Bolognese MA, Yuen CK, Kendler DL, Wang H, Liu Y, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women. J Clin Endocrinol Metab. 2008; 93:2149–2157. PMID: 18381571.
Article
26. Sohn W, Simiens MA, Jaeger K, Hutton S, Jang G. The pharmacokinetics and pharmacodynamics of denosumab in patients with advanced solid tumours and bone metastases: a systematic review. Br J Clin Pharmacol. 2014; 78:477–487. PMID: 24548274.
Article
27. Miller PD. A review of the efficacy and safety of denosumab in postmenopausal women with osteoporosis. Ther Adv Musculoskelet Dis. 2011; 3:271–282. PMID: 22870485.
Article
28. Martin M, Bell R, Bourgeois H, Brufsky A, Diel I, Eniu A, et al. Bone-related complications and quality of life in advanced breast cancer: results from a randomized phase III trial of denosumab versus zoledronic acid. Clin Cancer Res. 2012; 18:4841–4849. PMID: 22893628.
Article
29. Kendler DL, Roux C, Benhamou CL, Brown JP, Lillestol M, Siddhanti S, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res. 2010; 25:72–81. PMID: 19594293.
Article
30. Fizazi K, Carducci M, Smith M, Damião R, Brown J, Karsh L, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011; 377:813–822. PMID: 21353695.
Article
31. Anastasilakis AD, Polyzos SA, Anastasilakis CD, Toulis KA, Makras P. Denosumab and bisphosphonates: rivals or potential “partners”? A “hybrid” molecule hypothesis. Med Hypotheses. 2011; 77:109–111. PMID: 21482033.
Article
32. Chiu YG, Ritchlin CT. Denosumab: targeting the RANKL pathway to treat rheumatoid arthritis. Expert Opin Biol Ther. 2017; 17:119–128. PMID: 27871200.
Article
33. Tsai JN, Uihlein AV, Lee H, Kumbhani R, Siwila-Sackman E, McKay EA, et al. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet. 2013; 382:50–56. PMID: 23683600.
Article
Full Text Links
  • KJP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr