Skip to main content
Log in

Population Pharmacokinetic Meta-Analysis of Denosumab in Healthy Subjects and Postmenopausal Women with Osteopenia or Osteoporosis

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objective: Inhibition of the receptor activator of nuclear factor k-B ligand (RANKL) is a therapeutic target for treatment of bone disorders associated with increased bone resorption, such as osteoporosis. The objective of this analysis was to characterize the population pharmacokinetics of denosumab (AMG 162; Prolia®), a fully human IgG2 monoclonal antibody that binds to RANKL, in healthy subjects and postmenopausal women with osteopenia or osteoporosis.

Methods: A total of 22944 serum free denosumab concentrations from 495 healthy subjects and 1069 post-menopausal women with osteopenia or osteoporosis were pooled. Denosumab was administered as either a single intravenous dose (n = 36), a single subcutaneous dose (n = 469) or multiple subcutaneous doses (n= 1059), ranging from 0.01 to 3 mg/kg (or 6–210 mg as fixed mass dosages), every 3 or 6 months for up to 48 months. An open, two-compartment pharmacokinetic model with a quasi-steady-state approximation of the target-mediated drug disposition model was used to describe denosumab pharmacokinetics, using NONMEM Version 7.1.0 software. Subcutaneous absorption was characterized by the first-order absorption rate constant (ka), with constant absolute bioavailability over the range of doses that were evaluated. Clearance and volume of distribution parameters were scaled by body weight, using a power model. Model evaluation was performed through visual predictive checks.

Results: The subcutaneous bioavailability of denosumab was 64%, and the ka was 0.00883 h−1. The central volume of distribution and linear clearance were 2.49 L/66 kg and 3.06 mL/h/66 kg, respectively. The baseline RANKL level, quasi-steady-state constant and RANKL degradation rate were 614ng/mL, 138 ng/mL and 0.00148 h−1, respectively. Between-subject variability in model parameters was moderate. A fixed dose of 60 mg provided RANKL inhibition similar to that achieved by equivalent body weight-based dosing. The effects of age and race on the area under the serum concentration-time curve of denosumab were less than 15% over the range of covariate values that were evaluated.

Conclusions: The non-linearity in denosumab pharmacokinetics is probably due to RANKL binding, and denosumab dose adjustment based on the patient demographics is not warranted.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Table I
Fig. 1
Table II
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Anderson DM, Maraskovsky E, Billingsley WL, et al. A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 1997; 390: 175–9

    Article  PubMed  CAS  Google Scholar 

  2. Lacey DL, Timms E, Tan HL, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998; 93: 165–76

    Article  PubMed  CAS  Google Scholar 

  3. 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 U S A 1998; 95: 3597–602

    Article  PubMed  CAS  Google Scholar 

  4. Burgess TL, Qian Y, Kaufman S, et al. The ligand for osteoprotegerin (OPGL) directly activates mature osteoclasts. J Cell Biol 1999; 145: 527–38

    Article  PubMed  CAS  Google Scholar 

  5. Kostenuik PJ. Osteoprotegerin and RANKL regulate bone resorption, density, geometry and strength. Curr Opin Pharmacol 2005; 5: 618–25

    Article  PubMed  CAS  Google Scholar 

  6. Bucay N, Sarosi I, Dunstan CR, et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998; 12: 1260–8

    Article  PubMed  CAS  Google Scholar 

  7. Bekker PJ, 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

    Article  PubMed  CAS  Google Scholar 

  8. Kostenuik PJ, Shalhoub V. Osteoprotegerin: a physiological and pharmacological inhibitor of bone resorption. Curr Pharm Des 2001; 7: 613–35

    Article  PubMed  CAS  Google Scholar 

  9. Elliott R, Kostenuik PJ, Chen C, et al. Denosumab is a selective inhibitor of human receptor activator of NF-KB ligand (RANKL) that blocks osteoclast formation and function [abstract]. Osteoporos Int 2007; 18: S54

    Google Scholar 

  10. Simonet WS, Lacey DL, Dunstan CR, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997; 89: 309–19

    Article  PubMed  CAS  Google Scholar 

  11. Akatsu T, Murakami T, Nishikawa M, et al. Osteoclastogenesis inhibitory factor suppresses osteoclast survival by interfering in the interaction of stromal cells with osteoclast. Biochem Biophys Res Commun 1998; 250: 229–34

    Article  PubMed  CAS  Google Scholar 

  12. Nakashima T, Kobayashi Y, Yamasaki S, et al. Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-kappaB ligand: modulation of the expression by osteotropic factors and cytokines. Biochem Biophys Res Commun 2000; 275: 768–75

    Article  PubMed  CAS  Google Scholar 

  13. Cummings SR, San MJ, McClung MR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 2009; 361: 756–65

    Article  PubMed  CAS  Google Scholar 

  14. Bekker PJ, Holloway DL, Rasmussen AS, et al. A single-dose placebocontrolled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 2004; 19: 1059–66

    Article  PubMed  CAS  Google Scholar 

  15. Miller PD, Bolognese MA, Lewiecki EM, et al. Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after long-term continued, discontinued, and restarting of therapy: a randomized blinded phase 2 clinical trial. Bone 2008; 43: 222–9

    Article  PubMed  CAS  Google Scholar 

  16. Bone HG, Bolognese MA, Yuen CK, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women. J Clin Endocrinol Metab 2008; 93: 2149–57

    Article  PubMed  CAS  Google Scholar 

  17. Smith MR, Egerdie B, Hernandez TN, et al. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 2009; 361: 745–55

    Article  PubMed  CAS  Google Scholar 

  18. Amgen Inc. Prolia (denosumab) investigator’s brochure. Thousand Oaks (CA): Amgen Inc., 2010

    Google Scholar 

  19. European Medicines Agency. CHMP assessment report for Prolia [document reference EMA/21672/2010; online]. Available from URL: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Public_assessment_report/human/001120/WC500093529.pdf [Accessed 2011 Sep 26]

  20. Mager DE, Jusko WJ. General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J Pharmacokinet Pharmacodyn 2001; 28: 507–32

    Article  PubMed  CAS  Google Scholar 

  21. Mager DE, Krzyzanski W. Quasi-equilibrium pharmacokinetic model for drugs exhibiting target-mediated drug disposition. Pharm Res 2005; 22: 1589–96

    Article  PubMed  CAS  Google Scholar 

  22. Gibiansky L, Gibiansky E, Kakkar T, et al. Approximations of the target-mediated drug disposition model and identifiability of model parameters. J Pharmacokinet Pharmacodyn 2008; 35: 573–91

    Article  PubMed  CAS  Google Scholar 

  23. Gibiansky L, Gibiansky E. Target-mediated drug disposition model: approximations, identifiability of model parameters and applications to the population pharmacokinetic-pharmacodynamic modeling of biologics. Expert Opin Drug Metab Toxicol 2009; 5: 803–12

    Article  PubMed  CAS  Google Scholar 

  24. Girard P. Data transformation and parameter transformations in NONMEM. Eleventh Meeting, Population Approach Group in Europe; 2002 Jun 6–7; Paris [online]. Available from URL: http://www.page-meeting.org/page/page2002/PascalGirardPage2002.pdf [Accessed 2011 Sep 26]

    Google Scholar 

  25. Bergstrand M, Karlsson MO. Handling data below the limit of quantification in mixed effect models. AAPS J 2009; 11: 371–80

    Article  PubMed  CAS  Google Scholar 

  26. Beal SL. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn 2001; 28: 481–504

    Article  PubMed  CAS  Google Scholar 

  27. Ahn JE, Karlsson MO, Dunne A, et al. Likelihood based approaches to handling data below the quantification limit using NONMEM VI. J Pharmacokinet Pharmacodyn 2008; 35: 401–21

    Article  PubMed  Google Scholar 

  28. Byon W, Fletcher CV, Brundage RC. Impact of censoring data below an arbitrary quantification limit on structural model misspecification. J Pharmacokinet Pharmacodyn 2008; 35: 101–16

    Article  PubMed  Google Scholar 

  29. Mahmood I. Clinical pharmacology of therapeutic proteins. Rockville (MD): Pine House Publishers, 2006

    Google Scholar 

  30. Kuester K, Kloft C. Pharmacokinetics of monoclonal antibodies. In: Meibohm B, editor. Pharmacokinetics and pharmacodynamics of biotech drugs: principles and case studies in drug development. Weinheim: Wiley-VCH Verlag GmbH & Co. KGaA, 2006: 45–91

    Chapter  Google Scholar 

  31. Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 2008; 84: 548–58

    Article  PubMed  CAS  Google Scholar 

  32. Lobo ED, Hansen RJ, Balthasar JP. Antibody pharmacokinetics and pharmacodynamics. J Pharm Sci 2004; 93: 2645–68

    Article  PubMed  CAS  Google Scholar 

  33. Block G, Bone HG, Fang L, et al. A single dose study of denosumab in patients with various degrees of renal impairment [abstract]. J Amer Kidney Dis 2010; 55(4): B46

    Article  Google Scholar 

  34. Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/pharmacodynamic models using the posterior predictive check. J Pharmacokinet Pharmacodyn 2001; 28: 171–92

    Article  PubMed  CAS  Google Scholar 

  35. Yan X, Mager DE, Krzyzanski W. Selection between Michaelis-Menten and target-mediated drug disposition pharmacokinetic models. J Pharmacokinet Pharmacodyn 2010; 37: 25–47

    Article  PubMed  CAS  Google Scholar 

  36. Krippendorff BF, Kuester K, Kloft C, et al. Nonlinear pharmacokinetics of therapeutic proteins resulting from receptor mediated endocytosis. J Pharmacokinet Pharmacodyn 2009; 36: 239–60

    Article  PubMed  CAS  Google Scholar 

  37. Perez-Ruixo JJ, Piotrovskij V, Zhang S, et al. Population pharmacokinetics of tipifarnib in healthy subjects and adult cancer patients. Br J Clin Pharmacol 2006; 62: 81–96

    Article  PubMed  CAS  Google Scholar 

  38. Frame B, Koup J, Miller R, et al. Population pharmacokinetics of clinafloxacin in healthy volunteers and patients with infections: experience with heterogeneous pharmacokinetic data. Clin Pharmacokinet 2001; 40: 307–15

    Article  PubMed  CAS  Google Scholar 

  39. Savic RM, Karlsson MO. Importance of shrinkage in empirical Bayes estimates for diagnostics: problems and solutions. AAPS J 2009; 11: 558–69

    Article  PubMed  Google Scholar 

  40. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet 2010; 49: 633–59

    Article  PubMed  CAS  Google Scholar 

  41. Olsson-Gisleskog P, Jacqmin P, Perez-Ruixo JJ. Population pharmacokinetics meta-analysis of recombinant human erythropoietin in healthy subjects. Clin Pharmacokinet 2007; 46: 159–73

    Article  PubMed  CAS  Google Scholar 

  42. Agoram BM, Martin SW, van der Graaf PH. The role of mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modelling in translational research of biologics. Drug Discov Today 2007; 12: 1018–24

    Article  PubMed  CAS  Google Scholar 

  43. Roskos LK, Davis CG, Schwab GM. The clinical pharmacology of therapeutic monoclonal antibodies. Drug Dev Res 2004; 61: 108–20

    Article  CAS  Google Scholar 

  44. 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

    Article  PubMed  CAS  Google Scholar 

  45. Kostenuik PJ, Nguyen HQ, McCabe J, et al. Denosumab, a fully human monoclonal antibody to RANKL, inhibits bone resorption and increases BMD in knock-in mice that express chimeric (murine/human) RANKL. J Bone Miner Res 2009; 24: 182–95

    Article  PubMed  CAS  Google Scholar 

  46. Hofbauer LC, Schoppet M, Schuller P, et al. Effects of oral contraceptives on circulating osteoprotegerin and soluble RANK ligand serum levels in healthy young women. Clin Endocrinol (Oxf) 2004; 60: 214–9

    Article  CAS  Google Scholar 

  47. Liu JM, Zhao HY, Ning G, et al. Relationships between the changes of serum levels of OPG and RANKL with age, menopause, bone biochemical markers and bone mineral density in Chinese women aged 20–75. Calcif Tissue Int 2005; 76: 1–6

    Article  PubMed  CAS  Google Scholar 

  48. Findlay D, Chehade M, Tsangari H, et al. Circulating RANKL is inversely related to RANKL mRNA levels in bone in osteoarthritic males. Arthritis Res Ther 2008; 10: R2 [online]. Available from URL: http://arthritis-research.com/content/pdf/ar2348.pdf [Accessed 2011 Oct 6]

    Article  PubMed  Google Scholar 

  49. Marathe A, Peterson MC, Mager DE. Integrated cellular bone homeostasis model for denosumab pharmacodynamics in multiple myeloma patients. J Pharmacol Exp Ther 2008; 326: 555–62

    Article  PubMed  CAS  Google Scholar 

  50. Hayashi N, Tsukamoto Y, Sallas WM, et al. A mechanism-based binding model for the population pharmacokinetics and pharmacodynamics of omalizumab. Br J Clin Pharmacol 2007; 63: 548–61

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank Ed Lee and Leonid Gibiansky for the support and comments they provided during the completion of this analysis. The authors also thank the hundreds of patients, investigators, and medical, nursing and laboratory staff who participated in the clinical studies that were included in the present analysis.

This study was sponsored by Amgen Inc., which was involved in the study design; the data collection, analysis and interpretation; the writing of the manuscript; and the decision to submit the manuscript for publication. Liviawati Sutjandra, Sameer Doshi, Mark Ma, Graham Jang, Andrew Chow and Juan José Pérez-Ruixo were employees of Amgen Inc. and owned stock in Amgen Inc. at the time when the analysis was conducted. Rachelle Rodriguez and Mark Peterson are former employees of Amgen Inc. The authors have no other conflicts of interest to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juan José Pérez-Ruixo.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sutjandra, L., Rodriguez, R.D., Doshi, S. et al. Population Pharmacokinetic Meta-Analysis of Denosumab in Healthy Subjects and Postmenopausal Women with Osteopenia or Osteoporosis. Clin Pharmacokinet 50, 793–807 (2011). https://doi.org/10.2165/11594240-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11594240-000000000-00000

Keywords

Navigation