Skip to main content
Log in

Refinement of the Population Pharmacokinetic Model for the Monoclonal Antibody Matuzumab

External Model Evaluation and Simulations

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

Abstract

Objectives

A developed population pharmacokinetic model of the humanized monoclonal antibody (mAb) matuzumab was evaluated by external evaluation. Based on the estimates of the final model, simulations of different dosing regimens and the covariate effect were performed.

Methods

The development dataset included 90 patients, and the evaluation dataset included 81 patients; the two sets of patients were from three different studies. In all studies, the patients had different types of advanced carcinoma — mainly colon, rectal and pancreatic cancer. They received matuzumab as multiple 1-hour intravenous infusions in a wide range of dosing regimens (development dataset: from 400 mg every 3 weeks to 2000 mg in the first week followed by 1600 mg weekly; evaluation dataset: from 100 mg weekly to 800 mg weekly). In addition to 1256 serum mAb concentrations for model development, there were 1124 concentrations available for model evaluation. Serum concentration-time data were simultaneously fitted using NONMEM™ software. The developed two-compartment model — with the parameters central volume of distribution (V1) and peripheral volume of distribution (V2), intercompartmental clearance and linear clearance (CLL), an additional nonlinear elimination pathway (Michaelis-Menten constant: the concentration with the half-maximal elimination rate and Vmax: the maximum elimination rate) and covariate relations — was evaluated by an external dataset. Different simulation scenarios were performed to demonstrate the impact of the incorporated covariate effect and the influence of different dosing regimens and dosing strategies on the concentration-time profiles.

Results

The developed model included the covariate fat-free mass (FFM) on V1 and on CLL. The evaluation did not support the covariate FFM on V1 and, after deletion of this covariate, the model parameters of the refined model were estimated. The model showed good precision for all parameters: the relative standard errors (RSEs) were <42% for the development dataset and ≤51% for the evaluation dataset (excluding the higher RSEs for the correlation between V2 and Vmax and the interindividual variability on V2 for the evaluation dataset). The model showed good robustness for the ability to estimate highly precise parameters for the combined dataset of 171 patients (RSE <29%). Simulations revealed that variability in concentration-time profiles for minimum and maximum steady-state concentrations was reduced to a marginal extent by a proposed dose adaptation.

Conclusion

The population pharmacokinetic model for matuzumab was improved by evaluation with an external dataset. The new model obtained precise parameter estimates and demonstrated robustness. After correlation with efficacy data simulation results in particular could serve as a tool to guide dose selection for this ‘targeted’ cancer therapy.

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.

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

Similar content being viewed by others

References

  1. De Luca A, Carotenuto A, Rachiglio A, et al. The role of the EGFR signaling in tumor microenvironment. J Cell Physiol 2008; 214: 559–67

    Article  PubMed  Google Scholar 

  2. Cohen S, Carpenter G, King Jr L, et al. Epidermal growth factor-receptorprotein kinase interactions: co-purification of receptor and epidermal growth factor-enhanced phosphorylation activity. J Biol Chem 1980; 255: 4834–42

    PubMed  CAS  Google Scholar 

  3. Oda K, Matsuoka Y, Funahashi A, et al. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 2005; 1: 2005–10

    Article  PubMed  Google Scholar 

  4. Baselga J. The EGFR as a target for anticancer therapy: focus on cetuximab. Eur J Cancer 2001; 37 Suppl. 4: S16–22

    Article  PubMed  CAS  Google Scholar 

  5. Baselga J. Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 2002; 7 Suppl. 4: 2–8

    Article  PubMed  CAS  Google Scholar 

  6. Baselga J. New therapeutic agents targeting the epidermal growth factor receptor. J Clin Oncol 2000; 18: 54S–9S

    PubMed  CAS  Google Scholar 

  7. Goel S, Mani S, Perez-Soler R. Tyrosine kinase inhibitors: a clinical perspective. Curr Oncol Rep 2002; 4: 9–19

    Article  PubMed  Google Scholar 

  8. Raymond E, Faivre S, Armand JP. Epidermal growth factor receptor tyrosine kinase as a target for anticancer therapy. Drugs 2000; 60 Suppl. 1: 15–23; discussion 41–2

    Article  PubMed  CAS  Google Scholar 

  9. Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med 2004; 351: 337–45

    Article  PubMed  CAS  Google Scholar 

  10. Walker RA, Dearing SJ. Expression of epidermal growth factor receptor mRNA and protein in primary breast carcinomas. Breast Cancer Res Treat 1999; 53: 167–76

    Article  PubMed  CAS  Google Scholar 

  11. Brabender J, Danenberg KD, Metzger R, et al. Epidermal growth factor receptor and HER2-neu mRNA expression in non-small cell lung cancer is correlated with survival. Clin Cancer Res 2001; 7: 1850–5

    PubMed  CAS  Google Scholar 

  12. Ohsaki Y, Tanno S, Fujita Y, et al. Epidermal growth factor receptor expression correlates with poor prognosis in non-small cell lung cancer patients with p53 overexpression. Oncol Rep 2000; 7: 603–7

    PubMed  CAS  Google Scholar 

  13. Hemming AW, Davis NL, Kluftinger A, et al. Prognostic markers of colorectal cancer: an evaluation of DNA content, epidermal growth factor receptor, and Ki-67. J Surg Oncol 1992; 51: 147–52

    Article  PubMed  CAS  Google Scholar 

  14. Bruno R, Washington CB, Lu JF, et al. Population pharmacokinetics of trastuzumab in patients with HER2+ metastatic breast cancer. Cancer Chemother Pharmacol 2005; 56: 361–9

    Article  PubMed  CAS  Google Scholar 

  15. Ritter CA, Arteaga CL. The epidermal growth factor receptor-tyrosine kinase: a promising therapeutic target in solid tumors. Semin Oncol 2003; 30: 3–11

    Article  PubMed  CAS  Google Scholar 

  16. Wells A. EGF receptor. Int J Biochem Cell Biol 1999; 31: 637–43

    Article  PubMed  CAS  Google Scholar 

  17. Herbst RS, Shin DM. Monoclonal antibodies to target epidermal growth factor receptor-positive tumors: a new paradigm for cancer therapy. Cancer 2002; 94: 1593–611

    Article  PubMed  CAS  Google Scholar 

  18. Harding J, Burtness B. Cetuximab: an epidermal growth factor receptor chemeric human-murine monoclonal antibody. Drugs Today (Barc) 2005; 41: 107–27

    Article  CAS  Google Scholar 

  19. Office of Oncology Drug Products, US FDA Center for Drug Evaluation and Research. Erbitux (cetuximab) approved for use in combination with radiation therapy [media release]. 2006 Jul 20 [online]. Available from URL: http://www.fda.gov/cder/Offices/OODP/whatsnew/cetuximab.htm [Accessed 2008 May 18]

  20. US FDA Center for Drug Evaluation and Research. FDA expands labeling for cetuximab (marketed as Erbitux) [media release]. 2007 Oct 12 [online]. Available from URL: http://www.fda.gov/cder/Offices/OODP/whatsnew/centuximab200710.htm [Accessed 2008 May 18]

  21. Socinski MA. Antibodies to the epidermal growth factor receptor in non small cell lung cancer: current status of matuzumab and panitumumab. Clin Cancer Res 2007; 13: s4597–601

    Article  PubMed  Google Scholar 

  22. Vallbohmer D, Lenz HJ. Epidermal growth factor receptor as a target for chemotherapy. Clin Colorectal Cancer 2005; 5 Suppl. 1: S19–27

    Article  PubMed  CAS  Google Scholar 

  23. Graeven U, Kremer B, Sudhoff T, et al. Phase I study of the humanised antiEGFR monoclonal antibody matuzumab (EMD 72000) combined with gemcitabine in advanced pancreatic cancer. Br J Cancer 2006; 94: 1293–9

    Article  PubMed  CAS  Google Scholar 

  24. Rodeck U, Williams N, Murthy U, et al. Monoclonal antibody 425 inhibits growth stimulation of carcinoma cells by exogenous EGF and tumor-derived EGF/TGF-alpha. J Cell Biochem 1990; 44: 69–79

    Article  PubMed  CAS  Google Scholar 

  25. Murthy U, Basu A, Rodeck U, et al. Binding of an antagonistic monoclonal antibody to an intact and fragmented EGF-receptor polypeptide. Arch Biochem Biophys 1987; 252: 549–60

    Article  PubMed  CAS  Google Scholar 

  26. Kettleborough CA, Saldanha J, Heath VJ, et al. Humanization of a mouse monoclonal antibody by CDR-grafting: the importance of framework residues on loop conformation. Protein Eng 1991; 4: 773–83

    Article  PubMed  CAS  Google Scholar 

  27. Kuester K, Kovar A, Lupfert C, et al. Population pharmacokinetic data analysis of three phase I studies of matuzumab, a humanised anti-EGFR monoclonal antibody in clinical cancer development. Br J Cancer 2008; 98: 900–6

    Article  PubMed  CAS  Google Scholar 

  28. Janmahasatian S, Duffull SB, Ash S, et al. Quantification of lean bodyweight. Clin Pharmacokinet 2005; 44: 1051–65

    Article  PubMed  Google Scholar 

  29. Brendel K, Dartois C, Comets E, et al. Are population pharmacokinetic and/or pharmacodynamic models adequately evaluated? A survey of the literature from 2002 to 2004. Clin Pharmacokinet 2007; 46: 221–34

    Article  PubMed  Google Scholar 

  30. Vanhoefer U, Tewes M, Rojo F, et al. Phase I study of the humanized antiepidermal growth factor receptor monoclonal antibody EMD72000 in patients with advanced solid tumors that express the epidermal growth factor receptor. J Clin Oncol 2004; 22: 175–84

    Article  PubMed  CAS  Google Scholar 

  31. DeSilva B, Smith W, Weiner R, et al. Recommendations for the bioanalytical method validation of ligand-binding assays to support pharmacokinetic assessments of macromolecules. Pharm Res 2003; 20: 1885–900

    Article  PubMed  CAS  Google Scholar 

  32. Bonate PL. Pharmacokinetic-pharmacodynamic modeling and simulation. New York: Springer Science and Business Media, 2006

    Google Scholar 

  33. Bonate PL. The effect of collinearity on parameter estimates in nonlinear mixed effect models. Pharm Res 1999; 16: 709–17

    Article  PubMed  CAS  Google Scholar 

  34. Fliss G, Staab A, Tillmann C, et al. Population pharmacokinetic data analysis of cilobradine, an I(f) channel blocker. Pharm Res 2008; 25: 359–68

    Article  PubMed  CAS  Google Scholar 

  35. 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 

  36. Michels GH. The use of collinearity diagnostics in PK model building [online]. Available from URL: http://www.ecpag.org/presentations/2006/0725/2_GregoryMichelsv4.pdf [Accessed 2008 Feb 27]

  37. Belsley DA, Kuh E, Welsh RE. Regression diagnostics: identifying influential data and sources of collinearity. New York: John Wiley & Sons, Inc., 1980

    Book  Google Scholar 

  38. Mager DE. Target-mediated drug disposition and dynamics. Biochem Pharmacol 2006; 72: 1–10

    Article  PubMed  CAS  Google Scholar 

  39. Hing JP, Piotrovsky V, Kimko H, et al. Pharmacokinetic simulation for switching from galantamine immediate-release to extended-release formulation. Curr Med Res Opin 2005; 21: 483–8

    Article  PubMed  CAS  Google Scholar 

  40. Aarons L, Karlsson MO, Mentre F, et al. Role of modelling and simulation in phase I drug development. Eur J Pharm Sci 2001; 13: 115–22

    Article  PubMed  CAS  Google Scholar 

  41. Rombout F, Aarons L, Karlsson M, et al. Modelling and simulation in the development and use of anti-cancer agents: an underused tool? J Pharmacokinet Pharmacodyn 2004; 31: 419–40

    Article  PubMed  Google Scholar 

  42. Bauer RJ, Guzy S, Ng C. A survey of population analysis methods and software for complex pharmacokinetic and pharmacodynamic models with examples. AAPS J 2007; 9: E60–83

    Article  PubMed  Google Scholar 

  43. Dingemanse J, Appel-Dingemanse S. Integrated pharmacokinetics and pharmacodynamics in drug development. Clin Pharmacokinet 2007; 46: 713–37

    Article  PubMed  CAS  Google Scholar 

  44. Gieschke R, Steimer JL. Pharmacometrics: modelling and simulation tools to improve decision making in clinical drug development. Eur J Drug Metab Pharmacokinet 2000; 25: 49–58

    Article  PubMed  CAS  Google Scholar 

  45. Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: general principles. Eur J Pediatr 2006; 165: 741–6

    Article  PubMed  Google Scholar 

  46. Hortobagyi GN. Overview of treatment results with trastuzumab (Herceptin) in metastatic breast cancer. Semin Oncol 2001; 28: 43–7

    Article  PubMed  CAS  Google Scholar 

  47. Kloft C, Wallin J, Henningsson A, et al. Population pharmacokineticpharmacodynamic model for neutropenia with patient subgroup identification: comparison across anticancer drugs. Clin Cancer Res 2006; 12: 5481–90

    Article  PubMed  CAS  Google Scholar 

  48. Kloft C, Graefe EU, Tanswell P, et al. Population pharmacokinetics of sibrotuzumab, a novel therapeutic monoclonal antibody, in cancer patients. Invest New Drugs 2004; 22: 39–52

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Bioanalytical samples were measured at the Institute of Drug Metabolism and Pharmacokinetics, Merck KGaA (Grafing, Germany). Raw datasets were provided by Merck KGaA (Darmstadt, Germany). Katharina Kuester received a travel/research grant and Charlotte Kloft received research funding from Merck KGaA. Andreas Kovar, Christian Lüpfert and Brigitte Brockhaus are current employees at Merck KGaA. The authors have no other conflicts of interest to declare.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Charlotte Kloft.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kuester, K., Kovar, A., Lüpfert, C. et al. Refinement of the Population Pharmacokinetic Model for the Monoclonal Antibody Matuzumab. Clin Pharmacokinet 48, 477–487 (2009). https://doi.org/10.2165/11313400-000000000-00000

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/11313400-000000000-00000

Keywords

Navigation