Skip to main content
Top
Published in: Clinical Pharmacokinetics 7/2009

01-07-2009 | Original Research Article

Refinement of the Population Pharmacokinetic Model for the Monoclonal Antibody Matuzumab

External Model Evaluation and Simulations

Authors: Katharina Kuester, Andreas Kovar, Christian Lüpfert, Brigitte Brockhaus, Prof. Charlotte Kloft

Published in: Clinical Pharmacokinetics | Issue 7/2009

Login to get access

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference De Luca A, Carotenuto A, Rachiglio A, et al. The role of the EGFR signaling in tumor microenvironment. J Cell Physiol 2008; 214: 559–67PubMedCrossRef De Luca A, Carotenuto A, Rachiglio A, et al. The role of the EGFR signaling in tumor microenvironment. J Cell Physiol 2008; 214: 559–67PubMedCrossRef
2.
go back to reference 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: 4834–42PubMed 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: 4834–42PubMed
3.
go back to reference Oda K, Matsuoka Y, Funahashi A, et al. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 2005; 1: 2005–10PubMedCrossRef Oda K, Matsuoka Y, Funahashi A, et al. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol Syst Biol 2005; 1: 2005–10PubMedCrossRef
4.
go back to reference Baselga J. The EGFR as a target for anticancer therapy: focus on cetuximab. Eur J Cancer 2001; 37 Suppl. 4: S16–22PubMedCrossRef Baselga J. The EGFR as a target for anticancer therapy: focus on cetuximab. Eur J Cancer 2001; 37 Suppl. 4: S16–22PubMedCrossRef
5.
go back to reference Baselga J. Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 2002; 7 Suppl. 4: 2–8PubMedCrossRef Baselga J. Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 2002; 7 Suppl. 4: 2–8PubMedCrossRef
6.
go back to reference Baselga J. New therapeutic agents targeting the epidermal growth factor receptor. J Clin Oncol 2000; 18: 54S–9SPubMed Baselga J. New therapeutic agents targeting the epidermal growth factor receptor. J Clin Oncol 2000; 18: 54S–9SPubMed
7.
go back to reference Goel S, Mani S, Perez-Soler R. Tyrosine kinase inhibitors: a clinical perspective. Curr Oncol Rep 2002; 4: 9–19PubMedCrossRef Goel S, Mani S, Perez-Soler R. Tyrosine kinase inhibitors: a clinical perspective. Curr Oncol Rep 2002; 4: 9–19PubMedCrossRef
8.
go back to reference 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 41–2PubMedCrossRef 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 41–2PubMedCrossRef
9.
go back to reference 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: 337–45PubMedCrossRef 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: 337–45PubMedCrossRef
10.
go back to reference Walker RA, Dearing SJ. Expression of epidermal growth factor receptor mRNA and protein in primary breast carcinomas. Breast Cancer Res Treat 1999; 53: 167–76PubMedCrossRef Walker RA, Dearing SJ. Expression of epidermal growth factor receptor mRNA and protein in primary breast carcinomas. Breast Cancer Res Treat 1999; 53: 167–76PubMedCrossRef
11.
go back to reference 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: 1850–5PubMed 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: 1850–5PubMed
12.
go back to reference 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: 603–7PubMed 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: 603–7PubMed
13.
go back to reference 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: 147–52PubMedCrossRef 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: 147–52PubMedCrossRef
14.
go back to reference Bruno R, Washington CB, Lu JF, et al. Population pharmacokinetics of trastuzumab in patients with HER2+ metastatic breast cancer. Cancer Chemother Pharmacol 2005; 56: 361–9PubMedCrossRef Bruno R, Washington CB, Lu JF, et al. Population pharmacokinetics of trastuzumab in patients with HER2+ metastatic breast cancer. Cancer Chemother Pharmacol 2005; 56: 361–9PubMedCrossRef
15.
go back to reference Ritter CA, Arteaga CL. The epidermal growth factor receptor-tyrosine kinase: a promising therapeutic target in solid tumors. Semin Oncol 2003; 30: 3–11PubMedCrossRef Ritter CA, Arteaga CL. The epidermal growth factor receptor-tyrosine kinase: a promising therapeutic target in solid tumors. Semin Oncol 2003; 30: 3–11PubMedCrossRef
17.
go back to reference Herbst RS, Shin DM. Monoclonal antibodies to target epidermal growth factor receptor-positive tumors: a new paradigm for cancer therapy. Cancer 2002; 94: 1593–611PubMedCrossRef Herbst RS, Shin DM. Monoclonal antibodies to target epidermal growth factor receptor-positive tumors: a new paradigm for cancer therapy. Cancer 2002; 94: 1593–611PubMedCrossRef
18.
go back to reference Harding J, Burtness B. Cetuximab: an epidermal growth factor receptor chemeric human-murine monoclonal antibody. Drugs Today (Barc) 2005; 41: 107–27CrossRef Harding J, Burtness B. Cetuximab: an epidermal growth factor receptor chemeric human-murine monoclonal antibody. Drugs Today (Barc) 2005; 41: 107–27CrossRef
21.
go back to reference 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: s4597–601PubMedCrossRef 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: s4597–601PubMedCrossRef
22.
go back to reference Vallbohmer D, Lenz HJ. Epidermal growth factor receptor as a target for chemotherapy. Clin Colorectal Cancer 2005; 5 Suppl. 1: S19–27PubMedCrossRef Vallbohmer D, Lenz HJ. Epidermal growth factor receptor as a target for chemotherapy. Clin Colorectal Cancer 2005; 5 Suppl. 1: S19–27PubMedCrossRef
23.
go back to reference 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: 1293–9PubMedCrossRef 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: 1293–9PubMedCrossRef
24.
go back to reference 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: 69–79PubMedCrossRef 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: 69–79PubMedCrossRef
25.
go back to reference 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: 549–60PubMedCrossRef 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: 549–60PubMedCrossRef
26.
go back to reference 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: 773–83PubMedCrossRef 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: 773–83PubMedCrossRef
27.
go back to reference 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: 900–6PubMedCrossRef 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: 900–6PubMedCrossRef
28.
go back to reference Janmahasatian S, Duffull SB, Ash S, et al. Quantification of lean bodyweight. Clin Pharmacokinet 2005; 44: 1051–65PubMedCrossRef Janmahasatian S, Duffull SB, Ash S, et al. Quantification of lean bodyweight. Clin Pharmacokinet 2005; 44: 1051–65PubMedCrossRef
29.
go back to reference 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: 221–34PubMedCrossRef 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: 221–34PubMedCrossRef
30.
go back to reference 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: 175–84PubMedCrossRef 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: 175–84PubMedCrossRef
31.
go back to reference 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: 1885–900PubMedCrossRef 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: 1885–900PubMedCrossRef
32.
go back to reference Bonate PL. Pharmacokinetic-pharmacodynamic modeling and simulation. New York: Springer Science and Business Media, 2006 Bonate PL. Pharmacokinetic-pharmacodynamic modeling and simulation. New York: Springer Science and Business Media, 2006
33.
go back to reference Bonate PL. The effect of collinearity on parameter estimates in nonlinear mixed effect models. Pharm Res 1999; 16: 709–17PubMedCrossRef Bonate PL. The effect of collinearity on parameter estimates in nonlinear mixed effect models. Pharm Res 1999; 16: 709–17PubMedCrossRef
34.
go back to reference Fliss G, Staab A, Tillmann C, et al. Population pharmacokinetic data analysis of cilobradine, an I(f) channel blocker. Pharm Res 2008; 25: 359–68PubMedCrossRef Fliss G, Staab A, Tillmann C, et al. Population pharmacokinetic data analysis of cilobradine, an I(f) channel blocker. Pharm Res 2008; 25: 359–68PubMedCrossRef
35.
go back to reference Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/pharmacodynamic models using the posterior predictive check. J Pharmacokinet Pharmacodyn 2001; 28: 171–92PubMedCrossRef Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/pharmacodynamic models using the posterior predictive check. J Pharmacokinet Pharmacodyn 2001; 28: 171–92PubMedCrossRef
37.
go back to reference Belsley DA, Kuh E, Welsh RE. Regression diagnostics: identifying influential data and sources of collinearity. New York: John Wiley & Sons, Inc., 1980CrossRef Belsley DA, Kuh E, Welsh RE. Regression diagnostics: identifying influential data and sources of collinearity. New York: John Wiley & Sons, Inc., 1980CrossRef
38.
39.
go back to reference 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: 483–8PubMedCrossRef 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: 483–8PubMedCrossRef
40.
go back to reference Aarons L, Karlsson MO, Mentre F, et al. Role of modelling and simulation in phase I drug development. Eur J Pharm Sci 2001; 13: 115–22PubMedCrossRef Aarons L, Karlsson MO, Mentre F, et al. Role of modelling and simulation in phase I drug development. Eur J Pharm Sci 2001; 13: 115–22PubMedCrossRef
41.
go back to reference 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: 419–40PubMedCrossRef 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: 419–40PubMedCrossRef
42.
go back to reference 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: E60–83PubMedCrossRef 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: E60–83PubMedCrossRef
43.
go back to reference Dingemanse J, Appel-Dingemanse S. Integrated pharmacokinetics and pharmacodynamics in drug development. Clin Pharmacokinet 2007; 46: 713–37PubMedCrossRef Dingemanse J, Appel-Dingemanse S. Integrated pharmacokinetics and pharmacodynamics in drug development. Clin Pharmacokinet 2007; 46: 713–37PubMedCrossRef
44.
go back to reference Gieschke R, Steimer JL. Pharmacometrics: modelling and simulation tools to improve decision making in clinical drug development. Eur J Drug Metab Pharmacokinet 2000; 25: 49–58PubMedCrossRef Gieschke R, Steimer JL. Pharmacometrics: modelling and simulation tools to improve decision making in clinical drug development. Eur J Drug Metab Pharmacokinet 2000; 25: 49–58PubMedCrossRef
45.
go back to reference Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: general principles. Eur J Pediatr 2006; 165: 741–6PubMedCrossRef Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: general principles. Eur J Pediatr 2006; 165: 741–6PubMedCrossRef
46.
go back to reference Hortobagyi GN. Overview of treatment results with trastuzumab (Herceptin) in metastatic breast cancer. Semin Oncol 2001; 28: 43–7PubMedCrossRef Hortobagyi GN. Overview of treatment results with trastuzumab (Herceptin) in metastatic breast cancer. Semin Oncol 2001; 28: 43–7PubMedCrossRef
47.
go back to reference 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: 5481–90PubMedCrossRef 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: 5481–90PubMedCrossRef
48.
go back to reference 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: 39–52PubMedCrossRef 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: 39–52PubMedCrossRef
Metadata
Title
Refinement of the Population Pharmacokinetic Model for the Monoclonal Antibody Matuzumab
External Model Evaluation and Simulations
Authors
Katharina Kuester
Andreas Kovar
Christian Lüpfert
Brigitte Brockhaus
Prof. Charlotte Kloft
Publication date
01-07-2009
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 7/2009
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.2165/11313400-000000000-00000