Skip to main content
Log in

Exploring the Relationship Between Expression of Cytochrome P450 Enzymes and Gefitinib Pharmacokinetics

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

Abstract

Background and objectives

Exposure to gefitinib (IRESSA®, ZD1839), an epidermal growth factor receptor-tyrosine kinase inhibitor, is highly variable between subjects. In an attempt to explain this variability, three pharmacokinetic studies were carried out in healthy volunteers to investigate the relationship between exposure to gefitinib and cytochrome P450 (CYP) 3A phenotype (study 1), CYP3A5 genotype (study 2) and CYP2D6 genotype (study 3).

Methods

In study 1 all 15 healthy volunteers received single oral doses of midazolam (7.5mg), as a CYP3A probe, and gefitinib (500mg), separated by an appropriate washout period. Plasma concentrations of midazolam and gefitinib were measured. In study 2, 73 healthy volunteers with previously defined single-dose gefitinib pharmacokinetic profiles were genotyped for CYP3A5. In study 3 a single oral dose of gefitinib (250mg) was administered to poor and extensive CYP2D6 metabolisers (n= 15 in each group). Plasma concentrations of gefitinib and its major metabolite, M523595, were measured. Plasma concentrations of gefitinib, M523595 and midazolam were measured using high-performance liquid chromatography with tandem mass spectrometric detection, and appropriate pharmacokinetic parameters were determined by non-compartmental methods. Genetic analysis of CYP3A5 (study 2) and CYP2D6 (study 3) alleles was carried out using standard methodology.

Results

In study 1 there was some indication of a correlation between the area under the plasma concentration-time curve from time zero to infinity (AUC) values of midazolam and gefitinib, although this did not reach statistical significance (p = 0.062, regression analysis). In study 2 eight of 73 volunteers (11%) were identified as CYP3A5 expressers. No apparent relationship was observed between the occurrence of the CYP3A5 expresser genotype and gefitinib plasma clearance or terminal elimination halflife. In study 3 M523595 was not detected in any plasma samples collected from poor CYP2D6 metabolisers. Gefitinib geometric mean AUC and peak plasma drug concentration were higher in poor CYP2D6 metabolisers compared with extensive metabolisers (AUC 3060 vs 1430 ng · h/mL, p < 0.05, ANOVA), although the range of values was wide with considerable overlap between the groups. Gefitinib was well tolerated in both groups.

Conclusions

Individual differences in CYP3A expression do not explain all the interindividual variability in gefitinib exposure. There is no apparent relationship between CYP3A5 genotype and gefitinib clearance. The lack of measurable levels of M523595 in poor CYP2D6 metabolisers confirms that production of this metabolite is mediated by CYP2D6. Although higher exposure to gefitinib occurs in individuals who are poor CYP2D6 metabolisers, genotyping prior to initiation of therapy and dosage adjustment are 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
Table II
Table III
Fig. 1
Table IV
Table V
Fig. 2
Fig. 3
Table VI
Table VII
Fig. 4
Fig. 5

Similar content being viewed by others

Notes

  1. The use of trade names is for product identification purposes only and does not imply endorsement.

References

  1. Frampton JE, Easthope SE. Gefitinib: a review of its use in the management of advanced non-small-cell lung cancer. Drugs 2004; 64: 2475–92

    Article  PubMed  CAS  Google Scholar 

  2. Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer. J Clin Oncol 2003; 21: 2237–46

    Article  PubMed  CAS  Google Scholar 

  3. Kris MG, Natale RB, Herbst RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small-cell lung cancer: a randomized trial. JAMA 2003; 290: 2149–58

    Article  PubMed  CAS  Google Scholar 

  4. Thatcher N, Chang A, Parikh P, et al. Results of a Phase III placebo-controlled study (ISEL) of gefitinib (IRESSA) plus best supportive care (BSC) in patients with advanced non-small-cell lung cancer (NSCLC) who had received 1 or 2 prior chemotherapy regimens [abstract LB-L6]. Proceedings of 96th Annual Meeting of American Association for Cancer Research (AACR); 2005 Apr 16–20; Anaheim/Orange County (CA)

    Google Scholar 

  5. Botwood N, Caroll K, Thatcher N. A double blind, placebo controlled, parallel group, multicentre, randomised phase III survival study comparing ZD1839 (IRESSA™) [250mg tablets] plus best supportive care versus placebo plus best supportive care in patients with advanced NSCLC who have received one or two prior chemotherapy regimens and are refractory or intolerant to their most recent regimen. AstraZeneca Study Number D7913C00709. Macclesfield, UK: AstraZeneca; 2005 Jun 6

    Google Scholar 

  6. Swaisland HC, Smith RP, Laight A, et al. Single-dose clinical pharmacokinetic studies of gefitinib (IRESSA, ZD1839), an oral epidermal growth factor receptor tyrosine kinase inhibitor. Clin Pharmacokinet 2005; 44(11): 1165–77

    Article  PubMed  CAS  Google Scholar 

  7. Averbuch S, Swaisland H, Wolf M, et al. ZD1839 (IRESSA™) New drug application. Item 6. Human pharmacokinetics and bioavailability. 2001 Dec 5

  8. Gibbs MA, Hosea NA. Factors affecting the clinical development of cytochrome P450 3A substrates. Clin Pharmacokinet 2003; 42: 969–84

    Article  PubMed  CAS  Google Scholar 

  9. Watkins PB. Noninvasive tests of CYP3A enzymes. Pharmacogenetics 1994; 4: 171–84

    Article  PubMed  CAS  Google Scholar 

  10. McKillop D, McCormick AD, Millar A, et al. Cytochrome P450-dependent metabolism of gefitinib. Xenobiotica 2005; 35: 39–50

    Article  PubMed  CAS  Google Scholar 

  11. Von Moltke LL, Greenblatt DJ, Schmider J, et al. Metabolism of drugs by cytochrome P450 3A isoforms. Implications for drug interactions in psychopharmacology. Clin Pharmacokinet 1995; 29 Suppl. 1: 33–44

    Article  Google Scholar 

  12. Wandel C, Bocker RH, Bohrer H, et al. Relationship between hepatic cytochrome P450 3A content and activity and the disposition of midazolam administered orally. Drug Metab Dispos 1998; 26: 110–4

    PubMed  CAS  Google Scholar 

  13. Thummel KE, Shen DD, Podoll TD, et al. Use of midazolam as a human cytochrome P450 3A probe: I. In vitro-in vivo correlations in liver transplant patients. J Pharmacol Exp Ther 1994; 271: 549–56

    PubMed  CAS  Google Scholar 

  14. Hustert E, Haberl M, Burk O, et al. The genetic determinants of the CYP3A5 polymorphism. Pharmacogenetics 2001; 11: 773–9

    Article  PubMed  CAS  Google Scholar 

  15. Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterisation of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001; 27: 383–91

    Article  PubMed  CAS  Google Scholar 

  16. Sachse C, Brockmöller J, Bauer S, et al. Cytochrome P450 2D6 variants in a Caucasian population: allele frequencies and phenotypic consequences. Am J Hum Genet 1997; 60: 284–95

    PubMed  CAS  Google Scholar 

  17. Kivisto KT, Kroemer HK. Use of probe drugs as predictors of drug metabolism in humans. J Clin Pharmacol 1997; 37: 40S–8S

    Article  PubMed  CAS  Google Scholar 

  18. Laight A. An open, two period, single centre phase I trial to determine the multiple dose pharmacokinetics of ZD1839 following administration over a 14-day period to healthy male volunteers and to assess the effects of a loading dose regimen on the time to reach steady state. AstraZeneca Trial Number 1839IL/0034. Macclesfield, UK: AstraZeneca; 2000 Nov 27

    Google Scholar 

  19. Swaisland HC, Ranson M, Smith RP, et al. Pharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol. Clin Pharmacokinet 2005; 44(10): 1067–81

    Article  PubMed  CAS  Google Scholar 

  20. Swaisland H, Laight A, Stafford L, et al. Pharmacokinetics and tolerability of the orally active selective epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 in healthy volunteers. Clin Pharmacokinet 2001; 40: 297–306

    Article  PubMed  CAS  Google Scholar 

  21. Jones HK, Stafford LE, Swaisland HC, et al. A sensitive assay for ZD1839 (Iressa) in human plasma by liquid-liquid extraction and high performance liquid chromatography with mass spectrometric detection: validation and use in phase I clinical trials. J Pharm Biomed Anal 2002; 29: 221–8

    Article  PubMed  CAS  Google Scholar 

  22. Zaigler M, Tantcheva-Poör I, Fuhr U. Problems and perspectives of phenotyping for drug-metabolizing enzymes in man. Int J Clin Pharmacol Ther 2000; 38: 1–9

    PubMed  CAS  Google Scholar 

  23. Goh BB-C, Lee S-C, Wang L-Z, et al. Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol 2002; 20: 3683–90

    Article  PubMed  CAS  Google Scholar 

  24. Hirth J, Watkins PB, Strawderman M, et al. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 2000; 6: 1255–8

    PubMed  CAS  Google Scholar 

  25. Lown KS, Thummel KE, Benedict PE, et al. The erythromycin breath test predicts the clearance of midazolam. Clin Pharmacol Ther 1995; 57: 16–24

    Article  PubMed  CAS  Google Scholar 

  26. Xie HG, Wood AJ, Kim RB, et al. Genetic variability in CYP3A5 and its possible consequences. Pharmacogenomics 2004; 5: 243–72

    Article  PubMed  CAS  Google Scholar 

  27. Forsythe B, Faulkner K. Overview of the tolerability of gefitinib (IRESSA™) monotherapy: clinical experience in non-small-cell lung cancer. Drug Saf 2004; 27: 1081–92

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the analytical and technical support provided by Analytico Medinet BV, Pharma Bio-Research and within the Drug Metabolism and Pharmacokinetics Department of AstraZeneca, for analysis of the plasma samples. We thank Nancy Robertson from the Clinical Genotyping Group of AstraZeneca for CYP3A5 genotyping and Epidauros Biotechnologie AG for CYP2D6 genotyping. Editorial assistance was provided by Jenny Stewart and financed by AstraZeneca.

No sources of funding were used to assist in conducting this study. The authors have no potential conflicts of interest that are directly relevant to the contents of this study.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Helen C. Swaisland.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Swaisland, H.C., Cantarini, M.V., Fuhr, R. et al. Exploring the Relationship Between Expression of Cytochrome P450 Enzymes and Gefitinib Pharmacokinetics. Clin Pharmacokinet 45, 633–644 (2006). https://doi.org/10.2165/00003088-200645060-00006

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-200645060-00006

Keywords

Navigation