Show simple item record

dc.contributor.authorSwaisland, Helen C
dc.contributor.authorRanson, Malcolm R
dc.contributor.authorSmith, Robert P
dc.contributor.authorLeadbetter, Joanna
dc.contributor.authorLaight, Alison
dc.contributor.authorMcKillop, David
dc.contributor.authorWild, Martin J
dc.date.accessioned2009-07-22T09:15:11Z
dc.date.available2009-07-22T09:15:11Z
dc.date.issued2005
dc.identifier.citationPharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol. 2005, 44 (10):1067-81 Clin Pharmacokineten
dc.identifier.issn0312-5963
dc.identifier.pmid16176119
dc.identifier.urihttp://hdl.handle.net/10541/74893
dc.description.abstractBACKGROUND AND OBJECTIVES: Gefitinib (IRESSA, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor, has been approved in several countries for the treatment of advanced non-small-cell lung cancer. Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Based on these findings, three clinical studies were carried out to investigate pharmacokinetic drug interactions in vivo with gefitinib. METHODS: In preclinical studies radiolabelled gefitinib was incubated with: (i) hepatic microsomal protein in the presence of selective CYP inhibitors; and (ii) expressed CYP enzymes. Human hepatic microsomal protein was incubated with selective CYP substrates in the presence of gefitinib. Clinical studies were all phase I, open-label, single-centre studies; two had a randomised, two-way crossover design and the third was nonrandomised. The first and second studies investigated the pharmacokinetics of gefitinib in the presence of a potent CYP3A4 inducer (rifampicin [rifampin]) or inhibitor (itraconazole) in healthy male volunteers. The third study investigated the effects that gefitinib had on the pharmacokinetics of metoprolol, a CYP2D6 substrate, in patients with solid tumours. RESULTS: The results of preclinical studies demonstrated that CYP3A4 is involved in the metabolism of gefitinib and that gefitinib is a weak inhibitor of CYP2D6 activity. In clinical studies when gefitinib was administered in the presence of rifampicin, geometric mean (gmean) maximum concentration and area under the plasma concentration-time curve (AUC) were reduced by 65% and 83%, respectively; these changes were statistically significant. When gefitinib was administered in the presence of itraconazole, gmean AUC increased by 78% and 61% at gefitinib doses of 250 and 500 mg, respectively; these changes also being statistically significant. Coadministration of metoprolol with gefitinib resulted in a 35% increase in the metoprolol area under plasma concentration-time curve from time zero to the time of the last quantifiable concentration; this change was not statistically significant. There was no apparent change in the safety profile of gefitinib as a result of coadministration with other agents. CONCLUSIONS: Although CYP3A4 inducers may reduce exposure to gefitinib, further work is required to define any resultant effect on the efficacy of gefitinib. Exposure to gefitinib is increased by coadministration with CYP3A4 inhibitors, but since gefitinib is known to have a good tolerability profile, a dosage reduction is not recommended. Gefitinib is unlikely to exert a clinically relevant effect on the pharmacokinetics of drugs that are dependent on CYP2D6-mediated metabolism for their clearance, but the potential to increase plasma concentrations should be considered if gefitinib is coadministered with CYP2D6 substrates that have a narrow therapeutic index or are individually dose titrated.
dc.language.isoenen
dc.subjectCanceren
dc.subject.meshAdolescent
dc.subject.meshAdult
dc.subject.meshAntineoplastic Agents
dc.subject.meshCross-Over Studies
dc.subject.meshCytochrome P-450 CYP2D6
dc.subject.meshCytochrome P-450 CYP3A
dc.subject.meshCytochrome P-450 Enzyme System
dc.subject.meshDrug Combinations
dc.subject.meshDrug Interactions
dc.subject.meshEnzyme Induction
dc.subject.meshEnzyme Inhibitors
dc.subject.meshFemale
dc.subject.meshHumans
dc.subject.meshItraconazole
dc.subject.meshMale
dc.subject.meshMetoprolol
dc.subject.meshMicrosomes, Liver
dc.subject.meshMiddle Aged
dc.subject.meshNeoplasms
dc.subject.meshQuinazolines
dc.subject.meshRifampin
dc.titlePharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol.en
dc.typeArticleen
dc.contributor.departmentAstraZeneca Pharmaceuticals, Macclesfield, UK. helen.swaisland@astrazeneca.comen
dc.identifier.journalClinical Pharmacokineticsen
html.description.abstractBACKGROUND AND OBJECTIVES: Gefitinib (IRESSA, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor, has been approved in several countries for the treatment of advanced non-small-cell lung cancer. Preclinical studies were conducted to determine the cytochrome P450 (CYP) isoenzymes involved in the metabolism of gefitinib and to evaluate the potential of gefitinib to cause drug interactions through inhibition of CYP isoenzymes. Based on these findings, three clinical studies were carried out to investigate pharmacokinetic drug interactions in vivo with gefitinib. METHODS: In preclinical studies radiolabelled gefitinib was incubated with: (i) hepatic microsomal protein in the presence of selective CYP inhibitors; and (ii) expressed CYP enzymes. Human hepatic microsomal protein was incubated with selective CYP substrates in the presence of gefitinib. Clinical studies were all phase I, open-label, single-centre studies; two had a randomised, two-way crossover design and the third was nonrandomised. The first and second studies investigated the pharmacokinetics of gefitinib in the presence of a potent CYP3A4 inducer (rifampicin [rifampin]) or inhibitor (itraconazole) in healthy male volunteers. The third study investigated the effects that gefitinib had on the pharmacokinetics of metoprolol, a CYP2D6 substrate, in patients with solid tumours. RESULTS: The results of preclinical studies demonstrated that CYP3A4 is involved in the metabolism of gefitinib and that gefitinib is a weak inhibitor of CYP2D6 activity. In clinical studies when gefitinib was administered in the presence of rifampicin, geometric mean (gmean) maximum concentration and area under the plasma concentration-time curve (AUC) were reduced by 65% and 83%, respectively; these changes were statistically significant. When gefitinib was administered in the presence of itraconazole, gmean AUC increased by 78% and 61% at gefitinib doses of 250 and 500 mg, respectively; these changes also being statistically significant. Coadministration of metoprolol with gefitinib resulted in a 35% increase in the metoprolol area under plasma concentration-time curve from time zero to the time of the last quantifiable concentration; this change was not statistically significant. There was no apparent change in the safety profile of gefitinib as a result of coadministration with other agents. CONCLUSIONS: Although CYP3A4 inducers may reduce exposure to gefitinib, further work is required to define any resultant effect on the efficacy of gefitinib. Exposure to gefitinib is increased by coadministration with CYP3A4 inhibitors, but since gefitinib is known to have a good tolerability profile, a dosage reduction is not recommended. Gefitinib is unlikely to exert a clinically relevant effect on the pharmacokinetics of drugs that are dependent on CYP2D6-mediated metabolism for their clearance, but the potential to increase plasma concentrations should be considered if gefitinib is coadministered with CYP2D6 substrates that have a narrow therapeutic index or are individually dose titrated.


This item appears in the following Collection(s)

Show simple item record