Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 1/2012

01-01-2012 | Original Article

In vitro assessment of cytochrome P450 inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin

Authors: Jinping Gan, Peggy Liu-Kreyche, W. Griffith Humphreys

Published in: Cancer Chemotherapy and Pharmacology | Issue 1/2012

Login to get access

Abstract

Purpose

To assess the inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin (17-AG) on cytochrome P450 (CYP) enzymes.

Methods

The inhibitory effect of tanespimycin and 17-AG on various CYP enzymes was determined in human liver microsomes. The inductive effects of tanespimycin and 17-AG on CYP1A2, CYP2B6, and CYP3A4/5 were determined in cultured primary human hepatocytes.

Results

Tanespimycin did not inhibit the activities of CYP1A2, 2A6, 2B6, and 2E1 up to a concentration of 60 μM, while it moderately inhibited CYP3A4/5 and 2C19, and weakly inhibited CYP2C8, 2C9, and 2D6. In addition, its inhibition on CYP3A4/5 was time-dependent. 17-AG moderately inhibited the activities of CYP3A4/5 and CYP2C19, but did not inhibit other CYPs up to a concentration of 30 μM. The inhibition of CYP3A4/5 by 17-AG was not time-dependent. Tanespimycin and 17-AG did not significantly induce the activities of CYP1A2, CYP2B6, or CYP3A4/5 in cultured human hepatocytes at concentrations up to 40 and 20 μM for tanespimycin and 17-AG, respectively.

Conclusions

Tanespimycin together with its active metabolite, 17-AG are moderate inhibitors of CYP3A4/5 and CYP2C19, but not inducers of CYPs. Therefore, co-administration of tanespimycin has the potential to increase the exposure of substrates of CYP2C19 and CYP3A4/5.
Literature
1.
go back to reference Erlichman C (2009) Tanespimycin: the opportunities and challenges of targeting heat shock protein 90. Expert Opin on Investig Drugs 18:861–868CrossRef Erlichman C (2009) Tanespimycin: the opportunities and challenges of targeting heat shock protein 90. Expert Opin on Investig Drugs 18:861–868CrossRef
2.
go back to reference Lang W, Caldwell GW, Li J, Leo GC, Jones WJ, Masucci JA (2007) Biotransformation of geldanamycin and 17-allylamino-17-demethoxygeldanamycin by human liver microsomes: reductive versus oxidative metabolism and implications. Drug Metab Dispos 35:21–29PubMedCrossRef Lang W, Caldwell GW, Li J, Leo GC, Jones WJ, Masucci JA (2007) Biotransformation of geldanamycin and 17-allylamino-17-demethoxygeldanamycin by human liver microsomes: reductive versus oxidative metabolism and implications. Drug Metab Dispos 35:21–29PubMedCrossRef
3.
go back to reference Egorin MJ, Rosen DM, Wolff JH, Callery PS, Musser SM, Eiseman JL (1998) Metabolism of 17-(allylamino)-17-demethoxygeldanamycin (nsc 330507) by murine and human hepatic preparations. Cancer Res 58:2385–2396PubMed Egorin MJ, Rosen DM, Wolff JH, Callery PS, Musser SM, Eiseman JL (1998) Metabolism of 17-(allylamino)-17-demethoxygeldanamycin (nsc 330507) by murine and human hepatic preparations. Cancer Res 58:2385–2396PubMed
4.
go back to reference Schnur RC, Corman ML, Gallaschun RJ, Cooper BA, Dee MF, Doty JL, Muzzi ML, Moyer JD, DiOrio CI, Barbacci EG et al (1995) Inhibition of the oncogene product p185erbb-2 in vitro and in vivo by geldanamycin and dihydrogeldanamycin derivatives. J Med Chem 38:3806–3812PubMedCrossRef Schnur RC, Corman ML, Gallaschun RJ, Cooper BA, Dee MF, Doty JL, Muzzi ML, Moyer JD, DiOrio CI, Barbacci EG et al (1995) Inhibition of the oncogene product p185erbb-2 in vitro and in vivo by geldanamycin and dihydrogeldanamycin derivatives. J Med Chem 38:3806–3812PubMedCrossRef
5.
go back to reference Walsky RL, Obach RS (2004) Validated assays for human cytochrome p450 activities. Drug Metab Dispos 32:647–660PubMedCrossRef Walsky RL, Obach RS (2004) Validated assays for human cytochrome p450 activities. Drug Metab Dispos 32:647–660PubMedCrossRef
6.
go back to reference Yao M, Zhu M, Sinz MW, Zhang H, Humphreys WG, Rodrigues AD, Dai R (2007) Development and full validation of six inhibition assays for five major cytochrome p450 enzymes in human liver microsomes using an automated 96-well microplate incubation format and lc-ms/ms analysis. J Pharm Biomed Anal 44:211–223PubMedCrossRef Yao M, Zhu M, Sinz MW, Zhang H, Humphreys WG, Rodrigues AD, Dai R (2007) Development and full validation of six inhibition assays for five major cytochrome p450 enzymes in human liver microsomes using an automated 96-well microplate incubation format and lc-ms/ms analysis. J Pharm Biomed Anal 44:211–223PubMedCrossRef
7.
go back to reference LeCluyse EL (2001) Human hepatocyte culture systems for the in vitro evaluation of cytochrome p450 expression and regulation. Eur J Pharm Sci 13:343–368PubMedCrossRef LeCluyse EL (2001) Human hepatocyte culture systems for the in vitro evaluation of cytochrome p450 expression and regulation. Eur J Pharm Sci 13:343–368PubMedCrossRef
8.
go back to reference Wortelboer HM, de Kruif CA, van Iersel AA, Falke HE, Noordhoek J, Blaauboer BJ (1990) The isoenzyme pattern of cytochrome p450 in rat hepatocytes in primary culture, comparing different enzyme activities in microsomal incubations and in intact monolayers. Biochem Pharmacol 40:2525–2534PubMedCrossRef Wortelboer HM, de Kruif CA, van Iersel AA, Falke HE, Noordhoek J, Blaauboer BJ (1990) The isoenzyme pattern of cytochrome p450 in rat hepatocytes in primary culture, comparing different enzyme activities in microsomal incubations and in intact monolayers. Biochem Pharmacol 40:2525–2534PubMedCrossRef
9.
go back to reference Czerwinski M, Opdam P, Madan A, Carroll K, Mudra DR, Gan LL, Luo G, Parkinson A (2002) Analysis of cyp mRNA expression by branched DNA technology. Methods Enzymol 357:170–179PubMedCrossRef Czerwinski M, Opdam P, Madan A, Carroll K, Mudra DR, Gan LL, Luo G, Parkinson A (2002) Analysis of cyp mRNA expression by branched DNA technology. Methods Enzymol 357:170–179PubMedCrossRef
10.
go back to reference Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP, Miwa G, Ni L, Kumar G, McLeod J, Obach RS, Roberts S, Roe A, Shah A, Snikeris F, Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA (2003) The conduct of in vitro and in vivo drug-drug interaction studies: a pharmaceutical research and manufacturers of america (phrma) perspective. Drug Metab Dispos 31:815–832PubMedCrossRef Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP, Miwa G, Ni L, Kumar G, McLeod J, Obach RS, Roberts S, Roe A, Shah A, Snikeris F, Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA (2003) The conduct of in vitro and in vivo drug-drug interaction studies: a pharmaceutical research and manufacturers of america (phrma) perspective. Drug Metab Dispos 31:815–832PubMedCrossRef
11.
go back to reference FDA (2006) Draft guidance for industry—drug interaction studies: study design, data analysis, and implications for dosing and labeling, food and drug administration FDA (2006) Draft guidance for industry—drug interaction studies: study design, data analysis, and implications for dosing and labeling, food and drug administration
12.
go back to reference Ramanathan RK, Trump DL, Eiseman JL, Belani CP, Agarwala SS, Zuhowski EG, Lan J, Potter DM, Ivy SP, Ramalingam S, Brufsky AM, Wong MK, Tutchko S, Egorin MJ (2005) Phase i pharmacokinetic-pharmacodynamic study of 17-(allylamino)-17-demethoxygeldanamycin (17aag, nsc 330507), a novel inhibitor of heat shock protein 90, in patients with refractory advanced cancers. Clin Cancer Res 11:3385–3391PubMedCrossRef Ramanathan RK, Trump DL, Eiseman JL, Belani CP, Agarwala SS, Zuhowski EG, Lan J, Potter DM, Ivy SP, Ramalingam S, Brufsky AM, Wong MK, Tutchko S, Egorin MJ (2005) Phase i pharmacokinetic-pharmacodynamic study of 17-(allylamino)-17-demethoxygeldanamycin (17aag, nsc 330507), a novel inhibitor of heat shock protein 90, in patients with refractory advanced cancers. Clin Cancer Res 11:3385–3391PubMedCrossRef
13.
go back to reference Guo W, Reigan P, Siegel D, Ross D (2008) Enzymatic reduction and glutathione conjugation of benzoquinone ansamycin heat shock protein 90 inhibitors: relevance for toxicity and mechanism of action. Drug Metab Dispos 36:2050–2057PubMedCrossRef Guo W, Reigan P, Siegel D, Ross D (2008) Enzymatic reduction and glutathione conjugation of benzoquinone ansamycin heat shock protein 90 inhibitors: relevance for toxicity and mechanism of action. Drug Metab Dispos 36:2050–2057PubMedCrossRef
14.
go back to reference Premdas PD, Bowers RJ, Forkert PG (2000) Inactivation of hepatic cyp2e1 by an epoxide of diallyl sulfone. J Pharmacol Exp Ther 293:1112–1120PubMed Premdas PD, Bowers RJ, Forkert PG (2000) Inactivation of hepatic cyp2e1 by an epoxide of diallyl sulfone. J Pharmacol Exp Ther 293:1112–1120PubMed
15.
go back to reference Yoshinari K, Kobayashi K, Moore R, Kawamoto T, Negishi M (2003) Identification of the nuclear receptor car:Hsp90 complex in mouse liver and recruitment of protein phosphatase 2a in response to phenobarbital. FEBS Lett 548:17–20PubMedCrossRef Yoshinari K, Kobayashi K, Moore R, Kawamoto T, Negishi M (2003) Identification of the nuclear receptor car:Hsp90 complex in mouse liver and recruitment of protein phosphatase 2a in response to phenobarbital. FEBS Lett 548:17–20PubMedCrossRef
Metadata
Title
In vitro assessment of cytochrome P450 inhibition and induction potential of tanespimycin and its major metabolite, 17-amino-17-demethoxygeldanamycin
Authors
Jinping Gan
Peggy Liu-Kreyche
W. Griffith Humphreys
Publication date
01-01-2012
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 1/2012
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-011-1672-2

Other articles of this Issue 1/2012

Cancer Chemotherapy and Pharmacology 1/2012 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine