Accelerated Communication INDUCTION OF CYTOCHROME P450 3A4 IN PRIMARY HUMAN HEPATOCYTES AND ACTIVATION OF THE HUMAN PREGNANE X RECEPTOR BY TAMOXIFEN AND 4-HYDROXYTAMOXIFEN

نویسندگان

  • PANKAJ B. DESAI
  • SRIKANTH C. NALLANI
  • RUCHA S. SANE
  • LINDA B. MOORE
  • BRYAN J. GOODWIN
  • DONNA J. BUCKLEY
  • ARTHUR R. BUCKLEY
چکیده

Tamoxifen is a widely utilized antiestrogen in the treatment and chemoprevention of breast cancer. Clinical studies document that tamoxifen administration markedly enhances the systemic elimination of other drugs. Additionally, tamoxifen enhances its own clearance following repeated dosing. The mechanisms that underlie these clinically important events remain unresolved. Here, we report that tamoxifen and its metabolite 4-hydroxytamoxifen markedly induce cytochrome P450 3A4, a drug-metabolizing enzyme of central importance, in primary cultures of human hepatocytes. Tamoxifen and 4-hydroxytamoxifen (1–10 M) significantly increased the CYP3A4 expression and activity (measured as the rate of testosterone 6 -hydroxylation). Maximal induction was achieved at the 5 M level. At this level, tamoxifen and 4-hydroxytamoxifen caused a 1.5to 3.3-fold (mean, 2.1-fold) and 3.4to 17-fold (mean, 7.5-fold) increase in the CYP3A4 activity, respectively. In comparison, rifampicin treatment resulted in a 6to 16fold (mean, 10.5-fold) increase. We also observed corresponding increase in the CYP3A4 immunoreactive protein and mRNA levels. Furthermore, tamoxifen and 4-hydroxytamoxifen efficaciously activated the human pregnane X receptor (hPXR; also known as the steroid xenobiotic receptor), a key regulator of CYP3A4 expression. The efficacy of tamoxifen and 4-hydroxytamoxifen relative to rifampicin for hPXR activation was 30 and 60%, respectively. Our results indicate that the mechanism of tamoxifen-mediated alteration in drug clearance pathways in humans may involve CYP3A4 induction by the parent drug and/or its metabolite. Furthermore, the CYP3A4 induction may be a result of hPXR activation. These findings have important implications for optimizing the use of tamoxifen and in the development of newer antiestrogens. Tamoxifen, a nonsteroidal triphenylethylene, is currently the endocrine therapeutic agent of choice for all stages of breast cancer. It was also recently approved for use as a chemopreventive agent in women with high risk of contracting this disease in the future. Despite its well documented beneficial effects, tamoxifen use is associated with several major problems including serious drug-drug interactions. Several clinical trials indicate that tamoxifen has the propensity to alter the drug elimination pathway(s), resulting in markedly reduced plasma levels of coadministered compounds. In this regard, recently completed clinical trials indicate that tamoxifen reduced the plasma levels of aromatase inhibitors letrozole and anastrozole by 37 and 27%, respectively (Dowsett et al., 1999; ATAC Trialists’ Group, 2001). Such drug-drug interactions are of special concern with tamoxifen since numerous women are required to take tamoxifen daily for an extended time period and as such are likely to be simultaneously exposed to many drugs and nutraceuticals. Another consequence of tamoxifen-induced changes in drug disposition is that tamoxifen pharmacokinetics exhibit time-dependent changes. There are marked differences in the single dose versus steady-state (when multiple doses have been taken) pharmacokinetics of tamoxifen (Etienne et al., 1989). The relative abundance of tamoxifen (as a fraction of the combined levels of tamoxifen and its metabolites) is lower at steady state compared with that following single dose. This is accompanied with increased relative abundance of tamoxifen metabolites at steady state. The extent of tamoxifen conversion to its metabolites has serious implications for its efficacy and toxicity since some of its metabolites are potent antiestrogens whereas some others are impliSupported by a grant from the American Cancer Society (Ohio Division), Women’s Health Program, University of Cincinnati Medical Center, the National Institutes of Health (DK53452), and the American Institute for Cancer Research. 1 Abbreviations used are: P450, cytochrome P450; hPXR, human pregnane X receptor; XREM, xenobiotic response element module; DMSO, dimethyl sulfoxide; HuH7, human hepatocellular carcinoma; HPLC, high-performance liquid

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4-hydroxytamoxifen.

Tamoxifen is a widely utilized antiestrogen in the treatment and chemoprevention of breast cancer. Clinical studies document that tamoxifen administration markedly enhances the systemic elimination of other drugs. Additionally, tamoxifen enhances its own clearance following repeated dosing. The mechanisms that underlie these clinically important events remain unresolved. Here, we report that ta...

متن کامل

Role of human pregnane X receptor in tamoxifen- and 4-hydroxytamoxifen-mediated CYP3A4 induction in primary human hepatocytes and LS174T cells.

Previously we observed that the antiestrogens tamoxifen and 4-hydroxytamoxifen (4OHT) induce CYP3A4 in primary human hepatocytes and activate human pregnane X receptor (PXR) in cell-based reporter assays. Given the complex cross-talk between nuclear receptors, tissue-specific expression of CYP3A4, and the potential for tamoxifen and 4OHT to interact with a myriad of receptors, this study was un...

متن کامل

alpha-Hydroxytamoxifen, a metabolite of tamoxifen with exceptionally high DNA-binding activity in rat hepatocytes.

It has been proposed that the antiestrogen tamoxifen induces liver tumors in rats and genotoxic effects in vitro through metabolic activation involving, initially, alpha-hydroxylation of the ethyl group. To test this hypothesis, the extent of DNA adduct formation in primary rat hepatocytes treated with tamoxifen and alpha-hydroxytamoxifen was investigated. Hepatocytes from female Fischer F-344 ...

متن کامل

Sex differences in the activation of tamoxifen to DNA binding species in rat liver in vivo and in rat hepatocytes in vitro: role of sulfotransferase induction.

Previous work has indicated that metabolic activation of tamoxifen in rat liver cells involves cytochrome P450-mediated alpha-hydroxylation, followed by sulfate ester formation, mediated by hydroxysteroid sulfotransferase a (rHSTa), a member of the SULT2A subfamily, which efficiently metabolizes dehydroepiandrosterone. Because it is known that the expression of rHSTa and other SULT2A forms is s...

متن کامل

Activation of protein kinase B/cAkt in hepatocytes is sufficient for the induction of expression of the gene encoding glucokinase.

Inhibitors of signalling pathways were used to dissect the mechanism of insulin action on expression of the gene encoding glucokinase in cultured rat hepatocytes. Wortmannin and LY 294002 completely prevented the insulin-induced increase in glucokinase mRNA seen in unhibited cells, indicating that the phosphoinositide 3-kinase module has a key role. A ligand inducible protein kinase B (PKB, als...

متن کامل

ذخیره در منابع من


  با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

ثبت نام

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

عنوان ژورنال:

دوره   شماره 

صفحات  -

تاریخ انتشار 2002