Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 2/2014

01-08-2014 | Review Article

Role of pregnane X receptor in chemotherapeutic treatment

Authors: Wei Zhuo, Lei Hu, Jinfeng Lv, Hongbing Wang, Honghao Zhou, Lan Fan

Published in: Cancer Chemotherapy and Pharmacology | Issue 2/2014

Login to get access

Abstract

Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily that differently expresses not only in human normal tissues but also in numerous types of human cancers. PXR can be activated by many endogenous substances and exogenous chemicals, and thus affects chemotherapeutic effects and intervenes drug–drug interactions by regulating its target genes involving drug metabolism and transportation, cell proliferation and apoptosis, and modulating endobiotic homeostasis. Tissue and context-specific regulation of PXR contributes to diverse effects in the treatment for numerous cancers. Genetic variants of PXR lead to intra- and inter-individual differences in the expression and inducibility of PXR, resulting in different responses to chemotherapy in PXR-positive cancers. The purpose of this review is to summarize and discuss the role of PXR in the metabolism and clearance of anticancer drugs. It is also expected that this review will provide insights into PXR-mediated enhancement for chemotherapeutic treatment, prediction of drug–drug interactions and personalized medicine.
Literature
1.
go back to reference Blumberg B, Kang H, Bolado J Jr, Chen H, Craig AG, Moreno TA, Umesono K, Perlmann T, De Robertis EM, Evans RM (1998) BXR, an embryonic orphan nuclear receptor activated by a novel class of endogenous benzoate metabolites. Genes Dev 12:1269–1277PubMedCentralPubMed Blumberg B, Kang H, Bolado J Jr, Chen H, Craig AG, Moreno TA, Umesono K, Perlmann T, De Robertis EM, Evans RM (1998) BXR, an embryonic orphan nuclear receptor activated by a novel class of endogenous benzoate metabolites. Genes Dev 12:1269–1277PubMedCentralPubMed
2.
go back to reference Blumberg B, Sabbagh W Jr, Juguilon H, Bolado J Jr, van Meter CM, Ong ES, Evans RM (1998) SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev 12:3195–3205PubMedCentralPubMed Blumberg B, Sabbagh W Jr, Juguilon H, Bolado J Jr, van Meter CM, Ong ES, Evans RM (1998) SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev 12:3195–3205PubMedCentralPubMed
3.
go back to reference Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB, Willson TM, Zetterström RH, Perlmann T, Lehmann JM (1998) An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 92:73–82PubMed Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB, Willson TM, Zetterström RH, Perlmann T, Lehmann JM (1998) An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 92:73–82PubMed
4.
go back to reference Nishimura M, Naito S, Yokoi T (2004) Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet 19:135–149PubMed Nishimura M, Naito S, Yokoi T (2004) Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet 19:135–149PubMed
5.
go back to reference Pondugula SR, Mani S (2013) Pregnane xenobiotic receptor in cancer pathogenesis and therapeutic response. Cancer Lett 328:1–9PubMedCentralPubMed Pondugula SR, Mani S (2013) Pregnane xenobiotic receptor in cancer pathogenesis and therapeutic response. Cancer Lett 328:1–9PubMedCentralPubMed
6.
go back to reference Hu M, Fan L, Zhou HH, Tomlinson B (2012) Theranostics meets traditional Chinese medicine: rational prediction of drug-herb interactions. Expert Rev Mol Diagn 12:815–830PubMed Hu M, Fan L, Zhou HH, Tomlinson B (2012) Theranostics meets traditional Chinese medicine: rational prediction of drug-herb interactions. Expert Rev Mol Diagn 12:815–830PubMed
7.
go back to reference Zhang B, Xie W, Krasowski MD (2008) PXR: a xenobiotic receptor of diverse function implicated in pharmacogenetics. Pharmacogenomics 9:1695–1709PubMedCentralPubMed Zhang B, Xie W, Krasowski MD (2008) PXR: a xenobiotic receptor of diverse function implicated in pharmacogenetics. Pharmacogenomics 9:1695–1709PubMedCentralPubMed
8.
go back to reference Orans J, Teotico DG, Redinbo MR (2005) The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges. Mol Endocrinol 19:2891–2900PubMed Orans J, Teotico DG, Redinbo MR (2005) The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges. Mol Endocrinol 19:2891–2900PubMed
9.
go back to reference Squires EJ, Sueyoshi T, Negishi M (2004) Cytoplasmic localization of pregnane X receptor and ligand-dependent nuclear translocation in mouse liver. J Biol Chem 279:49307–49314PubMed Squires EJ, Sueyoshi T, Negishi M (2004) Cytoplasmic localization of pregnane X receptor and ligand-dependent nuclear translocation in mouse liver. J Biol Chem 279:49307–49314PubMed
10.
go back to reference Horwitz KB, Jackson TA, Bain DL, Richer JK, Takimoto GS, Tung L (1996) Nuclear receptor coactivators and corepressors. Mol Endocrinol 10:1167–1177PubMed Horwitz KB, Jackson TA, Bain DL, Richer JK, Takimoto GS, Tung L (1996) Nuclear receptor coactivators and corepressors. Mol Endocrinol 10:1167–1177PubMed
11.
go back to reference Chen Y, Nie D (2009) Pregnane X receptor and its potential role in drug resistance in cancer treatment. Recent Pat Anticancer Drug Discov 4:19–27PubMed Chen Y, Nie D (2009) Pregnane X receptor and its potential role in drug resistance in cancer treatment. Recent Pat Anticancer Drug Discov 4:19–27PubMed
12.
go back to reference McKenna NJ, Lanz RB, O’Malley BW (1999) Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 20:321–344PubMed McKenna NJ, Lanz RB, O’Malley BW (1999) Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 20:321–344PubMed
13.
go back to reference Zhou C, Verma S, Blumberg B (2009) The steroid and xenobiotic receptor (SXR), beyond xenobiotic metabolism. Nucl Recept Signal 7:e001PubMedCentralPubMed Zhou C, Verma S, Blumberg B (2009) The steroid and xenobiotic receptor (SXR), beyond xenobiotic metabolism. Nucl Recept Signal 7:e001PubMedCentralPubMed
14.
go back to reference Chai X, Zeng S, Xie W (2013) Nuclear receptors PXR and CAR: implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab Toxicol 9:253–266PubMed Chai X, Zeng S, Xie W (2013) Nuclear receptors PXR and CAR: implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab Toxicol 9:253–266PubMed
15.
go back to reference Chen Y, Tang Y, Guo C, Wang J, Boral D, Nie D (2012) Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters. Biochem Pharmacol 83:1112–1126PubMedCentralPubMed Chen Y, Tang Y, Guo C, Wang J, Boral D, Nie D (2012) Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters. Biochem Pharmacol 83:1112–1126PubMedCentralPubMed
16.
go back to reference Rosenfeld JM, Vargas R Jr, Xie W, Evans RM (2003) Genetic profiling defines the xenobiotic gene network controlled by the nuclear receptor pregnane X receptor. Mol Endocrinol 17:1268–1282PubMed Rosenfeld JM, Vargas R Jr, Xie W, Evans RM (2003) Genetic profiling defines the xenobiotic gene network controlled by the nuclear receptor pregnane X receptor. Mol Endocrinol 17:1268–1282PubMed
17.
go back to reference Maglich JM, Stoltz CM, Goodwin B, Hawkins-Brown D, Moore JT, Kliewer SA (2002) Nuclear pregnane X receptor and constitutive androstane receptor regulate overlapping but distinct sets of genes involved in xenobiotic detoxification. Mol Pharmacol 62:638–646PubMed Maglich JM, Stoltz CM, Goodwin B, Hawkins-Brown D, Moore JT, Kliewer SA (2002) Nuclear pregnane X receptor and constitutive androstane receptor regulate overlapping but distinct sets of genes involved in xenobiotic detoxification. Mol Pharmacol 62:638–646PubMed
18.
go back to reference Xie W, Yeuh MF, Radominska-Pandya A, Saini SP, Negishi Y, Bottroff BS, Cabrera GY, Tukey RH, Evans RM (2003) Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor. Proc Natl Acad Sci USA 100:4150–4155PubMedCentralPubMed Xie W, Yeuh MF, Radominska-Pandya A, Saini SP, Negishi Y, Bottroff BS, Cabrera GY, Tukey RH, Evans RM (2003) Control of steroid, heme, and carcinogen metabolism by nuclear pregnane X receptor and constitutive androstane receptor. Proc Natl Acad Sci USA 100:4150–4155PubMedCentralPubMed
19.
go back to reference Miki Y, Suzuki T, Kitada K, Yabuki N, Shibuya R, Moriya T, Ishida T, Ohuchi N, Blumberg B, Sasano H (2006) Expression of the steroid and xenobiotic receptor and its possible target gene, organic anion transporting polypeptide-A, in human breast carcinoma. Cancer Res 66:535–542PubMed Miki Y, Suzuki T, Kitada K, Yabuki N, Shibuya R, Moriya T, Ishida T, Ohuchi N, Blumberg B, Sasano H (2006) Expression of the steroid and xenobiotic receptor and its possible target gene, organic anion transporting polypeptide-A, in human breast carcinoma. Cancer Res 66:535–542PubMed
20.
go back to reference Guzelian J, Barwick JL, Hunter L, Phang TL, Quattrochi LC, Guzelian PS (2006) Identification of genes controlled by the pregnane X receptor by microarray analysis of mRNAs from pregnenolone 16alpha-carbonitrile-treated rats. Toxicol Sci 94:379–387PubMedCentralPubMed Guzelian J, Barwick JL, Hunter L, Phang TL, Quattrochi LC, Guzelian PS (2006) Identification of genes controlled by the pregnane X receptor by microarray analysis of mRNAs from pregnenolone 16alpha-carbonitrile-treated rats. Toxicol Sci 94:379–387PubMedCentralPubMed
21.
go back to reference Cheng J, Shah YM, Gonzalez FJ (2012) Pregnane X receptor as a target for treatment of inflammatory bowel disorders. Trends Pharmacol Sci 33:323–330PubMedCentralPubMed Cheng J, Shah YM, Gonzalez FJ (2012) Pregnane X receptor as a target for treatment of inflammatory bowel disorders. Trends Pharmacol Sci 33:323–330PubMedCentralPubMed
22.
go back to reference Masuyama H, Nakatsukasa H, Takamoto N, Hiramatsu Y (2007) Down-regulation of pregnane X receptor contributes to cell growth inhibition and apoptosis by anticancer agents in endometrial cancer cells. Mol Pharmacol 72:1045–1053PubMed Masuyama H, Nakatsukasa H, Takamoto N, Hiramatsu Y (2007) Down-regulation of pregnane X receptor contributes to cell growth inhibition and apoptosis by anticancer agents in endometrial cancer cells. Mol Pharmacol 72:1045–1053PubMed
23.
go back to reference Zucchini N, de Sousa G, Bailly-Maitre B, Gugenheim J, Bars R, Lemaire G, Rahmani R (2005) Regulation of Bcl-2 and Bcl-xL anti-apoptotic protein expression by nuclear receptor PXR in primary cultures of human and rat hepatocytes. Biochim Biophys Acta 1745:48–58PubMed Zucchini N, de Sousa G, Bailly-Maitre B, Gugenheim J, Bars R, Lemaire G, Rahmani R (2005) Regulation of Bcl-2 and Bcl-xL anti-apoptotic protein expression by nuclear receptor PXR in primary cultures of human and rat hepatocytes. Biochim Biophys Acta 1745:48–58PubMed
24.
go back to reference Gupta D, Venkatesh M, Wang H, Kim S, Sinz M, Goldberg GL, Whitney K, Longley C, Mani S (2008) Expanding the roles for pregnane X receptor in cancer: proliferation and drug resistance in ovarian cancer. Clin Cancer Res 14:5332–5340PubMed Gupta D, Venkatesh M, Wang H, Kim S, Sinz M, Goldberg GL, Whitney K, Longley C, Mani S (2008) Expanding the roles for pregnane X receptor in cancer: proliferation and drug resistance in ovarian cancer. Clin Cancer Res 14:5332–5340PubMed
25.
go back to reference Zhang J, Kuehl P, Green ED, Touchman JW, Watkins PB, Daly A, Hall SD, Maurel P, Relling M, Brimer C, Yasuda K, Wrighton SA, Hancock M, Kim RB, Strom S, Thummel K, Russell CG, Hudson JR Jr, Schuetz EG, Boguski MS (2001) The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics 11:555–572PubMed Zhang J, Kuehl P, Green ED, Touchman JW, Watkins PB, Daly A, Hall SD, Maurel P, Relling M, Brimer C, Yasuda K, Wrighton SA, Hancock M, Kim RB, Strom S, Thummel K, Russell CG, Hudson JR Jr, Schuetz EG, Boguski MS (2001) The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics 11:555–572PubMed
26.
go back to reference King CR, Xiao M, Yu J, Minton MR, Addleman NJ, Van Booven DJ, Kwok PY, McLeod HL, Marsh S (2007) Identification of NR1I2 genetic variation using resequencing. Eur J Clin Pharmacol 63:547–554PubMed King CR, Xiao M, Yu J, Minton MR, Addleman NJ, Van Booven DJ, Kwok PY, McLeod HL, Marsh S (2007) Identification of NR1I2 genetic variation using resequencing. Eur J Clin Pharmacol 63:547–554PubMed
27.
go back to reference Lamba J, Lamba V, Strom S, Venkataramanan R, Schuetz E (2008) Novel single nucleotide polymorphisms in the promoter and intron 1 of human pregnane X receptor/NR1I2 and their association with CYP3A4 expression. Drug Metab Dispos 36:169–181PubMed Lamba J, Lamba V, Strom S, Venkataramanan R, Schuetz E (2008) Novel single nucleotide polymorphisms in the promoter and intron 1 of human pregnane X receptor/NR1I2 and their association with CYP3A4 expression. Drug Metab Dispos 36:169–181PubMed
28.
go back to reference Koyano S, Kurose K, Ozawa S, Saeki M, Nakajima Y, Hasegawa R, Komamura K, Ueno K, Kamakura S, Nakajima T, Saito H, Kimura H, Goto Y, Saitoh O, Katoh M, Ohnuma T, Kawai M, Sugai K, Ohtsuki T, Suzuki C, Minami N, Saito Y, Sawada J (2002) Eleven novel single nucleotide polymorphisms in the NR1I2 (PXR) gene, four of which induce non-synonymous amino acid alterations. Drug Metab Pharmacokinet 17:561–565PubMed Koyano S, Kurose K, Ozawa S, Saeki M, Nakajima Y, Hasegawa R, Komamura K, Ueno K, Kamakura S, Nakajima T, Saito H, Kimura H, Goto Y, Saitoh O, Katoh M, Ohnuma T, Kawai M, Sugai K, Ohtsuki T, Suzuki C, Minami N, Saito Y, Sawada J (2002) Eleven novel single nucleotide polymorphisms in the NR1I2 (PXR) gene, four of which induce non-synonymous amino acid alterations. Drug Metab Pharmacokinet 17:561–565PubMed
29.
go back to reference Koyano S, Kurose K, Saito Y, Ozawa S, Hasegawa R, Komamura K, Ueno K, Kamakura S, Kitakaze M, Nakajima T, Matsumoto K, Akasawa A, Saito H, Sawada J (2004) Functional characterization of four naturally occurring variants of human pregnane X receptor (PXR): one variant causes dramatic loss of both DNA binding activity and the transactivation of the CYP3A4 promoter/enhancer region. Drug Metab Dispos 32:149–154PubMed Koyano S, Kurose K, Saito Y, Ozawa S, Hasegawa R, Komamura K, Ueno K, Kamakura S, Kitakaze M, Nakajima T, Matsumoto K, Akasawa A, Saito H, Sawada J (2004) Functional characterization of four naturally occurring variants of human pregnane X receptor (PXR): one variant causes dramatic loss of both DNA binding activity and the transactivation of the CYP3A4 promoter/enhancer region. Drug Metab Dispos 32:149–154PubMed
30.
go back to reference Lim YP, Liu CH, Shyu LJ, Huang JD (2005) Functional characterization of a novel polymorphism of pregnane X receptor, Q158 K, in Chinese subjects. Pharmacogenet Genomics 15:337–341PubMed Lim YP, Liu CH, Shyu LJ, Huang JD (2005) Functional characterization of a novel polymorphism of pregnane X receptor, Q158 K, in Chinese subjects. Pharmacogenet Genomics 15:337–341PubMed
31.
go back to reference Hustert E, Zibat A, Presecan-Siedel E, Eiselt R, Mueller R, Fuss C, Brehm I, Brinkmann U, Eichelbaum M, Wojnowski L, Burk O (2001) Natural protein variants of pregnane X receptor with altered transactivation activity toward CYP3A4. Drug Metab Dispos 29:1454–1459PubMed Hustert E, Zibat A, Presecan-Siedel E, Eiselt R, Mueller R, Fuss C, Brehm I, Brinkmann U, Eichelbaum M, Wojnowski L, Burk O (2001) Natural protein variants of pregnane X receptor with altered transactivation activity toward CYP3A4. Drug Metab Dispos 29:1454–1459PubMed
32.
go back to reference Uno Y, Sakamoto Y, Yoshida K, Hasegawa T, Hasegawa Y, Koshino T, Inoue I (2003) Characterization of six base pair deletion in the putative HNF1-binding site of human PXR promoter. J Hum Genet 48:594–597PubMed Uno Y, Sakamoto Y, Yoshida K, Hasegawa T, Hasegawa Y, Koshino T, Inoue I (2003) Characterization of six base pair deletion in the putative HNF1-binding site of human PXR promoter. J Hum Genet 48:594–597PubMed
33.
go back to reference Lamba V, Yasuda K, Lamba JK, Assem M, Davila J, Strom S, Schuetz EG (2004) PXR (NR1I2): splice variants in human tissues, including brain, and identification of neurosteroids and nicotine as PXR activators. Toxicol Appl Pharmacol 199:251–265PubMed Lamba V, Yasuda K, Lamba JK, Assem M, Davila J, Strom S, Schuetz EG (2004) PXR (NR1I2): splice variants in human tissues, including brain, and identification of neurosteroids and nicotine as PXR activators. Toxicol Appl Pharmacol 199:251–265PubMed
34.
go back to reference Keightley MC (1998) Steroid receptor isoforms: exception or rule? Mol Cell Endocrinol 137:1–5PubMed Keightley MC (1998) Steroid receptor isoforms: exception or rule? Mol Cell Endocrinol 137:1–5PubMed
35.
go back to reference Gardner-Stephen D, Heydel JM, Goyal A, Lu Y, Xie W, Lindblom T, Mackenzie P, Radominska-Pandya A (2004) Human PXR variants and their differential effects on the regulation of human UDP-glucuronosyltransferase gene expression. Drug Metab Dispos 32:340–347PubMedCentralPubMed Gardner-Stephen D, Heydel JM, Goyal A, Lu Y, Xie W, Lindblom T, Mackenzie P, Radominska-Pandya A (2004) Human PXR variants and their differential effects on the regulation of human UDP-glucuronosyltransferase gene expression. Drug Metab Dispos 32:340–347PubMedCentralPubMed
36.
go back to reference Fukuen S, Fukuda T, Matsuda H, Sumida A, Yamamoto I, Inaba T, Azuma J (2002) Identification of the novel splicing variants for the hPXR in human livers. Biochem Biophys Res Commun 298:433–438PubMed Fukuen S, Fukuda T, Matsuda H, Sumida A, Yamamoto I, Inaba T, Azuma J (2002) Identification of the novel splicing variants for the hPXR in human livers. Biochem Biophys Res Commun 298:433–438PubMed
37.
go back to reference Mensah-Osman EJ, Thomas DG, Tabb MM, Larios JM, Hughes DP, Giordano TJ, Lizyness ML, Rae JM, Blumberg B, Hollenberg PF, Baker LH (2007) Expression levels and activation of a PXR variant are directly related to drug resistance in osteosarcoma cell lines. Cancer 109:957–965PubMedCentralPubMed Mensah-Osman EJ, Thomas DG, Tabb MM, Larios JM, Hughes DP, Giordano TJ, Lizyness ML, Rae JM, Blumberg B, Hollenberg PF, Baker LH (2007) Expression levels and activation of a PXR variant are directly related to drug resistance in osteosarcoma cell lines. Cancer 109:957–965PubMedCentralPubMed
38.
go back to reference Lin YS, Yasuda K, Assem M, Cline C, Barber J, Li CW, Kholodovych V, Ai N, Chen JD, Welsh WJ, Ekins S, Schuetz EG (2009) The major human pregnane X receptor (PXR) splice variant, PXR.2, exhibits significantly diminished ligand-activated transcriptional regulation. Drug Metab Dispos 37:1295–1304PubMedCentralPubMed Lin YS, Yasuda K, Assem M, Cline C, Barber J, Li CW, Kholodovych V, Ai N, Chen JD, Welsh WJ, Ekins S, Schuetz EG (2009) The major human pregnane X receptor (PXR) splice variant, PXR.2, exhibits significantly diminished ligand-activated transcriptional regulation. Drug Metab Dispos 37:1295–1304PubMedCentralPubMed
39.
go back to reference Tompkins LM, Sit TL, Wallace AD (2008) Unique transcription start sites and distinct promoter regions differentiate the pregnane X receptor (PXR) isoforms PXR 1 and PXR 2. Drug Metab Dispos 36:923–929PubMed Tompkins LM, Sit TL, Wallace AD (2008) Unique transcription start sites and distinct promoter regions differentiate the pregnane X receptor (PXR) isoforms PXR 1 and PXR 2. Drug Metab Dispos 36:923–929PubMed
40.
go back to reference di Masi A, De Marinis E, Ascenzi P, Marino M (2009) Nuclear receptors CAR and PXR: Molecular, functional, and biomedical aspects. Mol Aspects Med 30:297–343PubMed di Masi A, De Marinis E, Ascenzi P, Marino M (2009) Nuclear receptors CAR and PXR: Molecular, functional, and biomedical aspects. Mol Aspects Med 30:297–343PubMed
41.
go back to reference Koutsounas I, Patsouris E, Theocharis S (2013) Pregnane X receptor and human malignancy. Histol Histopathol 28:405–420PubMed Koutsounas I, Patsouris E, Theocharis S (2013) Pregnane X receptor and human malignancy. Histol Histopathol 28:405–420PubMed
42.
go back to reference Ma X, Idle JR, Gonzalez FJ (2008) The pregnane X receptor: from bench to bedside. Expert Opin Drug Metab Toxicol 4:895–908PubMedCentralPubMed Ma X, Idle JR, Gonzalez FJ (2008) The pregnane X receptor: from bench to bedside. Expert Opin Drug Metab Toxicol 4:895–908PubMedCentralPubMed
43.
44.
go back to reference Koutsounas I, Theocharis S, Patsouris E, Giaginis C (2013) Pregnane X receptor (PXR) at the crossroads of human metabolism and disease. Curr Drug Metab 14:341–350PubMed Koutsounas I, Theocharis S, Patsouris E, Giaginis C (2013) Pregnane X receptor (PXR) at the crossroads of human metabolism and disease. Curr Drug Metab 14:341–350PubMed
45.
go back to reference Qiao E, Ji M, Wu J, Ma R, Zhang X, He Y, Zha Q, Song X, Zhu LW, Tang J (2013) Expression of the PXR gene in various types of cancer and drug resistance. Oncol Lett 5:1093–1100PubMedCentralPubMed Qiao E, Ji M, Wu J, Ma R, Zhang X, He Y, Zha Q, Song X, Zhu LW, Tang J (2013) Expression of the PXR gene in various types of cancer and drug resistance. Oncol Lett 5:1093–1100PubMedCentralPubMed
46.
go back to reference Handschin C, Meyer UA (2003) Induction of drug metabolism: the role of nuclear receptors. Pharmacol Rev 55:649–673PubMed Handschin C, Meyer UA (2003) Induction of drug metabolism: the role of nuclear receptors. Pharmacol Rev 55:649–673PubMed
47.
go back to reference Gollamudi R, Gupta D, Goel S, Mani S (2008) Novel orphan nuclear receptors–coregulator interactions controlling anti-cancer drug metabolism. Curr Drug Metab 9:611–613PubMed Gollamudi R, Gupta D, Goel S, Mani S (2008) Novel orphan nuclear receptors–coregulator interactions controlling anti-cancer drug metabolism. Curr Drug Metab 9:611–613PubMed
48.
go back to reference Lamba J, Lamba V, Schuetz E (2005) Genetic variants of PXR (NR1I2) and CAR (NR1I3) and their implications in drug metabolism and pharmacogenetics. Curr Drug Metab 6:369–383PubMed Lamba J, Lamba V, Schuetz E (2005) Genetic variants of PXR (NR1I2) and CAR (NR1I3) and their implications in drug metabolism and pharmacogenetics. Curr Drug Metab 6:369–383PubMed
49.
go back to reference Willson TM, Kliewer SA (2002) PXR, CAR and drug metabolism. Nat Rev Drug Discov 1:259–266PubMed Willson TM, Kliewer SA (2002) PXR, CAR and drug metabolism. Nat Rev Drug Discov 1:259–266PubMed
50.
go back to reference Verma S, Tabb MM, Blumberg B (2009) Activation of the steroid and xenobiotic receptor, SXR, induces apoptosis in breast cancer cells. BMC Cancer 9:3PubMedCentralPubMed Verma S, Tabb MM, Blumberg B (2009) Activation of the steroid and xenobiotic receptor, SXR, induces apoptosis in breast cancer cells. BMC Cancer 9:3PubMedCentralPubMed
51.
go back to reference Wang H, Venkatesh M, Li H, Goetz R, Mukherjee S, Biswas A, Zhu L, Kaubisch A, Wang L, Pullman J, Whitney K, Kuro-o M, Roig AI, Shay JW, Mohammadi M, Mani S (2011) Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice. J Clin Invest 121:3220–3232PubMedCentralPubMed Wang H, Venkatesh M, Li H, Goetz R, Mukherjee S, Biswas A, Zhu L, Kaubisch A, Wang L, Pullman J, Whitney K, Kuro-o M, Roig AI, Shay JW, Mohammadi M, Mani S (2011) Pregnane X receptor activation induces FGF19-dependent tumor aggressiveness in humans and mice. J Clin Invest 121:3220–3232PubMedCentralPubMed
52.
go back to reference Zhou J, Liu M, Zhai Y, Xie W (2008) The antiapoptotic role of pregnane X receptor in human colon cancer cells. Mol Endocrinol 22:868–880PubMedCentralPubMed Zhou J, Liu M, Zhai Y, Xie W (2008) The antiapoptotic role of pregnane X receptor in human colon cancer cells. Mol Endocrinol 22:868–880PubMedCentralPubMed
53.
go back to reference Ouyang N, Ke S, Eagleton N, Xie Y, Chen G, Laffins B, Yao H, Zhou B, Tian Y (2010) Pregnane X receptor suppresses proliferation and tumorigenicity of colon cancer cells. Br J Cancer 102:1753–1761PubMedCentralPubMed Ouyang N, Ke S, Eagleton N, Xie Y, Chen G, Laffins B, Yao H, Zhou B, Tian Y (2010) Pregnane X receptor suppresses proliferation and tumorigenicity of colon cancer cells. Br J Cancer 102:1753–1761PubMedCentralPubMed
54.
go back to reference Elias A, Wu J, Chen T (2013) Tumor suppressor protein p53 negatively regulates human pregnane X receptor activity. Mol Pharmacol 83:1229–1236PubMedCentralPubMed Elias A, Wu J, Chen T (2013) Tumor suppressor protein p53 negatively regulates human pregnane X receptor activity. Mol Pharmacol 83:1229–1236PubMedCentralPubMed
55.
go back to reference Swinnen JV, Brusselmans K, Verhoeven G (2006) Increased lipogenesis in cancer cells: new players, novel targets. Curr Opin Clin Nutr Metab Care 9:358–365PubMed Swinnen JV, Brusselmans K, Verhoeven G (2006) Increased lipogenesis in cancer cells: new players, novel targets. Curr Opin Clin Nutr Metab Care 9:358–365PubMed
56.
go back to reference Zhou J, Zhai Y, Mu Y, Gong H, Uppal H, Toma D, Ren S, Evans RM, Xie W (2006) A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J Biol Chem 281:15013–15020PubMedCentralPubMed Zhou J, Zhai Y, Mu Y, Gong H, Uppal H, Toma D, Ren S, Evans RM, Xie W (2006) A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J Biol Chem 281:15013–15020PubMedCentralPubMed
57.
go back to reference Zhou J, Febbraio M, Wada T, Zhai Y, Kuruba R, He J, Lee JH, Khadem S, Ren S, Li S, Silverstein RL, Xie W (2008) Hepatic fatty acid transporter Cd36 is a common target of LXR, PXR, and PPARgamma in promoting steatosis. Gastroenterology 134:556–567PubMed Zhou J, Febbraio M, Wada T, Zhai Y, Kuruba R, He J, Lee JH, Khadem S, Ren S, Li S, Silverstein RL, Xie W (2008) Hepatic fatty acid transporter Cd36 is a common target of LXR, PXR, and PPARgamma in promoting steatosis. Gastroenterology 134:556–567PubMed
58.
go back to reference Gong H, Singh SV, Singh SP, Mu Y, Lee JH, Saini SP, Toma D, Ren S, Kagan VE, Day BW, Zimniak P, Xie W (2006) Orphan nuclear receptor pregnane X receptor sensitizes oxidative stress responses in transgenic mice and cancerous cells. Mol Endocrinol 20:279–290PubMed Gong H, Singh SV, Singh SP, Mu Y, Lee JH, Saini SP, Toma D, Ren S, Kagan VE, Day BW, Zimniak P, Xie W (2006) Orphan nuclear receptor pregnane X receptor sensitizes oxidative stress responses in transgenic mice and cancerous cells. Mol Endocrinol 20:279–290PubMed
59.
go back to reference Ma MK, McLeod HL (2003) Lessons learned from the irinotecan metabolic pathway. Curr Med Chem 10:41–49PubMed Ma MK, McLeod HL (2003) Lessons learned from the irinotecan metabolic pathway. Curr Med Chem 10:41–49PubMed
60.
go back to reference Haaz MC, Rivory L, Riché C, Vernillet L, Robert J (1998) Metabolism of irinotecan (CPT-11) by human hepatic microsomes: participation of cytochrome P-450 3A and drug interactions. Cancer Res 58:468–472PubMed Haaz MC, Rivory L, Riché C, Vernillet L, Robert J (1998) Metabolism of irinotecan (CPT-11) by human hepatic microsomes: participation of cytochrome P-450 3A and drug interactions. Cancer Res 58:468–472PubMed
61.
go back to reference Mathijssen RH, van Alphen RJ, Verweij J, Loos WJ, Nooter K, Stoter G, Sparreboom A (2001) Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 7:2182–2194PubMed Mathijssen RH, van Alphen RJ, Verweij J, Loos WJ, Nooter K, Stoter G, Sparreboom A (2001) Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 7:2182–2194PubMed
62.
go back to reference Chu XY, Kato Y, Sugiyama Y (1999) Possible involvement of P-glycoprotein in biliary excretion of CPT-11 in rats. Drug Metab Dispos 27:440–441PubMed Chu XY, Kato Y, Sugiyama Y (1999) Possible involvement of P-glycoprotein in biliary excretion of CPT-11 in rats. Drug Metab Dispos 27:440–441PubMed
63.
go back to reference Chu XY, Suzuki H, Ueda K, Kato Y, Akiyama S, Sugiyama Y (1999) Active efflux of CPT-11 and its metabolites in human KB-derived cell lines. J Pharmacol Exp Ther 288:735–741PubMed Chu XY, Suzuki H, Ueda K, Kato Y, Akiyama S, Sugiyama Y (1999) Active efflux of CPT-11 and its metabolites in human KB-derived cell lines. J Pharmacol Exp Ther 288:735–741PubMed
64.
go back to reference Raynal C, Pascussi JM, Leguelinel G, Breuker C, Kantar J, Lallemant B, Poujol S, Bonnans C, Joubert D, Hollande F, Lumbroso S, Brouillet JP, Evrard A (2010) Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Mol Cancer 9:46PubMedCentralPubMed Raynal C, Pascussi JM, Leguelinel G, Breuker C, Kantar J, Lallemant B, Poujol S, Bonnans C, Joubert D, Hollande F, Lumbroso S, Brouillet JP, Evrard A (2010) Pregnane X Receptor (PXR) expression in colorectal cancer cells restricts irinotecan chemosensitivity through enhanced SN-38 glucuronidation. Mol Cancer 9:46PubMedCentralPubMed
65.
go back to reference Basseville A, Preisser L, de Carné Trécesson S, Boisdron-Celle M, Gamelin E, Coqueret O, Morel A (2011) Irinotecan induces steroid and xenobiotic receptor (SXR) signaling to detoxification pathway in colon cancer cells. Mol Cancer 10:80PubMedCentralPubMed Basseville A, Preisser L, de Carné Trécesson S, Boisdron-Celle M, Gamelin E, Coqueret O, Morel A (2011) Irinotecan induces steroid and xenobiotic receptor (SXR) signaling to detoxification pathway in colon cancer cells. Mol Cancer 10:80PubMedCentralPubMed
66.
go back to reference Yonemori K, Takeda Y, Toyota E, Kobayashi N, Kudo K (2004) Potential interactions between irinotecan and rifampin in a patient with small-cell lung cancer. Int J Clin Oncol 9:206–209PubMed Yonemori K, Takeda Y, Toyota E, Kobayashi N, Kudo K (2004) Potential interactions between irinotecan and rifampin in a patient with small-cell lung cancer. Int J Clin Oncol 9:206–209PubMed
67.
go back to reference Wentworth JM, Agostini M, Love J, Schwabe JW, Chatterjee VK (2000) St John’s wort, a herbal antidepressant, activates the steroid X receptor. J Endocrinol 166:R11–R16PubMed Wentworth JM, Agostini M, Love J, Schwabe JW, Chatterjee VK (2000) St John’s wort, a herbal antidepressant, activates the steroid X receptor. J Endocrinol 166:R11–R16PubMed
68.
go back to reference Mathijssen RH, Verweij J, de Bruijn P, Loos WJ, Sparreboom A (2002) Effects of St. John’s wort on irinotecan metabolism. J Natl Cancer Inst 94:1247–1249PubMed Mathijssen RH, Verweij J, de Bruijn P, Loos WJ, Sparreboom A (2002) Effects of St. John’s wort on irinotecan metabolism. J Natl Cancer Inst 94:1247–1249PubMed
69.
go back to reference Hennessy M, Kelleher D, Spiers JP, Barry M, Kavanagh P, Back D, Mulcahy F, Feely J (2002) St Johns wort increases expression of P-glycoprotein: implications for drug interactions. Br J Clin Pharmacol 53:75–82PubMedCentralPubMed Hennessy M, Kelleher D, Spiers JP, Barry M, Kavanagh P, Back D, Mulcahy F, Feely J (2002) St Johns wort increases expression of P-glycoprotein: implications for drug interactions. Br J Clin Pharmacol 53:75–82PubMedCentralPubMed
70.
go back to reference Dehal SS, Kupfer D (1997) CYP2D6 catalyzes tamoxifen 4-hydroxylation in human liver. Cancer Res 57:3402–3406PubMed Dehal SS, Kupfer D (1997) CYP2D6 catalyzes tamoxifen 4-hydroxylation in human liver. Cancer Res 57:3402–3406PubMed
71.
go back to reference Jacolot F, Simon I, Dreano Y, Beaune P, Riche C, Berthou F (1991) Identification of the cytochrome P450 IIIA family as the enzymes involved in the N-demethylation of tamoxifen in human liver microsomes. Biochem Pharmacol 41:1911–1919PubMed Jacolot F, Simon I, Dreano Y, Beaune P, Riche C, Berthou F (1991) Identification of the cytochrome P450 IIIA family as the enzymes involved in the N-demethylation of tamoxifen in human liver microsomes. Biochem Pharmacol 41:1911–1919PubMed
72.
go back to reference Teft WA, Mansell SE, Kim RB (2011) Endoxifen, the active metabolite of tamoxifen, is a substrate of the efflux transporter P-glycoprotein (multidrug resistance 1). Drug Metab Dispos 39:558–562PubMed Teft WA, Mansell SE, Kim RB (2011) Endoxifen, the active metabolite of tamoxifen, is a substrate of the efflux transporter P-glycoprotein (multidrug resistance 1). Drug Metab Dispos 39:558–562PubMed
73.
go back to reference Choi HK, Yang JW, Roh SH, Han CY, Kang KW (2007) Induction of multidrug resistance associated protein 2 in tamoxifen-resistant breast cancer cells. Endocr Relat Cancer 14:293–303PubMed Choi HK, Yang JW, Roh SH, Han CY, Kang KW (2007) Induction of multidrug resistance associated protein 2 in tamoxifen-resistant breast cancer cells. Endocr Relat Cancer 14:293–303PubMed
74.
go back to reference Chen Y, Tang Y, Chen S, Nie D (2009) Regulation of drug resistance by human pregnane X receptor in breast cancer. Cancer Biol Ther 8:1265–1272PubMedCentralPubMed Chen Y, Tang Y, Chen S, Nie D (2009) Regulation of drug resistance by human pregnane X receptor in breast cancer. Cancer Biol Ther 8:1265–1272PubMedCentralPubMed
75.
go back to reference Meyer zu Schwabedissen HE, Tirona RG, Yip CS, Ho RH, Kim RB (2008) Interplay between the nuclear receptor pregnane X receptor and the uptake transporter organic anion transporter polypeptide 1A2 selectively enhances estrogen effects in breast cancer. Cancer Res 68:9338–9347PubMedCentralPubMed Meyer zu Schwabedissen HE, Tirona RG, Yip CS, Ho RH, Kim RB (2008) Interplay between the nuclear receptor pregnane X receptor and the uptake transporter organic anion transporter polypeptide 1A2 selectively enhances estrogen effects in breast cancer. Cancer Res 68:9338–9347PubMedCentralPubMed
76.
go back to reference Desai PB, Nallani SC, Sane RS, Moore LB, Goodwin BJ, Buckley DJ, Buckley AR (2002) Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4-hydroxytamoxifen. Drug Metab Dispos 30:608–612PubMed Desai PB, Nallani SC, Sane RS, Moore LB, Goodwin BJ, Buckley DJ, Buckley AR (2002) Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4-hydroxytamoxifen. Drug Metab Dispos 30:608–612PubMed
77.
go back to reference Harmsen S, Meijerman I, Febus CL, Maas-Bakker RF, Beijnen JH, Schellens JH (2010) PXR-mediated induction of P-glycoprotein by anticancer drugs in a human colon adenocarcinoma-derived cell line. Cancer Chemother Pharmacol 66:765–771PubMedCentralPubMed Harmsen S, Meijerman I, Febus CL, Maas-Bakker RF, Beijnen JH, Schellens JH (2010) PXR-mediated induction of P-glycoprotein by anticancer drugs in a human colon adenocarcinoma-derived cell line. Cancer Chemother Pharmacol 66:765–771PubMedCentralPubMed
78.
go back to reference Nagaoka R, Iwasaki T, Rokutanda N, Takeshita A, Koibuchi Y, Horiguchi J, Shimokawa N, Iino Y, Morishita Y, Koibuchi N (2006) Tamoxifen activates CYP3A4 and MDR1 genes through steroid and xenobiotic receptor in breast cancer cells. Endocrine 30:261–268PubMed Nagaoka R, Iwasaki T, Rokutanda N, Takeshita A, Koibuchi Y, Horiguchi J, Shimokawa N, Iino Y, Morishita Y, Koibuchi N (2006) Tamoxifen activates CYP3A4 and MDR1 genes through steroid and xenobiotic receptor in breast cancer cells. Endocrine 30:261–268PubMed
79.
go back to reference Sane RS, Buckley DJ, Buckley AR, Nallani SC, Desai PB (2008) Role of human pregnane X receptor in tamoxifen- and 4-hydroxytamoxifen-mediated CYP3A4 induction in primary human hepatocytes and LS174T cells. Drug Metab Dispos 36:946–954PubMed Sane RS, Buckley DJ, Buckley AR, Nallani SC, Desai PB (2008) Role of human pregnane X receptor in tamoxifen- and 4-hydroxytamoxifen-mediated CYP3A4 induction in primary human hepatocytes and LS174T cells. Drug Metab Dispos 36:946–954PubMed
80.
go back to reference Huizing MT, Misser VH, Pieters RC, ten Bokkel Huinink WW, Veenhof CH, Vermorken JB, Pinedo HM, Beijnen JH (1995) Taxanes: a new class of antitumor agents. Cancer Invest 13:381–404PubMed Huizing MT, Misser VH, Pieters RC, ten Bokkel Huinink WW, Veenhof CH, Vermorken JB, Pinedo HM, Beijnen JH (1995) Taxanes: a new class of antitumor agents. Cancer Invest 13:381–404PubMed
81.
go back to reference Schiff PB, Fant J, Horwitz SB (1979) Promotion of microtubule assembly in vitro by taxol. Nature 277:665–667PubMed Schiff PB, Fant J, Horwitz SB (1979) Promotion of microtubule assembly in vitro by taxol. Nature 277:665–667PubMed
82.
go back to reference Harris JW, Rahman A, Kim BR, Guengerich FP, Collins JM (1994) Metabolism of taxol by human hepatic microsomes and liver slices: participation of cytochrome P450 3A4 and an unknown P450 enzyme. Cancer Res 54:4026–4035PubMed Harris JW, Rahman A, Kim BR, Guengerich FP, Collins JM (1994) Metabolism of taxol by human hepatic microsomes and liver slices: participation of cytochrome P450 3A4 and an unknown P450 enzyme. Cancer Res 54:4026–4035PubMed
83.
go back to reference Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, Harris JW (1994) Selective biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 2C8. Cancer Res 54:5543–5546PubMed Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, Harris JW (1994) Selective biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 2C8. Cancer Res 54:5543–5546PubMed
84.
go back to reference van Asperen J, van Tellingen O, Sparreboom A, Schinkel AH, Borst P, Nooijen WJ, Beijnen JH (1997) Enhanced oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833. Br J Cancer 76:1181–1183PubMedCentralPubMed van Asperen J, van Tellingen O, Sparreboom A, Schinkel AH, Borst P, Nooijen WJ, Beijnen JH (1997) Enhanced oral bioavailability of paclitaxel in mice treated with the P-glycoprotein blocker SDZ PSC 833. Br J Cancer 76:1181–1183PubMedCentralPubMed
85.
go back to reference Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK, Borst P, Nooijen WJ, Beijnen JH, van Tellingen O (1997) Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci USA 94:2031–2035PubMedCentralPubMed Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK, Borst P, Nooijen WJ, Beijnen JH, van Tellingen O (1997) Limited oral bioavailability and active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in the intestine. Proc Natl Acad Sci USA 94:2031–2035PubMedCentralPubMed
86.
go back to reference Chen Y, Tang Y, Wang MT, Zeng S, Nie D (2007) Human pregnane X receptor and resistance to chemotherapy in prostate cancer. Cancer Res 67:10361–10367PubMed Chen Y, Tang Y, Wang MT, Zeng S, Nie D (2007) Human pregnane X receptor and resistance to chemotherapy in prostate cancer. Cancer Res 67:10361–10367PubMed
87.
go back to reference Masuyama H, Suwaki N, Tateishi Y, Nakatsukasa H, Segawa T, Hiramatsu Y (2005) The pregnane X receptor regulates gene expression in a ligand- and promoter-selective fashion. Mol Endocrinol 19:1170–1180PubMed Masuyama H, Suwaki N, Tateishi Y, Nakatsukasa H, Segawa T, Hiramatsu Y (2005) The pregnane X receptor regulates gene expression in a ligand- and promoter-selective fashion. Mol Endocrinol 19:1170–1180PubMed
88.
go back to reference Nallani SC, Goodwin B, Maglich JM, Buckley DJ, Buckley AR, Desai PB (2003) Induction of cytochrome P450 3A by paclitaxel in mice: pivotal role of the nuclear xenobiotic receptor, pregnane X receptor. Drug Metab Dispos 31:681–684PubMed Nallani SC, Goodwin B, Maglich JM, Buckley DJ, Buckley AR, Desai PB (2003) Induction of cytochrome P450 3A by paclitaxel in mice: pivotal role of the nuclear xenobiotic receptor, pregnane X receptor. Drug Metab Dispos 31:681–684PubMed
89.
go back to reference Synold TW, Dussault I, Forman BM (2001) The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med 7:584–590PubMed Synold TW, Dussault I, Forman BM (2001) The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med 7:584–590PubMed
90.
go back to reference Gewirtz DA (1999) A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol 57:727–741PubMed Gewirtz DA (1999) A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol 57:727–741PubMed
91.
go back to reference Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L (2004) Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 56:185–229PubMed Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L (2004) Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 56:185–229PubMed
92.
go back to reference Lee HJ, Lee MG (1999) Effects of dexamethasone on the pharmacokinetics of adriamycin after intravenous administration to rats. Res Commun Mol Pathol Pharmacol 105:87–96PubMed Lee HJ, Lee MG (1999) Effects of dexamethasone on the pharmacokinetics of adriamycin after intravenous administration to rats. Res Commun Mol Pathol Pharmacol 105:87–96PubMed
93.
go back to reference Lal S, Mahajan A, Chen WN, Chowbay B (2010) Pharmacogenetics of target genes across doxorubicin disposition pathway: a review. Curr Drug Metab 11:115–128PubMed Lal S, Mahajan A, Chen WN, Chowbay B (2010) Pharmacogenetics of target genes across doxorubicin disposition pathway: a review. Curr Drug Metab 11:115–128PubMed
94.
go back to reference Mensah-Osman E, Lin H-L, Reinke D, Hollenberg P, Baker L (2005) Ecteinascidin-743 is a potent inhibitor of P450 3A4 enzyme and accumulates cytoplasmic PXR to inhibit transcription of P450 3A4 and MDR1: implications for the enhancement of cytotoxicity to chemotherapeutic agents in osteosarcoma. J Clin Oncol (Meeting Abstracts) 23(16s):9026 Mensah-Osman E, Lin H-L, Reinke D, Hollenberg P, Baker L (2005) Ecteinascidin-743 is a potent inhibitor of P450 3A4 enzyme and accumulates cytoplasmic PXR to inhibit transcription of P450 3A4 and MDR1: implications for the enhancement of cytotoxicity to chemotherapeutic agents in osteosarcoma. J Clin Oncol (Meeting Abstracts) 23(16s):9026
95.
go back to reference Sandanaraj E, Lal S, Selvarajan V, Ooi LL, Wong ZW, Wong NS, Ang PC, Lee EJ, Chowbay B (2008) PXR pharmacogenetics: association of haplotypes with hepatic CYP3A4 and ABCB1 messenger RNA expression and doxorubicin clearance in Asian breast cancer patients. Clin Cancer Res 14:7116–7126PubMed Sandanaraj E, Lal S, Selvarajan V, Ooi LL, Wong ZW, Wong NS, Ang PC, Lee EJ, Chowbay B (2008) PXR pharmacogenetics: association of haplotypes with hepatic CYP3A4 and ABCB1 messenger RNA expression and doxorubicin clearance in Asian breast cancer patients. Clin Cancer Res 14:7116–7126PubMed
96.
go back to reference Noble RL (1990) The discovery of the vinca alkaloids–chemotherapeutic agents against cancer. Biochem Cell Biol 68:1344–1351PubMed Noble RL (1990) The discovery of the vinca alkaloids–chemotherapeutic agents against cancer. Biochem Cell Biol 68:1344–1351PubMed
97.
go back to reference Jordan MA, Wilson L (2004) Microtubules as a target for anticancer drugs. Nat Rev Cancer 4:253–265PubMed Jordan MA, Wilson L (2004) Microtubules as a target for anticancer drugs. Nat Rev Cancer 4:253–265PubMed
98.
go back to reference Levêque D, Wihlm J, Jehl F (1996) Pharmacology of Catharanthus alkaloids. Bull Cancer 83:176–186PubMed Levêque D, Wihlm J, Jehl F (1996) Pharmacology of Catharanthus alkaloids. Bull Cancer 83:176–186PubMed
99.
go back to reference Levêque D, Jehl F (2007) Molecular pharmacokinetics of catharanthus (vinca) alkaloids. J Clin Pharmacol 47:579–588PubMed Levêque D, Jehl F (2007) Molecular pharmacokinetics of catharanthus (vinca) alkaloids. J Clin Pharmacol 47:579–588PubMed
100.
go back to reference Gruol DJ, King MN, Kuehne ME (2002) Evidence for the locations of distinct steroid and Vinca alkaloid interaction domains within the murine mdr1b P-glycoprotein. Mol Pharmacol 62:1238–1248PubMed Gruol DJ, King MN, Kuehne ME (2002) Evidence for the locations of distinct steroid and Vinca alkaloid interaction domains within the murine mdr1b P-glycoprotein. Mol Pharmacol 62:1238–1248PubMed
101.
go back to reference Huang R, Murry DJ, Kolwankar D, Hall SD, Foster DR (2006) Vincristine transcriptional regulation of efflux drug transporters in carcinoma cell lines. Biochem Pharmacol 71:1695–1704PubMed Huang R, Murry DJ, Kolwankar D, Hall SD, Foster DR (2006) Vincristine transcriptional regulation of efflux drug transporters in carcinoma cell lines. Biochem Pharmacol 71:1695–1704PubMed
102.
go back to reference Smith NF, Mani S, Schuetz EG, Yasuda K, Sissung TM, Bates SE, Figg WD, Sparreboom A (2010) Induction of CYP3A4 by vinblastine: role of the nuclear receptor NR1I2. Ann Pharmacother 44:1709–1717PubMedCentralPubMed Smith NF, Mani S, Schuetz EG, Yasuda K, Sissung TM, Bates SE, Figg WD, Sparreboom A (2010) Induction of CYP3A4 by vinblastine: role of the nuclear receptor NR1I2. Ann Pharmacother 44:1709–1717PubMedCentralPubMed
103.
go back to reference Hong WK, Itri LM (1994) Retinoids and human cancer. In: Sporn MB, Roberts AB, Goodman DS (eds) The retinoids. Biology, chemistry and medicine, 2nd edn. Raven Press Ltd, New York, pp 597–630 Hong WK, Itri LM (1994) Retinoids and human cancer. In: Sporn MB, Roberts AB, Goodman DS (eds) The retinoids. Biology, chemistry and medicine, 2nd edn. Raven Press Ltd, New York, pp 597–630
104.
go back to reference Levin AA, Sturzenbecker LJ, Kazmer S, Bosakowski T, Huselton C, Allenby G, Speck J, Kratzeisen C, Rosenberger M, Lovey A et al (1992) 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXR alpha. Nature 355:359–361PubMed Levin AA, Sturzenbecker LJ, Kazmer S, Bosakowski T, Huselton C, Allenby G, Speck J, Kratzeisen C, Rosenberger M, Lovey A et al (1992) 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXR alpha. Nature 355:359–361PubMed
105.
go back to reference Nagy L, Thomázy VA, Shipley GL, Fésüs L, Lamph W, Heyman RA, Chandraratna RA, Davies PJ (1995) Activation of retinoid X receptors induces apoptosis in HL-60 cell lines. Mol Cell Biol 15:3540–3551PubMedCentralPubMed Nagy L, Thomázy VA, Shipley GL, Fésüs L, Lamph W, Heyman RA, Chandraratna RA, Davies PJ (1995) Activation of retinoid X receptors induces apoptosis in HL-60 cell lines. Mol Cell Biol 15:3540–3551PubMedCentralPubMed
106.
go back to reference Elstner E, Müller C, Koshizuka K, Williamson EA, Park D, Asou H, Shintaku P, Said JW, Heber D, Koeffler HP (1998) Ligands for peroxisome proliferator-activated receptorgamma and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice. Proc Natl Acad Sci USA 95:8806–8811PubMedCentralPubMed Elstner E, Müller C, Koshizuka K, Williamson EA, Park D, Asou H, Shintaku P, Said JW, Heber D, Koeffler HP (1998) Ligands for peroxisome proliferator-activated receptorgamma and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice. Proc Natl Acad Sci USA 95:8806–8811PubMedCentralPubMed
107.
go back to reference Muindi JR, Frankel SR, Huselton C, DeGrazia F, Garland WA, Young CW, Warrell RP Jr (1992) Clinical pharmacology of oral all-trans retinoic acid in patients with acute promyelocytic leukemia. Cancer Res 52:2138–2142PubMed Muindi JR, Frankel SR, Huselton C, DeGrazia F, Garland WA, Young CW, Warrell RP Jr (1992) Clinical pharmacology of oral all-trans retinoic acid in patients with acute promyelocytic leukemia. Cancer Res 52:2138–2142PubMed
108.
go back to reference Marill J, Cresteil T, Lanotte M, Chabot GG (2000) Identification of human cytochrome P450s involved in the formation of all-trans-retinoic acid principal metabolites. Mol Pharmacol 58:1341–1348PubMed Marill J, Cresteil T, Lanotte M, Chabot GG (2000) Identification of human cytochrome P450s involved in the formation of all-trans-retinoic acid principal metabolites. Mol Pharmacol 58:1341–1348PubMed
109.
go back to reference Wang T, Ma X, Krausz KW, Idle JR, Gonzalez FJ (2008) Role of pregnane X receptor in control of all-trans retinoic acid (ATRA) metabolism and its potential contribution to ATRA resistance. J Pharmacol Exp Ther 324:674–684PubMedCentralPubMed Wang T, Ma X, Krausz KW, Idle JR, Gonzalez FJ (2008) Role of pregnane X receptor in control of all-trans retinoic acid (ATRA) metabolism and its potential contribution to ATRA resistance. J Pharmacol Exp Ther 324:674–684PubMedCentralPubMed
110.
go back to reference Moore MJ (1991) Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet 20:194–208PubMed Moore MJ (1991) Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet 20:194–208PubMed
111.
go back to reference Dechant KL, Brogden RN, Pilkington T, Faulds D (1991) Ifosfamide/mesna. A review of its antineoplastic activity, pharmacokinetic properties and therapeutic efficacy in cancer. Drugs 42:428–467PubMed Dechant KL, Brogden RN, Pilkington T, Faulds D (1991) Ifosfamide/mesna. A review of its antineoplastic activity, pharmacokinetic properties and therapeutic efficacy in cancer. Drugs 42:428–467PubMed
112.
go back to reference Sladek NE (1988) Metabolism of oxazaphosphorines. Pharmacol Ther 37:301–355PubMed Sladek NE (1988) Metabolism of oxazaphosphorines. Pharmacol Ther 37:301–355PubMed
113.
go back to reference Zhang Jing, Tian Quan, Zhou Shu-Feng (2006) Clinical pharmacology of cyclophosphamide and ifosfamide. Curr Drug Ther 1:55–84 Zhang Jing, Tian Quan, Zhou Shu-Feng (2006) Clinical pharmacology of cyclophosphamide and ifosfamide. Curr Drug Ther 1:55–84
114.
go back to reference Chang TK, Yu L, Maurel P, Waxman DJ (1997) Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 57:1946–1954PubMed Chang TK, Yu L, Maurel P, Waxman DJ (1997) Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 57:1946–1954PubMed
115.
go back to reference Lee W, Lockhart AC, Kim RB, Rothenberg ML (2005) Cancer pharmacogenomics: powerful tools in cancer chemotherapy and drug development. Oncologist 10:104–111PubMed Lee W, Lockhart AC, Kim RB, Rothenberg ML (2005) Cancer pharmacogenomics: powerful tools in cancer chemotherapy and drug development. Oncologist 10:104–111PubMed
116.
go back to reference Chabner BA, Roberts TG Jr (2005) Timeline: chemotherapy and the war on cancer. Nat Rev Cancer 5:65–72PubMed Chabner BA, Roberts TG Jr (2005) Timeline: chemotherapy and the war on cancer. Nat Rev Cancer 5:65–72PubMed
117.
go back to reference Harmsen S, Meijerman I, Beijnen JH, Schellens JH (2007) The role of nuclear receptors in pharmacokinetic drug–drug interactions in oncology. Cancer Treat Rev 33:369–380PubMed Harmsen S, Meijerman I, Beijnen JH, Schellens JH (2007) The role of nuclear receptors in pharmacokinetic drug–drug interactions in oncology. Cancer Treat Rev 33:369–380PubMed
118.
go back to reference Meijerman I, Beijnen JH, Schellens JH (2006) Herb–drug interactions in oncology: focus on mechanisms of induction. Oncologist 11:742–752PubMed Meijerman I, Beijnen JH, Schellens JH (2006) Herb–drug interactions in oncology: focus on mechanisms of induction. Oncologist 11:742–752PubMed
119.
go back to reference Biswas A, Mani S, Redinbo MR, Krasowski MD, Li H, Ekins S (2009) Elucidating the ‘Jekyll and Hyde’ nature of PXR: the case for discovering antagonists or allosteric antagonists. Pharm Res 26:1807–1815PubMedCentralPubMed Biswas A, Mani S, Redinbo MR, Krasowski MD, Li H, Ekins S (2009) Elucidating the ‘Jekyll and Hyde’ nature of PXR: the case for discovering antagonists or allosteric antagonists. Pharm Res 26:1807–1815PubMedCentralPubMed
120.
go back to reference Robbins D, Chen T (2014) Tissue-specific regulation of pregnane X receptor in cancer development and therapy. Cell Biosci 4:17PubMed Robbins D, Chen T (2014) Tissue-specific regulation of pregnane X receptor in cancer development and therapy. Cell Biosci 4:17PubMed
Metadata
Title
Role of pregnane X receptor in chemotherapeutic treatment
Authors
Wei Zhuo
Lei Hu
Jinfeng Lv
Hongbing Wang
Honghao Zhou
Lan Fan
Publication date
01-08-2014
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 2/2014
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-014-2494-9

Other articles of this Issue 2/2014

Cancer Chemotherapy and Pharmacology 2/2014 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