1
|
Wang H, Chu F, Zhang XF, Zhang P, Li LX, Zhuang YL, Niu XF, He X, Li ZJ, Bai Y, Mao D, Liu ZW, Zhang DL, Li BA. TPX2 enhances the transcription factor activation of PXR and enhances the resistance of hepatocellular carcinoma cells to antitumor drugs. Cell Death Dis 2023; 14:64. [PMID: 36707511 PMCID: PMC9883482 DOI: 10.1038/s41419-022-05537-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 01/29/2023]
Abstract
The pregnane X receptor (PXR) is an important regulator of hepatocellular carcinoma cellular resistance to antitumor drugs. Activation of PXR was modulated by the co-regulators. The target protein for the Xenopus plus end-directed kinesin-like protein (Xklp2) known as TPX2 that was previously considered as a tubulin regulator, also functions as the regulator of some transcription factors and pro-oncogenes in human malignances. However, the actions of TPX2 on PXR and HCC cells are still unclear. In the present study, our results demonstrate that the high expression of endogenous mRNA level of TPX2 not only correlated with the poor prognosis of advanced HCC patients who received sorafenib treatment but also with expression of PXR's downstream genes, cyp3a4 and/or mdr-1. Results from luciferase and real-time polymerase chain reaction (qPCR) showed that TPX2 leads to enhancement of the transcription factor activation of PXR. Protein-protein interactions between PXR and TPX2 were identified using co-immunoprecipitation. Mechanically, overexpression of TPX2 led to enhancement of PXR recruitment to its downstream gene cyp3a4's promoter region (the PXRE region) or enhancer region (the XREM region). Treatment of HCC cells with paclitaxel, a microtubule promoter, led to enhancement of the effects of TPX2, whereas vincristine, a microtubule depolymerizing agent caused a decrease in TPX2-associated effects. TPX2 was found to cause acceleration of the metabolism or clearance of sorafenib, a typical tyrosine kinase inhibitor (TKI) in HCC cells and in turn led to the resistance to sorafenib by HCC cells. By establishing novel actions of TXP2 on PXR in HCC cells, the results indicate that TPX2 could be considered a promising therapeutic target to enhance HCC cells sensitivity to antitumor drugs.
Collapse
Affiliation(s)
- Hongbo Wang
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Fang Chu
- Department of Emergency, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Xiao-Feng Zhang
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Peng Zhang
- Department of Urology, Chinese People's Liberation Army (PLA) General Hospital/Chinese PLA Medical Academy, Beijing, 100853, China
| | - Li-Xin Li
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Yun-Long Zhuang
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Xiao-Feng Niu
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Xi He
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Zhi-Jie Li
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Ying Bai
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China
| | - Da Mao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100191, China
- Division of Chemical Metrology and Analytical Science, National Institute of Metrology, Beijing, 100029, China
| | - Zhen-Wen Liu
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China.
| | - Da-Li Zhang
- Senior Department of Hepatology, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China.
| | - Bo-An Li
- Clinical Laboratory, the Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100039, China.
| |
Collapse
|
2
|
Rachmale M, Rajput N, Jadav T, Sahu AK, Tekade RK, Sengupta P. Implication of metabolomics and transporter modulation based strategies to minimize multidrug resistance and enhance site-specific bioavailability: a needful consideration toward modern anticancer drug discovery. Drug Metab Rev 2022; 54:101-119. [PMID: 35254954 DOI: 10.1080/03602532.2022.2048007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Induction of drug-metabolizing enzymes and efflux transporters (DMET) through activation of pregnane x receptor (PXR) is the primary factor involved in almost all bioavailability and drug resistance-related problems of anticancer drugs. PXR is a transcriptional regulator of many metabolizing enzymes and efflux transporters proteins like p-glycoprotein (p-gp), multidrug resistant protein 1 and 2 (MRP 1 and 2), and breast cancer resistant protein (BCRP), etc. Several anticancer drugs are potent activators of PXR receptors and can modulate the gene expression of DMET proteins. Involvement of anticancer drugs in transcriptional regulation of DMET can prompt increased metabolism and efflux of their own or other co-administered drugs, which leads to poor site-specific bioavailability and increased drug resistance. In this review, we have discussed several novel strategies to evade drug-induced PXR activation and p-gp efflux including assessment of PXR ligand and p-gp substrate at early stages of drug discovery. Additionally, we have critically discussed the chemical structure and drug delivery-based approaches to avoid PXR binding and inhibit the p-gp activity of the drugs at their target sites.
Collapse
Affiliation(s)
- Megha Rachmale
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Niraj Rajput
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Tarang Jadav
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Amit Kumar Sahu
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Gandhinagar, Gujarat, India
| |
Collapse
|
3
|
Creusot N, Gassiot M, Alaterre E, Chiavarina B, Grimaldi M, Boulahtouf A, Toporova L, Gerbal-Chaloin S, Daujat-Chavanieu M, Matheux A, Rahmani R, Gongora C, Evrard A, Pourquier P, Balaguer P. The Anti-Cancer Drug Dabrafenib Is a Potent Activator of the Human Pregnane X Receptor. Cells 2020; 9:cells9071641. [PMID: 32650447 PMCID: PMC7407672 DOI: 10.3390/cells9071641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 12/22/2022] Open
Abstract
The human pregnane X receptor (hPXR) is activated by a large set of endogenous and exogenous compounds and plays a critical role in the control of detoxifying enzymes and transporters regulating liver and gastrointestinal drug metabolism and clearance. hPXR is also involved in both the development of multidrug resistance and enhanced cancer cells aggressiveness. Moreover, its unintentional activation by pharmaceutical drugs can mediate drug–drug interactions and cause severe adverse events. In that context, the potential of the anticancer BRAF inhibitor dabrafenib suspected to activate hPXR and the human constitutive androstane receptor (hCAR) has not been thoroughly investigated yet. Using different reporter cellular assays, we demonstrate that dabrafenib can activate hPXR as efficiently as its reference agonist SR12813, whereas it does not activate mouse or zebrafish PXR nor hCAR. We also showed that dabrafenib binds to recombinant hPXR, induces the expression of hPXR responsive genes in colon LS174T-hPXR cancer cells and human hepatocytes and finally increases the proliferation in LS174T-hPXR cells. Our study reveals that by using a panel of different cellular techniques it is possible to improve the assessment of hPXR agonist activity for new developed drugs.
Collapse
Affiliation(s)
- Nicolas Creusot
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Matthieu Gassiot
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Elina Alaterre
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Barbara Chiavarina
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Abdelhay Boulahtouf
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Lucia Toporova
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Sabine Gerbal-Chaloin
- IRMB, Université de Montpellier, INSERM, CHU Montpellier, 34090 Montpellier, France; (S.G.-C.); (M.D.-C.)
| | - Martine Daujat-Chavanieu
- IRMB, Université de Montpellier, INSERM, CHU Montpellier, 34090 Montpellier, France; (S.G.-C.); (M.D.-C.)
| | - Alice Matheux
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Roger Rahmani
- INRA UMR 1331 TOXALIM, 06560 Sophia Antipolis, France;
| | - Céline Gongora
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Alexandre Evrard
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
| | - Philippe Pourquier
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
- Correspondence: (P.P.); (P.B.); Tel.: +33-467613787 (P.P.); +33-467612409 (P.B.); Fax: +33-467613787 (P.P.); +33-467612337 (P.B.)
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Université de Montpellier, ICM, 34298 Montpellier, France; (N.C.); (M.G.); (E.A.); (B.C.); (M.G.); (A.B.); (L.T.); (A.M.); (C.G.); (A.E.)
- Correspondence: (P.P.); (P.B.); Tel.: +33-467613787 (P.P.); +33-467612409 (P.B.); Fax: +33-467613787 (P.P.); +33-467612337 (P.B.)
| |
Collapse
|
4
|
Mutation of a single amino acid of pregnane X receptor switches an antagonist to agonist by altering AF-2 helix positioning. Cell Mol Life Sci 2020; 78:317-335. [PMID: 32232515 DOI: 10.1007/s00018-020-03505-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 10/24/2022]
Abstract
Pregnane X receptor (PXR) is activated by chemicals to transcriptionally regulate drug disposition and possibly decrease drug efficacy and increase resistance, suggesting therapeutic value for PXR antagonists. We previously reported the antagonist SPA70 and its analog SJB7, which unexpectedly is an agonist. Here, we describe another unexpected observation: mutating a single residue (W299A) within the PXR ligand-binding domain converts SPA70 to an agonist. After characterizing wild-type and W299A PXR activity profiles, we used molecular dynamics simulations to reveal that in wild-type PXR, agonists stabilize the activation function 2 (AF-2) helix in an "inward" position, but SPA70 displaces the AF-2. In W299A, however, SPA70 stabilizes the AF-2 "inward", like agonists. We validated our model by predicting the antagonist SJC2 to be a W299A agonist, which was confirmed experimentally. Our work correlates previously unobserved ligand-induced conformational changes to PXR cellular activity and, for the first time, reveals how PXR antagonists work.
Collapse
|
5
|
Lin W, Bwayi M, Wu J, Li Y, Chai SC, Huber AD, Chen T. CITCO Directly Binds to and Activates Human Pregnane X Receptor. Mol Pharmacol 2020; 97:180-190. [PMID: 31882411 PMCID: PMC6978709 DOI: 10.1124/mol.119.118513] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
The xenobiotic receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are activated by structurally diverse chemicals to regulate the expression of target genes, and they have overlapping regulation in terms of ligands and target genes. Receptor-selective agonists are, therefore, critical for studying the overlapping function of PXR and CAR. An early effort identified 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) as a selective human CAR (hCAR) agonist, and this has since been widely used to distinguish the function of hCAR from that of human PXR (hPXR). The selectivity was demonstrated in a green monkey kidney cell line, CV-1, in which CITCO displayed >100-fold selectivity for hCAR over hPXR. However, whether the selectivity observed in CV-1 cells also represented CITCO activity in liver cell models was not hitherto investigated. In this study, we showed that CITCO: 1) binds directly to hPXR; 2) activates hPXR in HepG2 cells, with activation being blocked by an hPXR-specific antagonist, SPA70; 3) does not activate mouse PXR; 4) depends on tryptophan-299 to activate hPXR; 5) recruits steroid receptor coactivator 1 to hPXR; 6) activates hPXR in HepaRG cell lines even when hCAR is knocked out; and 7) activates hPXR in primary human hepatocytes. Together, these data indicate that CITCO binds directly to the hPXR ligand-binding domain to activate hPXR. As CITCO has been widely used, its confirmation as a dual agonist for hCAR and hPXR is important for appropriately interpreting existing data and designing future experiments to understand the regulation of hPXR and hCAR. SIGNIFICANCE STATEMENT: The results of this study demonstrate that 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) is a dual agonist for human constitutive androstane receptor (hCAR) and human pregnane X receptor (hPXR). As CITCO has been widely used to activate hCAR, and hPXR and hCAR have distinct and overlapping biological functions, these results highlight the value of receptor-selective agonists and the importance of appropriately interpreting data in the context of receptor selectivity of such agonists.
Collapse
Affiliation(s)
- Wenwei Lin
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Monicah Bwayi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yongtao Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Andrew D Huber
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
6
|
Mohandas S, Vairappan B. Pregnane X receptor activation by its natural ligand Ginkgolide-A improves tight junction proteins expression and attenuates bacterial translocation in cirrhosis. Chem Biol Interact 2020; 315:108891. [PMID: 31697926 DOI: 10.1016/j.cbi.2019.108891] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/02/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Pregnane X receptor (PXR) is a ligand-activated transcription factor and nuclear receptor expressed ubiquitously along gut-liver-axis. Inflammatory bowel disorders have been reported to implicate PXR in maintaining tight junction (TJ) integrity and countering inflammation. However, the hepatoprotective role of PXR activation in soothing bacterial translocation in liver cirrhosis has not been explored. Ginkgolide A (GA), a terpene trilactone from Ginkgo Biloba extract, is a natural ligand of rodent and human PXR. This study aims to investigate the effect of GA in activating PXR and improving associated tight junction integrity and reducing bacterial translocation in gut-liver axis of CCl4 induced cirrhosis model. METHODS Swiss albino mice were administered with CCl4 (0.5 ml/kg body weight, i.p) in corn oil for 12 weeks at an interval of two times a week. Following ascites induction, mice were randomized & administered 100 mg/kg body weight of GA through oral gavage for 2 weeks. At termination, blood, gut and liver tissues were collected for biochemical and molecular studies. RESULTS When compared to naïve mice, protein expression of hepatic and small intestinal PXR, CYP3A, ZO-1 and occludin were found to be significantly (p < 0.01) decreased in CCl4 induced cirrhotic mice. Treatment with GA to cirrhotic mice significantly (p < 0.05) induced the expression of both hepatic and small intestinal PXR, CYP3A, ZO-1 and Occludin. Furthermore, increased (p < 0.01) hepatic and small intestinal NFκB was observed in CCl4 induced cirrhotic mice that was significantly (p < 0.05) lowered following GA treatment. Over expression of TLR4/MyD88/NFκB axis and its downstream pro-inflammatory mediators TNF-α, IL6 and IFN-γ were observed in CCl4 induced mice, and these indices were abrogated significantly after GA treatment. Furthermore, significantly increased plasma levels of bacterial translocation markers LBP and procalcitonin were found in CCl4 mice, which were reduced significantly (p < 0.05 & p < 0.0001) after GA treatment. CONCLUSION In conclusion, our data supports the hypothesis that, GA treatment to CCl4 induced cirrhotic mice, activated hepatic and small intestinal PXR and diminished inflammation, thereby improving tight junction integrity and attenuating bacterial translocation.
Collapse
Affiliation(s)
- Sundhar Mohandas
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India
| | - Balasubramaniyan Vairappan
- Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India.
| |
Collapse
|
7
|
Bhagyaraj E, Ahuja N, Kumar S, Tiwari D, Gupta S, Nanduri R, Gupta P. TGF-β induced chemoresistance in liver cancer is modulated by xenobiotic nuclear receptor PXR. Cell Cycle 2019; 18:3589-3602. [PMID: 31739702 DOI: 10.1080/15384101.2019.1693120] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma appears as an extremely angiogenic solid tumor marked by apoptosis evasion, dysregulated cell cycle and low sensitivity to chemotherapy. TGF-β, a multifunctional cytokine, plays a pleiotropic role in the tumor microenvironment and has implications in cancer drug resistance. The current study provides novel evidence that TGF-β signaling contributes to drug resistance in liver cancer cells by inducing the expression of xenobiotic nuclear receptor PXR. We observed that PXR increases the expression of drug efflux transporters; therefore, accounting for exacerbated drug resistance. Additionally, anti-apoptotic nature of PXR contributes to TGF-β mediated chemoresistance as seen by procaspase-3 and Mcl-1 cellular levels. TGF-β binding to the TGF-β receptor triggers a complex downstream signaling cascade through a non-canonical SMAD-independent ERK pathway that leads to increased PXR expression. Activated ERK activates ETS1 transcription factor which is a critical regulator of endogenous PXR expression in hepatic cells. Loss of function of ETS1 abrogates the TGF-β induced PXR expression. Together these findings indicate that PXR modulates TGF-β induced resistance to chemotherapy in liver cancer cells. This underscores the need for combinatorial approaches with focus on PXR antagonism to improve drug effectiveness in hepatocellular carcinoma.Abbreviations: HCC: Hepatocellular Carcinoma; FDA: Food and Drug Administration; TGF-β: Transforming growth factor-β; PXR: Pregnane X receptor; CAR: Constitutive androstane receptor; P-gp/ABCB1: P-glycoproteins/ATP-binding cassette transporter subfamily B member 1; MRP1/ABCC1 and MRP2/ABCC2: Multidrug-resistance associated proteins; BCRP/ABCG2: Breast cancer resistant protein; DMEs: Drug-metabolizing enzymes; CFDA: 5,6-carboxyfluorescein diacetate; ETS1: Transcription factor E26 transformation specific sequence 1.
Collapse
Affiliation(s)
- Ella Bhagyaraj
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India.,Department of Infectious disease and Immunology, University of Florida, Gainesville, FL, USA
| | - Nancy Ahuja
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sumit Kumar
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Drishti Tiwari
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Shalini Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Ravikanth Nanduri
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India.,Laboratory of Metabolism, National Cancer Institute, Bethesda, MD, USA
| | - Pawan Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
8
|
Yuan P, Shentu J, Xu J, Burke W, Hsu K, Learoyd M, Zhu M, Xu B. Pharmacokinetics and safety of olaparib tablets as monotherapy and in combination with paclitaxel: results of a Phase I study in Chinese patients with advanced solid tumours. Cancer Chemother Pharmacol 2019; 83:963-974. [PMID: 30887180 DOI: 10.1007/s00280-019-03799-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/11/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE Chinese patients have been enrolled in multiple Phase III trials of the poly(ADP-ribose) polymerase (PARP) inhibitor olaparib (Lynparza); however, the pharmacokinetic (PK) profile of olaparib has not been investigated in this population. This two-part, open-label Phase I study was, therefore, carried out to determine the PK and safety profile of olaparib (tablet formulation) in Chinese patients with advanced solid tumours as monotherapy and in combination with paclitaxel (NCT02430311). METHODS The PK profile of olaparib 300 mg (twice daily [bid]; Cohort 1) as monotherapy after a single dose and at steady state, and 100 mg (bid; Cohort 2) as monotherapy (single dose and at steady state) and in combination (at steady state) with weekly paclitaxel (80 mg/m2) was assessed during Part A. Patients could continue to receive treatment (monotherapy, Cohort 1; combination therapy, Cohort 2) in Part B, which assessed safety and tolerability. RESULTS Twenty and 16 patients were enrolled into Cohorts 1 and 2, respectively. Steady-state olaparib exposure increased slightly less than proportionally with increasing monotherapy dose and inter-patient variability was high. A statistically significant decrease in olaparib exposure was seen when given in combination with paclitaxel. Discontinuation due to adverse events (AEs) was rare and haematological AEs were more common in patients receiving combination treatment. CONCLUSIONS The PK and safety profile of olaparib monotherapy in Chinese patients is consistent with that seen previously in Western and Japanese patients, and the recommended Phase III monotherapy tablet dose (300 mg bid) is suitable for use in this population.
Collapse
Affiliation(s)
- Peng Yuan
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), No. 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Jianzhong Shentu
- Research Center for Clinical Pharmacy, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Jianming Xu
- Affiliated Hospital Cancer Center, The 307th Hospital of Chinese People's Liberation Army, Academy of Military Medical Sciences, Beijing, China
| | | | | | | | - Min Zhu
- AstraZeneca, Shanghai, China
| | - Binghe Xu
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), No. 17 Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
9
|
Zhang YZ, Zuo YZ, Du ZH, Xia J, Zhang C, Wang H, Li XN, Li JL. Di (2-ethylhexyl) phthalate (DEHP)-induced hepatotoxicity in quails (Coturnix japonica) via triggering nuclear xenobiotic receptors and modulating cytochrome P450 systems. Food Chem Toxicol 2018; 120:287-293. [DOI: 10.1016/j.fct.2018.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/17/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022]
|
10
|
Chen JT, Wei L, Chen TL, Huang CJ, Chen RM. Regulation of cytochrome P450 gene expression by ketamine: a review. Expert Opin Drug Metab Toxicol 2018; 14:709-720. [PMID: 29888644 DOI: 10.1080/17425255.2018.1487397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Although used as an anesthetic drug for decades, ketamine appears to have garnered renewed interest due to its potential therapeutic uses in pain therapy, neurology, and psychiatry. Ketamine undergoes extensive oxidative metabolism by cytochrome P450 (CYP) enzymes. Considerable efforts have been expended to elucidate the ketamine-induced regulation of CYP gene expression. The safety profile of chronic ketamine administration is still unclear. Understanding how ketamine regulates CYP gene expression is clinically meaningful. Areas covered: In this article, the authors provide a brief review of clinical applications of ketamine and its metabolism by CYP enzymes. We discuss the effects of ketamine on the regulation of CYP gene expression, exploring aspects of cytoskeletal remodeling, mitochondrial functions, and calcium homeostasis. Expert opinion: Ketamine may inhibit CYP gene expression through inhibiting calcium signaling, decreasing ATP levels, producing excessive reactive oxygen species, and subsequently perturbing cytoskeletal dynamics. Further research is still needed to avoid possible ketamine-drug interactions during long-term use in the clinic.
Collapse
Affiliation(s)
- Jui-Tai Chen
- a Department of Anesthesiology, School of Medicine, College of Medicine , Taipei Medical University , Taipei City , Taiwan.,b Department of Anesthesiology, Wan-Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| | - Li Wei
- c Department of Neurosurgery, Wan-Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| | - Ta-Liang Chen
- d Anesthesiology and Health Policy Research Center , Taipei Medical University Hospital , Taipei City , Taiwan
| | - Chun-Jen Huang
- a Department of Anesthesiology, School of Medicine, College of Medicine , Taipei Medical University , Taipei City , Taiwan.,b Department of Anesthesiology, Wan-Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| | - Ruei-Ming Chen
- d Anesthesiology and Health Policy Research Center , Taipei Medical University Hospital , Taipei City , Taiwan.,e Graduate Institute of Medical Sciences, College of Medicine , Taipei Medical University , Taipei City , Taiwan.,f Cellular Physiology and Molecular Image Research Center, Wan-Fang Hospital , Taipei Medical University , Taipei City , Taiwan
| |
Collapse
|
11
|
Identification of KX2-391 as an inhibitor of HBV transcription by a recombinant HBV-based screening assay. Antiviral Res 2017. [PMID: 28624460 DOI: 10.1016/j.antiviral.2017.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Antiviral therapies for chronic hepatitis B virus (HBV) infection that are currently applicable for clinical use are limited to nucleos(t)ide analogs targeting HBV polymerase activity and pegylated interferon alpha (PEG-IFN). Towards establishing an effective therapy for HBV related diseases, it is important to develop a new anti-HBV agent that suppresses and eradicates HBV. This study used recombinant HBV encoding NanoLuc to screen anti-HBV compounds from 1827 US Food and Drug Administration approved compounds and identified several compounds that suppressed HBV infection. Among them, KX2-391, a non-ATP-competitive inhibitor of SRC kinase and tubulin polymerization, was identified as a lead candidate for an anti-HBV drug. Treatment of sodium taurocholate cotransporting polypeptide (NTCP) transduced-HepG2 (HepG2-NTCP) or primary human hepatocytes with KX2-391 suppressed HBV replication in a dose-dependent manner. The anti-HBV activity of KX2-391 appeared not to depend on SRC kinase activity because siRNA for SRC mRNA did not impair the HBV infection/replication. The anti-HBV activity of KX2-391 depended on the inhibitory effect of tubulin polymerization similar to other tubulin polymerization inhibitors, some of which were shown to inhibit HBV replication. KX2-391 inhibited HBV transcription driven by a HBV precore promoter in an HBV X protein-independent manner but did not inhibit the activity of HBV-S1, -S2, -X or cytomegalovirus promoters. Treatment with KX2-391 reduced the expression of several various factors including hepatocyte nuclear factor-4a.
Collapse
|
12
|
Mallick P, Basu S, Moorthy B, Ghose R. Role of Toll-like receptor 4 in drug-drug interaction between paclitaxel and irinotecan in vitro. Toxicol In Vitro 2017; 41:75-82. [PMID: 28242239 PMCID: PMC5479719 DOI: 10.1016/j.tiv.2017.02.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/10/2017] [Accepted: 02/23/2017] [Indexed: 12/22/2022]
Abstract
The bacterial receptor, Toll-like receptor (TLR) 4 mediates inflammatory responses and has been linked to a broad array of diseases. TLR4 agonists are being explored as potential treatments for cancer and other diseases. We have previously shown that activation of TLR4 by lipopolysaccharide (LPS) leads to down-regulation of drug metabolizing enzymes/transporters (DMETs), and altered pharmacokinetics/pharmacodynamics (PK/PD) of drugs. These changes can increase the risk of drug-drug interactions (DDIs) in patients on multiple medications. Clinically, DDI was observed for combination chemotherapy of paclitaxel (TLR4 ligand) and irinotecan. To determine the role of TLR4 in DDI between paclitaxel and irinotecan in vitro, primary hepatocytes from TLR4-wild-type (WT) and mutant mice were pre-treated with paclitaxel, followed by irinotecan. Gene expression of DMETs was determined. Paclitaxel treatment increased the levels of irinotecan metabolites, SN-38 and SN-38 glucuronide (SN-38G) in TLR4-dependent manner. Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. These novel findings demonstrate that paclitaxel can affect irinotecan metabolism by a TLR4-dependent mechanism. This provides a new perspective towards evaluation of marketed drugs according to their potential to exert DDIs in TLR4-dependent manner.
Collapse
Affiliation(s)
- Pankajini Mallick
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Sumit Basu
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, FL, USA
| | - Bhagavtula Moorthy
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA.
| |
Collapse
|
13
|
Wang X, Martínez MA, Wu Q, Ares I, Martínez-Larrañaga MR, Anadón A, Yuan Z. Fipronil insecticide toxicology: oxidative stress and metabolism. Crit Rev Toxicol 2016; 46:876-899. [DOI: 10.1080/10408444.2016.1223014] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Brewer CT, Chen T. PXR variants: the impact on drug metabolism and therapeutic responses. Acta Pharm Sin B 2016; 6:441-449. [PMID: 27709012 PMCID: PMC5045535 DOI: 10.1016/j.apsb.2016.07.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/21/2016] [Accepted: 05/04/2016] [Indexed: 01/30/2023] Open
Abstract
The pregnane X receptor (PXR) plays an important and diverse role in mediating xenobiotic induction of drug-metabolizing enzymes and transporters. Several protein isoforms of PXR exist, and they have differential transcriptional activity upon target genes; transcript variants 3 (PXR3) and 4 (PXR4) do not induce target gene expression, whereas transcript variants 1 (PXR1) and 2 (PXR2) respond to agonist by activating target gene expression. PXR protein variants also display differences in protein-protein interactions; PXR1 interacts with p53, whereas PXR3 does not. Furthermore, the transcript variants of PXR that encode these protein isoforms are differentially regulated by methylation and deletions in the respective promoters of the variants, and their expression differs in various human cancers and also in cancerous tissue compared to adjacent normal tissues. PXR1 and PXR4 mRNA are downregulated by methylation in cancerous tissue and have divergent effects on cellular proliferation when ectopically overexpressed. Additional detailed and comparative mechanistic studies are required to predict the effect of PXR transcript variant expression on carcinogenesis, therapeutic response, and the development of toxicity.
Collapse
Key Words
- AF, activating function
- BAMCA, bacterial artificial chromosome array–based methylated CpG island amplification
- CYP, cytochrome P450
- Drug metabolism
- GST, glutathione S-transferase
- MDR, multidrug resistance protein
- NHR, nuclear hormone receptor
- P-gp, P-glycoprotein
- PXR1, PXR transcript variant 1 (434 residues)
- PXR2, transcript variant 2 (473 residues)
- PXR3, transcript variant 3 (397 residues)
- PXR4, transcript variant 4 (322 residues;AK122990)
- Pregnane X receptor
- RACE, 5′ rapid amplification of cDNA ends
- Therapeutic responses
- Toxicity
- Transcript variants
- UGT, UDP-glucuronosyltransferase
- UTR, untranslated region
- shRNA, short hairpin RNA
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- C. Trent Brewer
- Department of Chemical Biology and Therapeutics, St. Jude Children′s Research Hospital, Memphis, TN 38105, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children′s Research Hospital, Memphis, TN 38105, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Corresponding author at: Department of Chemical Biology and Therapeutics, St. Jude Children′s Research Hospital, Mail Stop #1000, 262 Danny Thomas Place, Memphis, TN 38105, USA. Tel.: +1 901 595 5937; fax: +1 901 595 5715.Department of Chemical Biology and Therapeutics, St. Jude Children′s Research Hospital, Mail Stop #1000, 262 Danny Thomas PlaceMemphisTN38105USA
| |
Collapse
|
15
|
Zhao S, Zhang Y, Ju P, Gu L, Zhuang R, Zhao L, Tang X, Bi K, Chen X. Determination of 6258-70, a new semi-synthetic taxane, in rat plasma and tissues: Application to the pharmacokinetics and tissue distribution study. J Pharm Anal 2016; 6:219-225. [PMID: 29403986 PMCID: PMC5762609 DOI: 10.1016/j.jpha.2016.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/27/2015] [Accepted: 02/29/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer is the leading cause of death all over the world. Among the chemotherapy drugs, taxanes play an important role in cancer treatment. 6258-70 is a new semi-synthetic taxane which has a broad spectrum of antitumor activity. A fast and reliable high performance liquid chromatography-tandem mass spectrometry (HPLC–MS/MS) method was developed for quantification of 6258-70 in rat plasma and tissues in this paper. After extraction by liquid-liquid extraction method with methyl tert-butyl ether, the samples were separated on a Kinetex C18 column (50 mm×2.1 mm, 2.6 µm, Phenomenex, USA) within 3 min. The method was fully validated with the matrix effect between 87.7% and 99.5% and the recovery ranging from 80.3% to 90.1%. The intra- and inter-day precisions were less than 9.5% and the accuracy ranged from −3.8% to 6.5%. The reliable method was successfully applied to the pharmacokinetics and tissue distribution studies of 6258-70 after intravenous administration in rats. The pharmacokinetic results indicated that the pharmacokinetic behavior of 6258-70 in rats was in accordance with linear features within tested dosage of 1 to 4 mg/kg, and there was no significant difference between the two genders. The tissue distribution study showed that 6258-70 had an effective penetration, spread widely and rapidly and could cross blood-brain barrier. The results of pharmacokinetics and tissue distribution may provide a guide for future study.
Collapse
Affiliation(s)
- Simin Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yuanyuan Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Ping Ju
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Liqiang Gu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Rui Zhuang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Longshan Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xiaohui Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
16
|
Ghose R, Mallick P, Taneja G, Chu C, Moorthy B. In Vitro Approaches to Study Regulation of Hepatic Cytochrome P450 (CYP) 3A Expression by Paclitaxel and Rifampicin. Methods Mol Biol 2016; 1395:55-68. [PMID: 26910068 DOI: 10.1007/978-1-4939-3347-1_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer is the second leading cause of mortality worldwide; however the response rate to chemotherapy treatment remains slow, mainly due to narrow therapeutic index and multidrug resistance. Paclitaxel (taxol) has a superior outcome in terms of response rates and progression-free survival. However, numerous cancer patients are resistant to this drug. In this investigation, we tested the hypothesis that induction of cytochrome P450 (Cyp)3a11 gene by paclitaxel is downregulated by the inflammatory mediator, lipopolysaccharide (LPS), and that the pro-inflammatory cytokine, tumor necrosis factor (TNF)-α, attenuates human CYP3A4 gene induction by rifampicin. Primary mouse hepatocytes were pretreated with LPS (1 μg/ml) for 10 min, followed by paclitaxel (20 μM) or vehicle for 24 h. RNA was extracted from the cells by trizol method followed by cDNA synthesis and analysis by real-time PCR. Paclitaxel significantly induced gene expression of Cyp3a11 (~30-fold) and this induction was attenuated in LPS-treated samples. Induction and subsequent downregulation of CYP3A enzyme can impact paclitaxel treatment in cancer patients where inflammatory mediators are activated. It has been shown that the nuclear receptor, pregnane X receptor (PXR), plays a role in the induction of CYP enzymes. In order to understand the mechanisms of regulation of human CYP3A4 gene, we co-transfected HepG2 cells (human liver cell line) with CYP3A4-luciferase construct and a PXR expression plasmid. The cells were then treated with the pro-inflammatory cytokine, TNFα, followed by the prototype CYP3A inducer rifampicin. It is well established that rifampicin activates PXR, leading to CYP3A4 induction. We found that induction of CYP3A4-luciferase activity by rifampicin was significantly attenuated by TNFα. In conclusion, we describe herein several in vitro approaches entailing primary and cultured hepatocytes, real-time PCR, and transcriptional activation (transfection) assays to investigate the molecular regulation of CYP3A, which plays a pivotal role in the metabolism of numerous chemotherapeutic drugs. Genetic or drug-induced variation in CYP3A and/or PXR expression could contribute to drug resistance to chemotherapeutic agents in cancer patients.
Collapse
Affiliation(s)
- Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77030, USA
| | - Pankajini Mallick
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77030, USA
| | - Guncha Taneja
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77030, USA
| | - Chun Chu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Mbatchi LC, Schmitt A, Thomas F, Cazaubon Y, Robert J, Lumbroso S, Brouillet JP, Pourquier P, Chatelut E, Boyer JC, Evrard A. Polymorphisms in SLCO1B3 and NR1I2 as genetic determinants of hematotoxicity of carboplatin and paclitaxel combination. Pharmacogenomics 2015; 16:1439-50. [DOI: 10.2217/pgs.15.84] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The goal of our study was to assess the impact of patients’ genetic background on their sensitivity to carboplatin/paclitaxel hematotoxicity. Patients & methods: Parameters describing sensitivity to neutropenia and to thrombocytopenia of 201 patients were extracted from a previous pharmacokinetic/pharmacodynamics analysis, in order to assess their association with 52 candidates SNPs in 18 genes. Results: Carriers of a T allele of SLCO1B3-rs4149117 were 19% less sensitive to thrombocytopenia than the homozygotes for the G allele (p = 0.00279). Carriers of two copies of the ATG haplotypes of NR1I2-rs1523130, rs3814055 and rs1523127 were 19% less sensitive to thrombocytopenia than those harboring other haplotypes (p = 0.025). Conclusion: Our results revealed the importance of SLCO1B3 and NR1I2 in the sensitivity to carboplatin/paclitaxel thrombocytopenia.
Collapse
Affiliation(s)
- Litaty Céphanoée Mbatchi
- Laboratoire de biochimie, Centre Hospitalier Universitaire (CHU) of Nîmes, Hôpital Carémeau, Nîmes, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, INSERM, U1194 France
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, Montpellier, France
| | - Antonin Schmitt
- EA4553 Institut Claudius-Regaud, and Université Paul-Sabatier, Toulouse, France
| | - Fabienne Thomas
- EA4553 Institut Claudius-Regaud, and Université Paul-Sabatier, Toulouse, France
| | - Yoann Cazaubon
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, Montpellier, France
| | - Jacques Robert
- INSERM U916, Institut Bergonié, Université de Bordeaux, France
| | - Serge Lumbroso
- Laboratoire de biochimie, Centre Hospitalier Universitaire (CHU) of Nîmes, Hôpital Carémeau, Nîmes, France
| | - Jean-Paul Brouillet
- Laboratoire de biochimie, Centre Hospitalier Universitaire (CHU) of Nîmes, Hôpital Carémeau, Nîmes, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, INSERM, U1194 France
| | - Philippe Pourquier
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, INSERM, U1194 France
| | - Etienne Chatelut
- EA4553 Institut Claudius-Regaud, and Université Paul-Sabatier, Toulouse, France
| | - Jean-Christophe Boyer
- Laboratoire de biochimie, Centre Hospitalier Universitaire (CHU) of Nîmes, Hôpital Carémeau, Nîmes, France
| | - Alexandre Evrard
- Laboratoire de biochimie, Centre Hospitalier Universitaire (CHU) of Nîmes, Hôpital Carémeau, Nîmes, France
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, INSERM, U1194 France
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, Montpellier, France
| |
Collapse
|
18
|
Cherian MT, Chai SC, Chen T. Small-molecule modulators of the constitutive androstane receptor. Expert Opin Drug Metab Toxicol 2015; 11:1099-114. [PMID: 25979168 DOI: 10.1517/17425255.2015.1043887] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION The constitutive androstane receptor (CAR) induces drug-metabolizing enzymes for xenobiotic metabolism. AREAS COVERED This review covers recent advances in elucidating the biological functions of CAR and its modulation by a growing number of agonists and inhibitors. EXPERT OPINION Extrapolation of animal CAR function to that of humans should be carefully scrutinized, particularly when rodents are used in evaluating the metabolic profile and carcinogenic properties of clinical drugs and environmental chemicals. Continuous efforts are needed to discover novel CAR inhibitors, with extensive understanding of their inhibitory mechanism, species selectivity, and discriminating power against other xenobiotic sensors.
Collapse
Affiliation(s)
- Milu T Cherian
- Postdoctoral fellow, St. Jude Children's Research Hospital, Department of Chemical Biology and Therapeutics , 262 Danny Thomas Place, Memphis, TN 38105 , USA
| | | | | |
Collapse
|
19
|
Zhuo W, Hu L, Lv J, Wang H, Zhou H, Fan L. Role of pregnane X receptor in chemotherapeutic treatment. Cancer Chemother Pharmacol 2014; 74:217-27. [PMID: 24889719 DOI: 10.1007/s00280-014-2494-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/06/2014] [Indexed: 10/25/2022]
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.
Collapse
Affiliation(s)
- Wei Zhuo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | | | | | | | | | | |
Collapse
|
20
|
Rathod V, Jain S, Nandekar P, Sangamwar AT. Human pregnane X receptor: a novel target for anticancer drug development. Drug Discov Today 2014; 19:63-70. [DOI: 10.1016/j.drudis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/02/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023]
|
21
|
Pregnane X receptor dependent up-regulation of CYP2C9 and CYP3A4 in tumor cells by antitumor acridine agents, C-1748 and C-1305, selectively diminished under hypoxia. Biochem Pharmacol 2013; 86:231-41. [PMID: 23688499 DOI: 10.1016/j.bcp.2013.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 05/04/2013] [Accepted: 05/08/2013] [Indexed: 01/12/2023]
Abstract
Induction of proteins involved in drug metabolism and in drug delivery has a significant impact on drug-drug interactions and on the final therapeutic effects. Two antitumor acridine derivatives selected for present studies, C-1748 (9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine) and C-1305 (5-dimethylaminopropylamino-8-hydroxy-triazoloacridinone), expressed high and low susceptibility to metabolic transformations with liver microsomes, respectively. In the current study, we examined the influence of these compounds on cytochrome P450 3A4 (CYP3A4) and 2C9 (CYP2C9) enzymatic activity and gene expression in HepG2 tumor cells. Luminescence and HPLC examination, real-time RT-PCR and western blot analyses along with transfection of pregnane X receptor (PXR) siRNA and CYP3A4 reporter gene assays were applied. We found that both compounds strongly induced CYP3A4 and CYP2C9 activity and expression as well as expression of UGT1A1 and MDR1 in a concentration- and time-dependent manner. C-1748-mediated CYP3A4 and CYP2C9 mRNA induction equal to rifampicin occurred at extremely low concentrations (0.001 and 0.01μM), whereas 10μM C-1305 induced three-times higher CYP3A4 and CYP2C9 mRNA levels than rifampicin did. CYP3A4 and CYP2C9 expressions were shown to be PXR-dependent; however, neither compound influenced PXR expression. Thus, the observed drug-mediated induction of isoenzymes occurs on a PXR-mediated regulatory level. Furthermore, C-1748 and C-1305 were demonstrated to be selective PXR agonists. These effects are hypoxia-inhibited only in the case of C-1748, which is sensitive to P450 metabolism. In summary, PXR was found to be a new target of the studied compounds. Thus, possible combinations of these compounds with other therapeutics might lead to the PXR-dependent enzyme-mediated drug-drug interactions.
Collapse
|
22
|
Hester S, Moore T, Padgett WT, Murphy L, Wood CE, Nesnow S. The Hepatocarcinogenic Conazoles: Cyproconazole, Epoxiconazole, and Propiconazole Induce a Common Set of Toxicological and Transcriptional Responses. Toxicol Sci 2012; 127:54-65. [DOI: 10.1093/toxsci/kfs086] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
23
|
Nesnow S, Grindstaff RD, Lambert G, Padgett WT, Bruno M, Ge Y, Chen PJ, Wood CE, Murphy L. Propiconazole increases reactive oxygen species levels in mouse hepatic cells in culture and in mouse liver by a cytochrome P450 enzyme mediated process. Chem Biol Interact 2011; 194:79-89. [DOI: 10.1016/j.cbi.2011.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 01/14/2023]
|
24
|
Overcoming drug resistance by regulating nuclear receptors. Adv Drug Deliv Rev 2010; 62:1257-64. [PMID: 20691230 DOI: 10.1016/j.addr.2010.07.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 07/21/2010] [Accepted: 07/23/2010] [Indexed: 12/12/2022]
Abstract
Drug resistance involves multiple mechanisms. Multidrug resistance (MDR) is the leading cause of treatment failure in cancer therapy. Elevated levels of MDR proteins [members of the ATP-binding cassette (ABC) transporter family] increase cellular efflux and decrease the effectiveness of chemotherapeutic agents. As a salvage approach to overcome drug resistance, inhibitors of MDR proteins have been developed, but have had limited success mainly due to undesired toxicities. Nuclear receptors (NRs), including pregnane X receptor (PXR), regulate the expression of proteins (including MDR proteins) involved in drug metabolism and drug clearance, suggesting that it is possible to overcome drug resistance by regulating NR. This review discusses the progress in the development of MDR inhibitors, with a focus on MDR1 inhibitors. Recent development of PXR antagonists to pharmacologically modulate PXR is also reviewed. The review proposes that selectively preventing the elevation of MDR levels by regulating NRs rather than non-selectively inhibiting the MDR activity by using MDR inhibitors can be a less toxic approach to overcome drug resistance during cancer therapy.
Collapse
|
25
|
Sugawara M, Okamoto K, Kadowaki T, Kusano K, Fukamizu A, Yoshimura T. Inoculation of human tumor cells alters the basal expression but not the inducibility of cytochrome P450 enzymes in tumor-bearing mouse liver. Drug Metab Dispos 2009; 37:2244-54. [PMID: 19666990 DOI: 10.1124/dmd.109.028571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The athymic nude mouse is often used to grow tumors for in vivo oncology research, including the identification of anticancer drugs, whereas wild-type mice are usually used to assess the pharmacokinetics (PK) of new chemical entities. The relationship between PK and pharmacodynamics (PD) provides useful mechanistic information and helps guide of the clinical regimen. The aim of this study was to assess whether the inoculation of human hepatocellular carcinoma cells (PLC/PRF/5) into athymic nude mice alters the expression of genes encoding the drug-metabolizing enzymes and transporters in host liver. The livers from nontumor- and tumor-bearing mice were initially subjected to drug metabolism gene microarray analysis. Microarray analysis indicated that tumor inoculation had little effect on drug metabolism-related genes, including several cytochrome P450s: Cyp1a, Cyp2b, and Cyp3a. This result was further confirmed by reverse transcription-polymerase chain reaction (RT-PCR). However, immunoreactive proteins of Cyp1a, Cyp2b, and Cyp3a were suppressed by tumor inoculation. RT-PCR and Western immunoblotting analysis showed that the inducibility of Cyp1a, Cyp2b, and Cyp3a by 3-methylcholanthrene, phenobarbital, and dexamethasone, respectively, was similar between nontumor- and tumor-bearing mice. These results suggest that inoculation of human tumor cells into athymic nude mice suppresses the expression of certain drug-metabolizing enzymes, which may alter the PK and PD of antitumor drugs.
Collapse
Affiliation(s)
- Michiko Sugawara
- Drug Metabolism and Pharmacokinetics Research, Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
Nesnow S, Ward W, Moore T, Ren H, Hester SD. Discrimination of Tumorigenic Triazole Conazoles from Phenobarbital by Transcriptional Analyses of Mouse Liver Gene Expression. Toxicol Sci 2009; 110:68-83. [DOI: 10.1093/toxsci/kfp076] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
27
|
Wang H, Huang H, Li H, Teotico DG, Sinz M, Baker SD, Staudinger J, Kalpana G, Redinbo MR, Mani S. Activated pregnenolone X-receptor is a target for ketoconazole and its analogs. Clin Cancer Res 2007; 13:2488-95. [PMID: 17438109 DOI: 10.1158/1078-0432.ccr-06-1592] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Variations in biotransformation and elimination of microtubule-binding drugs are a major cause of unpredictable side effects during cancer therapy. Because the orphan receptor, pregnenolone X-receptor (PXR), coordinately regulates the expression of paclitaxel metabolizing and transport enzymes, controlling this process could improve therapeutic outcome. EXPERIMENTAL DESIGN In vitro RNA-, protein-, and transcription-based assays in multiple cell lines derived from hepatocytes and PXR wild-type and null mouse studies were employed to show the effects of ketoconazole and its analogues on ligand-activated PXR-mediated gene transcription and translation. RESULTS The transcriptional activation of genes regulating biotransformation and transport by the liganded human nuclear xenobiotic receptor, PXR, was inhibited by the commonly used antifungal ketoconazole and related azole analogs. Mutations at the AF-2 surface of the human PXR ligand-binding domain indicate that ketoconazole may interact with specific residues outside the ligand-binding pocket. Furthermore, in contrast to that observed in PXR (+/+) mice, genetic loss of PXR results in increased (preserved) blood levels of paclitaxel. CONCLUSIONS These studies show that some azole compounds repress the coordinated activation of genes involved in drug metabolism by blocking PXR activation. Because loss of PXR maintains blood levels of paclitaxel upon chronic dosing, ketoconazole analogues may also serve to preserve paclitaxel blood levels on chronic dosing of drugs. Our observations may facilitate new strategies to improve the clinical efficacy of drugs and to reduce therapeutic side effects.
Collapse
Affiliation(s)
- Hongwei Wang
- Albert Einstein Cancer Center, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The pregnane X receptor (PXR; NR1I2) is a nuclear hormone receptor (NR) that transcriptionally regulates genes encoding transporters and drug-metabolising enzymes in the liver and intestine. PXR activation leads to enhanced metabolism and elimination of xenobiotics and endogenous compounds such as hormones and bile salts. Relative to other vertebrate NRs, PXR has the broadest specificity for ligand activators by virtue of a large, flexible ligand-binding cavity. In addition, PXR has the most extensive sequence diversity across vertebrate species in the ligand-binding domain of any NR, with significant pharmacological differences between human and rodent PXRs, and especially marked divergence between mammalian and nonmammalian PXRs. The unusual properties of PXR complicate the use of in silico and animal models to predict in vivo human PXR pharmacology. Research into the evolutionary history of the PXR gene has also provided insight into the function of PXR in humans and other animals.
Collapse
Affiliation(s)
- Manisha Iyer
- University of Pittsburgh, Department of Pathology, Scaife Hall S-730, 3550 Terrace Street, Pittsburgh, PA 15261 USA
| | - Erica J. Reschly
- University of Pittsburgh, Department of Pathology, Scaife Hall S-730, 3550 Terrace Street, Pittsburgh, PA 15261 USA
| | - Matthew D. Krasowski
- University of Pittsburgh, Department of Pathology, Scaife Hall S-730, 3550 Terrace Street, Pittsburgh, PA 15261 USA
- Author for correspondence, Tel: 412-647-6517; Fax: 412-647-5934; E-mail:
| |
Collapse
|
29
|
Harmsen S, Meijerman I, Beijnen JH, Schellens JHM. The role of nuclear receptors in pharmacokinetic drug–drug interactions in oncology. Cancer Treat Rev 2007; 33:369-80. [PMID: 17451886 DOI: 10.1016/j.ctrv.2007.02.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 02/06/2007] [Accepted: 02/11/2007] [Indexed: 01/17/2023]
Abstract
Drug-drug interactions can have a major impact on treatment outcome in cancer patients. These patients are at high risk of such interactions, because they are treated with combinations of multiple cytotoxic anticancer drugs or hormonal agents often co-administered with prophylactic antiemetics and analgesics to provide palliation. Interactions between drugs can affect the pharmacokinetics of concomitantly administered chemotherapeutic agents. Especially, due to the specific properties of anticancer drugs, such as a narrow therapeutic index and steep dose-toxicity curve, small pharmacokinetic changes can have significant clinical consequences like decreased therapeutic efficacy or increased toxicity. An important mechanism that underlies these interactions is the induction of enzymes or efflux transporters involved in the biotransformation and clearance of anticancer drugs. Several nuclear receptors, like the pregnane X receptor (PXR), constitutively androstane receptor (CAR), have been shown to regulate induction. Activation of these receptors will lead to induction of important enzymes like cytochrome P450 3A4 (CYP3A4), which is involved in the biotransformation of more than 50% of all clinically used drugs. Therefore, concomitant administration of agents that activate PXR will affect the pharmacokinetics of drugs that are substrate for PXRs target genes, which include CYP3A4 and MDR-1. Understanding of the molecular mechanisms that underlie enzyme induction and the identification of (new) drugs involved in pharmacokinetic drug-drug interactions may contribute to the predictability of drug-drug interactions and eventually help to develop safer anticancer regimens.
Collapse
Affiliation(s)
- S Harmsen
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Biomedical Analysis, Sorbonnelaan 16, 3584 CA Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
30
|
Stanley LA, Horsburgh BC, Ross J, Scheer N, Wolf CR. PXR and CAR: nuclear receptors which play a pivotal role in drug disposition and chemical toxicity. Drug Metab Rev 2006; 38:515-97. [PMID: 16877263 DOI: 10.1080/03602530600786232] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Xenobiotic metabolism and detoxification is regulated by receptors (e.g., PXR, CAR) whose characterization has contributed significantly to our understanding of drug responses in humans. Technologies facilitating the screening of compounds for receptor interactions provide valuable tools applicable in drug development. Most use in vitro systems or mice humanized for receptors in vivo. In vitro assays are limited by the reporter systems and cell lines chosen and are uninformative about effects in vivo. Humanized mouse models provide novel, exciting ways of understanding the functions of these genes. This article evaluates these technologies and current knowledge on PXR/CAR-mediated regulation of gene expression.
Collapse
Affiliation(s)
- Lesley A Stanley
- Consultant in Investigative Toxicology, St. Andrews, Fife, United Kingdom
| | | | | | | | | |
Collapse
|
31
|
Chang TKH, Waxman DJ. Synthetic drugs and natural products as modulators of constitutive androstane receptor (CAR) and pregnane X receptor (PXR). Drug Metab Rev 2006; 38:51-73. [PMID: 16684648 DOI: 10.1080/03602530600569828] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are members of the nuclear receptor superfamily. These transcription factors are predominantly expressed in the liver, where they are activated by structurally diverse compounds, including many drugs and endogenous substances. CAR and PXR regulate the expression of a broad range of genes, which contribute to transcellular transport, bioactivation, and detoxification of numerous xenochemicals and endogenous substances. This article discusses the importance of these receptors for pharmacology and toxicology, emphasizing the role of individual drugs and natural products as agonists, indirect activators, inverse agonists, and antagonists of CAR and PXR.
Collapse
Affiliation(s)
- Thomas K H Chang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada.
| | | |
Collapse
|
32
|
Hariparsad N, Sane RS, Strom SC, Desai PB. In vitro methods in human drug biotransformation research: implications for cancer chemotherapy. Toxicol In Vitro 2006; 20:135-53. [PMID: 16359840 DOI: 10.1016/j.tiv.2005.06.049] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2005] [Revised: 05/01/2005] [Accepted: 06/01/2005] [Indexed: 11/28/2022]
Abstract
Anticancer drugs have a complex pharmacological and toxicological profile with a narrow therapeutic index. It is therefore critical to understand the factors that contribute to the marked intersubject variability in the pharmacokinetics and pharmacodynamics often observed with anticancer compounds. Since hepatic and extra-hepatic drug metabolism represents a major drug disposition pathway, extensive efforts are made to thoroughly investigate metabolism of anticancer compounds during the pre-clinical and clinical development phases as well as to address issues encountered during the clinical use of an approved drug. In recent years there has been a significant paradigm shift in pre-clinical/non-clinical drug metabolism studies. Most importantly, this has included a reduced reliance on animal models and increased use of human tissues (i.e. human liver microsomes and other cellular fractions, primary culture of human hepatocytes, cDNA expressed human-specific enzymes and cell-based reporter assays). Typically, experiments are performed using these tools to identify the phase I and/or phase II enzymes involved in metabolism of the drug/investigational agent and for metabolic fingerprinting. Additionally, issues pertaining to the rate, extent and mechanism(s) of the inhibition or induction of the metabolic pathways are also investigated. These studies provide important clues about various aspects of the disposition of a therapeutic agent including first-pass metabolism, elimination half-life, overall bioavailability and the potential for drug-drug interactions. The methodologies used for in vitro assessment of drug metabolism and their applications to drug development and clinical therapeutics with special emphasis on anticancer drugs are reviewed in this manuscript.
Collapse
Affiliation(s)
- N Hariparsad
- College of Pharmacy, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | | | | | | |
Collapse
|
33
|
Mathias AA, Maggio-Price L, Lai Y, Gupta A, Unadkat JD. Changes in pharmacokinetics of anti-HIV protease inhibitors during pregnancy: the role of CYP3A and P-glycoprotein. J Pharmacol Exp Ther 2006; 316:1202-9. [PMID: 16293714 DOI: 10.1124/jpet.105.095406] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human immunodeficiency virus (HIV)-infected women have reduced exposure [area under the curve (AUC)] to anti-HIV protease inhibitors [e.g., nelfinavir (NFV)] during pregnancy. To determine the mechanistic basis of this phenomenon, we administered NFV mesylate orally (2.5 mg) or intravenously (0.625 mg) to timed pregnant (gestational age: 18-19 days) and non-pregnant FVB mice. After oral but not after i.v. administration, the plasma clearance of NFV was higher (by 134%, p < 0.05) and bioavailability was lower (by 32%, p < 0.05) in pregnant (n = 3) versus nonpregnant mice (n = 3). These effects of pregnancy were not due to changes in plasma protein binding of NFV. The half-life of NFV depletion in hepatic S-9 fractions of pregnant mice (n = 8) was 2.2-fold faster (p < 0.05) than that in nonpregnant mice (n = 7). Hepatic CYP3A activity (testosterone 6beta-hydroxylation, n = 4) and expression (n = 8) were significantly higher (by 138 and 49%, p < 0.05) in pregnant mice than that in nonpregnant mice. In the intestine, no CYP3A activity was detected and CYP3A protein expression (n = 6, p > 0.05) was not significantly different between the two groups. P-glycoprotein expression (n = 6) in hepatic and intestinal tissue of pregnant mice was not significantly different from that in nonpregnant mice. These changes in disposition of NFV during pregnancy are predominately due to a change in its bioavailability. An increase in hepatic CYP3A can explain the reduced bioavailability of NFV during pregnancy. If such upregulation of hepatic CYP3A activity occurs in pregnant women, it has important implications for dose adjustment of a variety of drugs ingested by pregnant women and cleared predominately via CYP3A metabolism.
Collapse
Affiliation(s)
- Anita A Mathias
- Department of Pharmaceutics, University of Washington, Box 357610, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
34
|
Modrianský M, Dvořák Z. MICROTUBULE DISRUPTORS AND THEIR INTERACTION WITH BIOTRANSFORMATION ENZYMES. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2005. [DOI: 10.5507/bp.2005.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
35
|
Gustafson DL, Long ME, Bradshaw EL, Merz AL, Kerzic PJ. P450 induction alters paclitaxel pharmacokinetics and tissue distribution with multiple dosing. Cancer Chemother Pharmacol 2005; 56:248-54. [PMID: 15856231 DOI: 10.1007/s00280-004-0988-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Accepted: 11/16/2004] [Indexed: 11/26/2022]
Abstract
PURPOSE Paclitaxel (Taxol) is an effective agent against a broad range of human cancers. Studies on the metabolism and disposition of paclitaxel have shown that it is primarily eliminated via hepatic metabolism by P450 enzymes (2C8 and 3A4) to essentially inactive metabolites, and that biliary and gut transport by P-glycoprotein (PGP) as well as urinary elimination of the parent compound play relatively minor roles. Recent studies in vitro have shown that paclitaxel treatment increases the level of CYP2C8 and CYP3A4 in human hepatocytes as well as PGP in colon tumor cells. The data suggest that previous paclitaxel exposure may influence metabolism and elimination of subsequent doses. Further, since weekly paclitaxel dose schedules are becoming more common as opposed to the original every 21-day dosing, the likelihood of enzyme induction from previous doses impacting that from subsequent doses is increased. METHODS To study the potential for such sequence-dependent alterations in paclitaxel pharmacokinetics, we carried out pharmacokinetic studies in mouse plasma and tissues following day 1 and days 1 and 5 dosing at 20 mg/kg. Paclitaxel concentrations were determined by a sensitive LC/MS/MS assay out to 16 h post-dosing in plasma, liver, kidney, gut and heart. The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. RESULTS Pharmacokinetic analysis of the data showed that plasma and tissue AUC values after treatment on day 5 following a dose on day 1 were between 50% and 74% of those determined following a single dose on day 1. The terminal elimination half-life was not different. Activity and protein levels for CYP2C in liver were elevated at 24 and 96 h after paclitaxel dosing. Cremophor EL, a carrier solvent for paclitaxel, also caused elevated CYP2C activity. Neither CYP3A nor PGP levels in liver were altered by paclitaxel or Cremophor EL treatment at the 24-h and 96-h time points. The levels of 6alpha-OH-paclitaxel in feces were increased on day 5 as opposed to day 1 while paclitaxel levels in feces were unchanged. CONCLUSIONS The results of our studies showed that paclitaxel pharmacokinetics are altered by previous paclitaxel exposure up to 96 h earlier.
Collapse
Affiliation(s)
- Daniel L Gustafson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Health Sciences Center, C-238, Denver, CO 80262, USA.
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Vignati LA, Bogni A, Grossi P, Monshouwer M. A human and mouse pregnane X receptor reporter gene assay in combination with cytotoxicity measurements as a tool to evaluate species-specific CYP3A induction. Toxicology 2004; 199:23-33. [PMID: 15125996 DOI: 10.1016/j.tox.2003.12.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Revised: 11/14/2003] [Accepted: 12/11/2003] [Indexed: 01/03/2023]
Abstract
Recent studies have demonstrated that a member of the nuclear receptor family, pregnane X receptor (PXR) is a key regulator of the expression of cytochrome P450 3A (CYP3A) in humans and rodents. It is also known that species specificity in the induction of CYP3A by xenobiotics is likely a consequence of differences at the level of PXR activation. Because of the importance of CYP3A4 in drug metabolism, the development of rapid and accurate in vitro assays for predicting the effects of compounds on CYP3A4 expression or activity in humans has been a long-standing goal within pharmaceutical industries. PXR activation measurements using an in vitro reporter gene approach appears to provide a rapid and relatively inexpensive means for predicting whether compounds will induce CYP3A levels in vivo. In this study, using an HepG2 cell based human and mouse PXR reporter gene assay, 23 compounds were tested for their potential to activate hPXR or mPXR. Data demonstrated that potent activators of hPXR had virtually no activity on mPXR and efficient activators of mPXR had weak activity on hPXR. In addition, a third category of moderate/weak activators of both hPXR and mPXR was identified. Exemestane was a strong activator of mPXR ( approximately 22-fold activation) with only minor effect on hPXR ( approximately 5-fold activation). The importance of cell viability measurements as part of the PXR reporter gene assay was demonstrated as significant cytotoxicity or inhibition of cell proliferation might underestimate the potential for PXR activation.
Collapse
Affiliation(s)
- Luisella A Vignati
- Department of Pharmacokinetics, Dynamics and Metabolism, Pharmacia, Gruppo Pfizer Inc., Viale Pasteur 10, 20014 Nerviano, MI, Italy
| | | | | | | |
Collapse
|
38
|
McFadyen MCE, Melvin WT, Murray GI. Cytochrome P450 enzymes: Novel options for cancer therapeutics. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.363.3.3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abstract
The concept of overexpression of individual forms of cytochrome P450 enzymes in tumor cells is now becoming well recognized. Indeed, a growing body of research highlights the overexpression of P450s, particularly CYP1B1, in tumor cells as representing novel targets for anticancer therapy. The purpose of this review is to outline the novel therapeutic options and opportunities arising from both enhanced endogenous expression of cytochrome P450 in tumors and cytochrome P450-mediated gene therapy.
Collapse
Affiliation(s)
| | - William T. Melvin
- 2Molecular and Cell Biology, University of Aberdeen, Aberdeen, United Kingdom
| | | |
Collapse
|