1
|
Bachorz RA, Nowak D, Ratajewski M. QSPRmodeler - An open source application for molecular predictive analytics. FRONTIERS IN BIOINFORMATICS 2024; 4:1441024. [PMID: 39391332 PMCID: PMC11464749 DOI: 10.3389/fbinf.2024.1441024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
The drug design process can be successfully supported using a variety of in silico methods. Some of these are oriented toward molecular property prediction, which is a key step in the early drug discovery stage. Before experimental validation, drug candidates are usually compared with known experimental data. Technically, this can be achieved using machine learning approaches, in which selected experimental data are used to train the predictive models. The proposed Python software is designed for this purpose. It supports the entire workflow of molecular data processing, starting from raw data preparation followed by molecular descriptor creation and machine learning model training. The predictive capabilities of the resulting models were carefully validated internally and externally. These models can be easily applied to new compounds, including within more complex workflows involving generative approaches.
Collapse
Affiliation(s)
- Rafał A. Bachorz
- Institute of Medical Biology, Polish Academy of Sciences, Łódź, Poland
| | - Damian Nowak
- Institute of Medical Biology, Polish Academy of Sciences, Łódź, Poland
- Department of Quantum Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| | - Marcin Ratajewski
- Institute of Medical Biology, Polish Academy of Sciences, Łódź, Poland
| |
Collapse
|
2
|
Bulitta JB, Fang E, Stryjewski ME, Wang W, Atiee GJ, Stark JG, Hafkin B. Population pharmacokinetic rationale for intravenous contezolid acefosamil followed by oral contezolid dosage regimens. Antimicrob Agents Chemother 2024; 68:e0140023. [PMID: 38415667 PMCID: PMC10989001 DOI: 10.1128/aac.01400-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/09/2024] [Indexed: 02/29/2024] Open
Abstract
Contezolid is a novel oxazolidinone antibiotic with a promising safety profile. Oral contezolid and its intravenous (IV) prodrug contezolid acefosamil (CZA) are in development for treatment of diabetic foot and acute bacterial skin and skin structure infections (ABSSSI). The prodrug CZA is converted to active contezolid via intermediate MRX-1352. This study aimed to provide the pharmacokinetic rationale for safe, effective, and flexible dosage regimens with initial IV CZA followed by oral contezolid. We simultaneously modeled plasma concentrations from 110 healthy volunteers and 74 phase 2 patients with ABSSSI via population pharmacokinetics (using the importance sampling estimation algorithm), and optimized dosage regimens by Monte Carlo simulations. This included data on MRX-1352, contezolid, and its metabolite MRX-1320 from 66 healthy volunteers receiving intravenous CZA (150-2400 mg) for up to 28 days, and 74 patients receiving oral contezolid [800 mg every 12 h (q12h)] for 10 days. The apparent total clearance for 800 mg oral contezolid with food was 16.0 L/h (23.4% coefficient of variation) in healthy volunteers and 17.7 L/h (53.8%) in patients. CZA was rapidly converted to MRX-1352, which subsequently transformed to contezolid. The proposed dosage regimen used an IV CZA 2000 mg loading dose with 1000 mg IV CZA q12h as maintenance dose(s), followed by 800 mg oral contezolid q12h (with food). During each 24-h period, Monte Carlo simulations predicted this regimen to achieve consistent areas under the curve of 91.9 mg·h/L (range: 76.3-106 mg·h/L) under all scenarios. Thus, this regimen was predicted to reliably achieve efficacious contezolid exposures independent of timing of switch from IV CZA to oral contezolid.IMPORTANCEThis study provides the population pharmacokinetic rationale for the dosage regimen of the intravenous (IV) prodrug contezolid acefosamil (CZA) followed by oral contezolid. We developed the first integrated population model for the pharmacokinetics of the MRX-1352 intermediate prodrug, active contezolid, and its main metabolite MRX-1320 based on data from three clinical studies in healthy volunteers and phase 2 patients. The proposed regimen was predicted to reliably achieve efficacious contezolid exposures independent of timing of switch from IV CZA to oral contezolid.
Collapse
Affiliation(s)
- Jürgen B. Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Edward Fang
- MicuRx Pharmaceuticals, Inc., Foster City, California, USA
| | - Martin E. Stryjewski
- Department of Medicine, Division of Infectious Diseases, Centro de Educación Médica e Investigaciones Clínicas, Buenos Aires, Argentina
| | - Wen Wang
- MicuRx Pharmaceuticals, Inc., Foster City, California, USA
| | | | | | - Barry Hafkin
- MicuRx Pharmaceuticals, Inc., Foster City, California, USA
| |
Collapse
|
3
|
Bachorz RA, Pastwińska J, Nowak D, Karaś K, Karwaciak I, Ratajewski M. The application of machine learning methods to the prediction of novel ligands for ROR γ/ROR γT receptors. Comput Struct Biotechnol J 2023; 21:5491-5505. [PMID: 38022699 PMCID: PMC10663739 DOI: 10.1016/j.csbj.2023.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
In this work, we developed and applied a computational procedure for creating and validating predictive models capable of estimating the biological activity of ligands. The combination of modern machine learning methods, experimental data, and the appropriate setup of molecular descriptors led to a set of well-performing models. We thoroughly inspected both the methodological space and various possibilities for creating a chemical feature space. The resulting models were applied to the virtual screening of the ZINC20 database to identify new, biologically active ligands of RORγ receptors, which are a subfamily of nuclear receptors. Based on the known ligands of RORγ, we selected candidates and calculate their predicted activities with the best-performing models. We chose two candidates that were experimentally verified. One of these candidates was confirmed to induce the biological activity of the RORγ receptors, which we consider proof of the efficacy of the proposed methodology.
Collapse
Affiliation(s)
- Rafał A. Bachorz
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Joanna Pastwińska
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Damian Nowak
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Kaja Karaś
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Iwona Karwaciak
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| | - Marcin Ratajewski
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, Łódź, 93-232, Poland
| |
Collapse
|
4
|
Imarah AA, Jabir MS, Abood AH, Sulaiman GM, Albukhaty S, Mohammed HA, Khan RA, Al-Kuraishy HM, Al-Gareeb AI, Al-Azzawi WK, A Najm MA, Jawad SF. Graphene oxide-induced, reactive oxygen species-mediated mitochondrial dysfunctions and apoptosis: high-dose toxicity in normal cells. Nanomedicine (Lond) 2023; 18:875-887. [PMID: 37470184 DOI: 10.2217/nnm-2023-0129] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023] Open
Abstract
Aim: The cytotoxic effects of graphene oxide nanoparticles (GONPs) using MTT assays, observance of apoptotic markers, and oxidative stress were outlined. Materials & methods: Rat embryonic fibroblasts (REFs) and human epithelial breast cells (HBLs) were used at 250, 500 and 750 μg/ml concentrations. Results: Significant cytotoxic and apoptotic effects were observed. Analyses of CYP2E1 and malondialdehyde concentrations in REF and HBL-100 cell lines after exposing to GONPs confirmed the nanomaterials toxicity. However, the glutathione levels in REF and HBL-100 cell lines showed a substantial reduction compared with the control. The cytochrome CYP2E1, glutathione, malondialdehyde and caspase-3 alterations provided a plausible interlinked relationship. Conclusion: The study confirmed the GONPs cytotoxic effects on REF and HBL-100 cell lines. The outcome suggested caution in wide-spread applications of GONPs, which could have implications for occupational health also.
Collapse
Affiliation(s)
- Ameer A Imarah
- Department of Biology, Faculty of Science, University of Kufa, Kufa 540011, Iraq
| | - Majid S Jabir
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, 10066, Iraq
| | - Ali H Abood
- Department of Biology, Faculty of Science, University of Kufa, Kufa 540011, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, 10066, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan 62001, Iraq
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Al-Azhar University, Cairo 11371, Egypt
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology, Medicine and Therapeutics, College of Medicine, Mustansiriyah University, PO Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology, Medicine and Therapeutics, College of Medicine, Mustansiriyah University, PO Box 14132, Baghdad, Iraq
| | | | - Mazin A A Najm
- Department of Pharmaceutical Chemistry, College of Pharmacy, Al-Ayen University, Thi-Qar 64001, Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001, Hillah, Babylon, Iraq
| |
Collapse
|
5
|
Coe KJ, Feinstein M, Higgins JW, Leung P, Scott BP, Skaptason J, Tam Y, Volak LP, Kinong J, Bittner A, McAllister H, Lim NM, Hack M, Koudriakova T. Characterization of JNJ-2482272 [4-(4-Methyl-2-(4-(Trifluoromethyl)Phenyl)Thiazole-5-yl) Pyrimidine-2-Amine] As a Strong Aryl Hydrocarbon Receptor Activator in Rat and Human. Drug Metab Dispos 2022; 50:1064-1076. [PMID: 35680134 DOI: 10.1124/dmd.121.000825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
[4-(4-Methyl-2-(4-(trifluoromethyl)phenyl)thiazole-5-yl)pyrimidine-2-amine] (JNJ-2482272), under investigation as an anti-inflammatory agent, was orally administered to rats once daily at 60 mg/kg for 6 consecutive days. Despite high plasma exposure after single administration (Cmax of 7.1 μM), JNJ-2482272 had plasma concentrations beneath the lower limit of quantification (3 ng/ml) after 6 consecutive days of dosing. To determine if JNJ-2482272 is an autoinducer in rats, plated rat hepatocytes were treated with JNJ-2482272 for 2 days. The major hydroxylated metabolites of JNJ-2482272 were isolated and characterized by mass spectrometry and NMR analyses. Compared with the vehicle-treated cells, a concentration-dependent increase was observed in the formation of phase I- and II-mediated metabolites coinciding with greater expression of cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) in rat hepatocytes. CYP1A1, CYP1A2, CYP1B1, and UGT1A6 transcripts were predominantly induced, suggesting that JNJ-2482272 is an activator of the aryl hydrocarbon receptor (AhR). In a human AhR reporter assay, JNJ-2482272 demonstrated potent AhR activation with an EC50 value of 0.768 nM, a potency more comparable to the strong AhR activator and toxin 2,3,7,8-tetrachloro-dibenzodioxin than to weaker AhR activators 3-methylcholanthrene, β-naphthoflavone, and omeprazole. In plated human hepatocytes, JNJ-2482272 induced CYP1A1 gene expression with an EC50 of 20.4 nM and increased CYP1A activity >50-fold from basal levels. In human recombinant P450s, JNJ-2482272 was exclusively metabolized by the CYP1 family of enzymes and most rapidly by CYP1A1. The summation of these in vitro findings bridges the in vivo conclusion that JNJ-2482272 is a strong autoinducer in rats and potentially in humans through potent AhR activation. SIGNIFICANCE STATEMENT: Drugs that induce their own metabolism (autoinducers) can lack sustained exposures for pharmacology and safety assessment hindering their development. JNJ-2482272 is demonstrated herein as a strong aryl hydrocarbon receptor (AhR) activator and CYP1A autoinducer, explaining its near complete loss of exposure after repeat administration in rat, which is likely translatable to human (if progressed further) considering its nanomolar potency comparable to "classical" AhR ligands like 2,3,7,8-tetrachloro-dibenzo-dioxin despite bearing a "nonclassical" drug structure.
Collapse
Affiliation(s)
- Kevin J Coe
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Mark Feinstein
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - J William Higgins
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Perry Leung
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Brian P Scott
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Judy Skaptason
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Yuen Tam
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Laurie P Volak
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Jennifer Kinong
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Anton Bittner
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Heather McAllister
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Nathan M Lim
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Michael Hack
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| | - Tatiana Koudriakova
- Janssen Research & Development, L.L.C., San Diego, California (K.J.C., M.F., P.L., B.P.S., L.P.V., H.M., N.M.L., M.H., T.K.); Janssen Research & Development, L.L.C., San Francisco, California (Y.T.), Neurocrine Biosciences, Inc, San Diego, California (J.S.); Pfizer, San Diego, California (J.K.); Turnstone Biologics, La Jolla, California (A.B.); and Trestle Biotherapeutics, San Diego, California (J.W.H.)
| |
Collapse
|
6
|
Chen MH, Zhang SH, Jia SM, Wang LJ, Ma WL. In vitro biotransformation of tris(1,3-dichloro-2-propyl) phosphate and triphenyl phosphate by mouse liver microsomes: Kinetics and key CYP isoforms. CHEMOSPHERE 2022; 288:132504. [PMID: 34627810 DOI: 10.1016/j.chemosphere.2021.132504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 06/13/2023]
Abstract
As the result of the phase-out on polybrominated diphenyl ethers, organophosphate flame retardants (OPFRs) were widely used as substitutes in the world. Previous studies found that OPFRs were frequently detected in environmental, biological, and human samples. Considering their adverse effects, the absorption, bioaccumulation, metabolism and internal exposure processes of OPFRs attracted more attentions recently, especially for aryl-OPFR and Cl-OPFRs. In the present study, the biotransformation, metabolic kinetics and related CYP450 isoforms of typical Cl-OPFR (tris(1,3-dichloro-2-propyl) phosphate: TDCPP) and aryl-OPFR (triphenyl phosphate: TPhP) were studied in vitro by mouse liver microsomes. Metabolomic analysis revealed that TDCPP may be easier to bio-accumulate in organisms than TPhP, which can be explained by their metabolic rates and half-life values (TDCPP: t1/2 = 1.8083 h; TPhP: t1/2 = 0.1531 h). CYP2E1, CYP2D6, CYP1A2 and CYP2C19 were suggested to be the specific enzymes for the biotransformation of TDCPP via associated inhibition assay. CYP2E1 was the primary CYP450 isoform of metabolism in vitro for TPhP. These findings may provide new insights for the potential mechanism of hepatotoxicity in mammals induced by OPFRs and the detoxification process of OPFRs in hepatocytes.
Collapse
Affiliation(s)
- Mei-Hong Chen
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China; Heilongjiang Provincial Key Laboratory of Polar Environment and Ecosystem (HPKL-PEE), Harbin, 150090, China
| | - Sheng-Hu Zhang
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment of the People's Republic of China, Nanjing, 210042, China
| | - Shi-Ming Jia
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China; Heilongjiang Provincial Key Laboratory of Polar Environment and Ecosystem (HPKL-PEE), Harbin, 150090, China
| | - Li-Jun Wang
- Department of Physiology, Harbin Medical University, Harbin, 150081, China
| | - Wan-Li Ma
- International Joint Research Center for Persistent Toxic Substances (IJRC-PTS), State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150090, China; Heilongjiang Provincial Key Laboratory of Polar Environment and Ecosystem (HPKL-PEE), Harbin, 150090, China.
| |
Collapse
|
7
|
Buser MC, Pohl HR, Abadin HG. Windows of sensitivity to toxic chemicals in the development of the endocrine system: an analysis of ATSDR's toxicological profile database. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2022; 32:437-454. [PMID: 32495642 PMCID: PMC7714698 DOI: 10.1080/09603123.2020.1772204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/18/2020] [Indexed: 06/11/2023]
Abstract
This review utilizes the robust database of literature contained in toxicological profiles developed by the Agency for Toxic Substances and Disease Registry. The aim was to use this database to identify developmental toxicity studies reporting alterations in hormone levels in the developing fetus and offspring and identify windows of sensitivity. We identified 74 oral exposure studies in rats that provided relevant information on 30 chemicals from 21 profiles. Most studies located provided information on thyroid hormones, with fewer studies on anterior pituitary, adrenal medulla, ovaries, and testes. No studies pertaining to hormones of the posterior pituitary, pancreas, or adrenal cortex were located. The results demonstrate that development of the endocrine system may be affected by exposure to environmental contaminants at many different points, including gestational and/or lactational exposure. Moreover, this review demonstrates the need for more developmental toxicity studies focused on the endocrine system and specifically alterations in hormone levels.
Collapse
Affiliation(s)
- M C Buser
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| | - H R Pohl
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| | - H G Abadin
- US Department of Health and Human Services, Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA, USA
| |
Collapse
|
8
|
Smythe MA, Burns C, Liu Q, Garwood CL. Potential Dexamethasone-Direct Oral Anticoagulant Drug Interaction: Is This a Concern in COVID? Ann Pharmacother 2021; 56:319-329. [PMID: 34137279 DOI: 10.1177/10600280211025042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To evaluate the literature on a potential dexamethasone-direct oral anticoagulant (DOAC) drug interaction and provide management considerations with COVID hypercoagulability. DATA SOURCES A search of EMBASE, PubMed, and Google Scholar (January 1990 to May 2021), limited to the English language, using applicable search terms resulted in 137 articles, with 21 relevant articles included. Regulatory agency and clinical guidance documents were also reviewed. STUDY SELECTION AND DATA EXTRACTION Included articles describe in vitro or in vivo animal or human data for dexamethasone induction of cytochrome P450 (CYP) 3A4 or P-glycoprotein (P-gp). DATA SYNTHESIS Dexamethasone has the potential to interact with the DOACs via CYP3A4 and/or P-gp induction. Only apixaban and rivaroxaban have CYP3A4 metabolism. Dexamethasone can increase CYP3A4 activity by up to 70% and reduce the area under the concentration-time curve (AUC) of CYP3A4 substrates by >40%, which is consistent with criteria for a weak CYP inducer. In rodents, dexamethasone P-gp induction is associated with AUC reductions of 20% to 50%. Human data are lacking. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Severe COVID-19 infection is associated with hypercoagulability. Although heparins are the preferred anticoagulants for hospitalized COVID-19 patients, DOACs are being utilized. Dexamethasone is recommended for hospitalized COVID-19 patients requiring supplemental oxygen. The concurrent use of dexamethasone and apixaban or rivaroxaban in such patients carries the potential for reduced anticoagulant effect during a state of heightened thrombotic risk. CONCLUSIONS Concurrent use of dexamethasone and apixaban or rivaroxaban in hospitalized COVID-19 patients with laboratory evidence of COVID coagulopathy should be avoided until higher-quality data are available.
Collapse
Affiliation(s)
- Maureen A Smythe
- Wayne State University, Detroit, MI, USA.,Beaumont Hospital, Royal Oak, MI, USA
| | | | | | - Candice L Garwood
- Wayne State University, Detroit, MI, USA.,Detroit Medical Center, MI, USA
| |
Collapse
|
9
|
Bachmann F, Duthaler U, Meyer Zu Schwabedissen HE, Puchkov M, Huwyler J, Haschke M, Krähenbühl S. Metamizole is a Moderate Cytochrome P450 Inducer Via the Constitutive Androstane Receptor and a Weak Inhibitor of CYP1A2. Clin Pharmacol Ther 2020; 109:1505-1516. [PMID: 33336382 PMCID: PMC8247900 DOI: 10.1002/cpt.2141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/07/2020] [Indexed: 01/24/2023]
Abstract
Metamizole is an analgesic and antipyretic drug used intensively in certain countries. Previous studies have shown that metamizole induces cytochrome (CYP) 2B6 and possibly CYP3A4. So far, it is unknown whether metamizole induces additional CYPs and by which mechanism. Therefore, we assessed the activity of 6 different CYPs in 12 healthy male subjects before and after treatment with 3 g of metamizole per day for 1 week using a phenotyping cocktail approach. In addition, we investigated whether metamizole induces CYPs by an interaction with the constitutive androstane receptor (CAR) or the pregnane X receptor (PXR) in HepaRG cells. In the clinical study, we confirmed a moderate induction of CYP2B6 (decrease in the efavirenz area under the plasma concentration time curve (AUC) by 79%) and 3A4 (decrease in the midazolam AUC by 68%) by metamizole. In addition, metamizole weakly induced CYP2C9 (decrease in the flurbiprofen AUC by 22%) and moderately CYP2C19 (decrease in the omeprazole AUC by 66%) but did not alter CYP2D6 activity. In addition, metamizole weakly inhibited CYP1A2 activity (1.79‐fold increase in the caffeine AUC). We confirmed these results in HepaRG cells, where 4‐MAA, the principal metabolite of metamizole, induced the mRNA expression of CYP2B6, 2C9, 2C19, and 3A4. In HepaRG cells with a stable knockout of PXR or CAR, we could demonstrate that CYP induction by 4‐MAA depends on CAR and not on PXR. In conclusion, metamizole is a broad CYP inducer by an interaction with CAR and an inhibitor of CYP1A2. Regarding the widespread use of metamizole, these findings are of substantial clinical relevance.
Collapse
Affiliation(s)
- Fabio Bachmann
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Urs Duthaler
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Maxim Puchkov
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jörg Huwyler
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Manuel Haschke
- Clinical Pharmacology and Toxicology, Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology & Toxicology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Enzymatic deinduction phenomenon and clinical implications with a focus on direct-acting oral anticoagulants. Blood Coagul Fibrinolysis 2020; 31:283-286. [DOI: 10.1097/mbc.0000000000000914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Is there a sex difference in adult salivary clearance of caffeine (1,3,7-trimethylpurine-2,6-dione)? J Oral Biol Craniofac Res 2020; 10:20-22. [DOI: 10.1016/j.jobcr.2020.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/24/2019] [Accepted: 01/30/2020] [Indexed: 11/22/2022] Open
|
12
|
Bulutoglu B, Rey-Bedón C, Mert S, Tian L, Jang YY, Yarmush ML, Usta OB. A comparison of hepato-cellular in vitro platforms to study CYP3A4 induction. PLoS One 2020; 15:e0229106. [PMID: 32106230 PMCID: PMC7046200 DOI: 10.1371/journal.pone.0229106] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
In vitro studies of drug toxicity and drug-drug interactions are crucial for drug development efforts. Currently, the utilization of primary human hepatocytes (PHHs) is the de facto standard for this purpose, due to their functional xenobiotic response and drug metabolizing CYP450 enzyme metabolism. However, PHHs are scarce, expensive, require laborious maintenance, and exhibit lot-to-lot heterogeneity. Alternative human in vitro platforms include hepatic cell lines, which are easy to access and maintain, and induced pluripotent stem cell (iPSC) derived hepatocytes. In this study, we provide a direct comparison of drug induced CYP3A4 and PXR expression levels of PHHs, hepatic cell lines Huh7 and HepG2, and iPSC derived hepatocyte like cells. Confluently cultured Huh7s exhibited an improved CYP3A4 expression and were inducible by up to 4.9-fold, and hepatocytes differentiated from human iPSCs displayed a 3.3-fold CYP3A4 induction. In addition, an increase in PXR expression levels was observed in both hepatic cell lines and iPSC derived hepatocytes upon rifampicin treatment, whereas a reproducible increase in PXR expression was not achieved in PHHs. Our results indicate that both hepatoma originated cell lines and iPSCs may provide alternative sources to primary hepatocytes, providing reliable and reproducible results for CYP3A4/PXR metabolism, upon in vitro maturation. This study may serve as a guide for the selection of suitable and feasible in vitro platforms for drug-drug interaction and toxicology studies.
Collapse
Affiliation(s)
- Beyza Bulutoglu
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Camilo Rey-Bedón
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Safak Mert
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Lipeng Tian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yoon-Young Jang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Martin L. Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States of America
| | - O. Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| |
Collapse
|
13
|
Sekretarska J, Szczepaniak J, Sosnowska M, Grodzik M, Kutwin M, Wierzbicki M, Jaworski S, Bałaban J, Daniluk K, Sawosz E, Chwalibog A, Strojny B. Influence of Selected Carbon Nanostructures on the CYP2C9 Enzyme of the P450 Cytochrome. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E4149. [PMID: 31835701 PMCID: PMC6947289 DOI: 10.3390/ma12244149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022]
Abstract
Carbon nanostructures have recently gained significant interest from scientists due to their unique physicochemical properties and low toxicity. They can accumulate in the liver, which is the main expression site of cytochrome P450 (CYP450) enzymes. These enzymes play an important role in the metabolism of exogenous compounds, such as drugs and xenobiotics. Altered activity or expression of CYP450 enzymes may lead to adverse drug effects and toxicity. The objective of this study was to evaluate the influence of three carbon nanostructures on the activity and expression at the mRNA and protein levels of CYP2C9 isoenzyme from the CYP2C subfamily: Diamond nanoparticles, graphite nanoparticles, and graphene oxide platelets. The experiments were conducted using two in vitro models. A microsome model was used to assess the influence of the three-carbon nanostructures on the activity of the CYP2C9 isoenzyme. The CYP2C9 gene expression at the mRNA and protein levels was determined using a hepatoma-derived cell line HepG2. The experiments have shown that all examined nanostructures inhibit the enzymatic activity of the studied isoenzymes. Moreover, a decrease in the expression at the mRNA and protein levels was also observed. This indicates that despite low toxicity, the nanostructures can alter the enzymatic function of CYP450 enzymes, and the molecular pathways involved in their expression.
Collapse
Affiliation(s)
- Justyna Sekretarska
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Jarosław Szczepaniak
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Malwina Sosnowska
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Marta Grodzik
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Marta Kutwin
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Mateusz Wierzbicki
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Sławomir Jaworski
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Jaśmina Bałaban
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Karolina Daniluk
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - Ewa Sawosz
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| | - André Chwalibog
- Department of Veterinary and Animal Sciences, University of Copenhagen, Groennegaardsvej 3, 1870 Frederiksberg, Denmark;
| | - Barbara Strojny
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland; (J.S.); (J.S.); (M.S.); (M.G.); (M.K.); (M.W.); (S.J.); (J.B.); (K.D.); (E.S.)
| |
Collapse
|
14
|
Bulutoglu B, Mert S, Rey-Bedón C, Deng SL, Yarmush ML, Usta OB. Rapid maturation of the hepatic cell line Huh7 via CDK inhibition for PXR dependent CYP450 metabolism and induction. Sci Rep 2019; 9:15848. [PMID: 31676845 PMCID: PMC6825149 DOI: 10.1038/s41598-019-52174-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/12/2019] [Indexed: 12/26/2022] Open
Abstract
CYP3A4, a cytochrome P450 enzyme regulated by the nuclear receptor PXR, is involved in most of the drug metabolizing pathways. Studying the regulation/induction of CYP3A4 and PXR is critical in toxicology and drug-drug interaction (DDI) studies. Primary human hepatocytes constitute the preferred in vitro platform for drug development efforts. However, they are expensive, scarce and heterogeneous. Hepatic cell lines, such as Huh7, could provide a cost-effective alternative, however, they express negligible amounts of CYP450s and PXR. In this study, we show that dinaciclib, a potent cyclin dependent kinase inhibitor, significantly increases the basal CYP3A4 and PXR levels in 24 hours. We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. More importantly, through a direct demonstration using amiodarone and rifampicin as model drugs, we showed that matured Huh7s present a suitable platform for DDI studies.
Collapse
Affiliation(s)
- Beyza Bulutoglu
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Safak Mert
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Camilo Rey-Bedón
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Sarah L Deng
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA.
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| | - O Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, 02114, USA.
| |
Collapse
|
15
|
Miyauchi Y, Tanaka Y, Nagata K, Yamazoe Y, Mackenzie PI, Yamada H, Ishii Y. UDP-Glucuronosyltransferase (UGT)-mediated attenuations of cytochrome P450 3A4 activity: UGT isoform-dependent mechanism of suppression. Br J Pharmacol 2019; 177:1077-1089. [PMID: 31660580 DOI: 10.1111/bph.14900] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 09/19/2019] [Accepted: 09/28/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND PURPOSE Cytochrome P450 (CYP, P450) 3A4 is involved in the metabolism of 50% of drugs and its catalytic activity in vivo is not explained only by hepatic expression levels. We previously demonstrated that UDP-glucuronosyltransferase (UGT) 2B7 suppressed CYP3A4 activity through an interaction. In the present study, we target UGT1A9 as another candidate modulator of CYP3A4. EXPERIMENTAL APPROACH We prepared co-expressed enzymes using the baculovirus-insect cell expression system and compared CYP3A4 activity in the presence and absence of UGT1A9. Wistar rats were treated with dexamethasone and liver microsomes were used to elucidate the role of CYP3A-UGT1A interactions. KEY RESULTS UGT1A9 and UGT2B7 interacted with and suppressed CYP3A4. Kinetic analyses showed that both of the UGTs significantly reduced Vmax of CYP3A4 activity. In addition, C-terminal truncated mutants of UGT1A9 and UGT2B7 still retained the suppressive capacity. Dexamethasone treatment induced hepatic CYP3As and UGT1As at different magnitudes. Turnover of CYP3A was enhanced about twofold by this treatment. CONCLUSION AND IMPLICATIONS The changes of kinetic parameters suggested that UGT1A9 suppressed CYP3A4 activity with almost the same mechanism as UGT2B7. The luminal domain of UGTs contains the suppressive interaction site(s), whereas the C-terminal domain may contribute to modulating suppression in a UGT isoform-specific manner. CYP3A-UGT1A interaction seemed to be disturbed by dexamethasone treatment and the suppression was partially cancelled. CYP3A4-UGT interactions would help to better understand the causes of inter/intra-individual differences in CYP3A4 activity.
Collapse
Affiliation(s)
- Yuu Miyauchi
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiyoshi Nagata
- Department of Environmental and Health Science, School of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yasushi Yamazoe
- Food Safety Commission, Cabinet Office, Government of Japan, Tokyo, Japan
| | - Peter I Mackenzie
- Department of Clinical Pharmacology, Flinders Medical Centre and Flinders University, Adelaide, SA, Australia
| | - Hideyuki Yamada
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Parvez MK, Rishi V. Herb-Drug Interactions and Hepatotoxicity. Curr Drug Metab 2019; 20:275-282. [PMID: 30914020 DOI: 10.2174/1389200220666190325141422] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND In recent times, herbals or phytomedicines have become very popular due to their global acceptance as a complementary and alternative remedy. While modern drugs are commercially available only after laboratory validations, clinical trials, as well as approval from drug regulatory authorities, majority of the marketed herbal products lack such scientific evidence of efficacy and safety. This results in herb or herb-drug interaction induced unfavorable clinical outcomes without crucial documentation on their temporal relations and concomitant use. METHODS An online literature search for peer-reviewed articles was conducted on the PubMed, Europe PMC, Medline and Google Scholar portals, using the phrases: complementary & alternative medicine, traditional Chinese medicine, herb-drug interaction, mechanisms of herb-drug interaction, herb-induced toxicity, herbal hepatotoxicity and causality, traditional medicine, viral hepatitis, etc. RESULTS The retrieved data showed that globally, patients are attracted to herbal remedies with the misconception that these are completely safe and therefore, use them simultaneously with prescription drugs. Notably, there exists a potential risk of herb-drug interactions leading to some adverse side effects, including hepatotoxicity. The toxicological effect of a drug or herb is due to the inhibition of drug metabolizing enzymes (e.g., cytochrome P450), including interactions with certain prescription drugs through various mechanisms. Several cases of hepatotoxicity due to use of herbals in viral hepatitis-related liver diseases have been recently reported. However, limited experimental data and clinical evidence on herbal pharmacokinetics hamper the evaluation and reporting of adverse reactions and the underlying mechanisms. CONCLUSION Herb-drug interaction related morbidity is thus an emerging serious public health issue with broad implications for clinicians, pharmaceutical industries and health authorities. Nonetheless, despite increasing recognition of herb-drug interaction, a standard system for interaction prediction and evaluation is still nonexistent. This review article discusses the herb-drug interactions related hepatotoxicity and underlying mechanisms, including drug metabolizing enzymes and their regulation.
Collapse
Affiliation(s)
- Mohammad K Parvez
- Department of Pharmacognosy, King Saud University College of Pharmacy, Riyadh 11451, Saudi Arabia
| | - Vikas Rishi
- National Agri-Food Biotechnology Institute, Mohali, Punjab 140306, India
| |
Collapse
|
17
|
Weiss J. Herb⁻Drug Interaction Potential of Anti-Borreliae Effective Extracts from Uncaria tomentosa (Samento) and Otoba parvifolia (Banderol) Assessed In Vitro. Molecules 2018; 24:molecules24010137. [PMID: 30602711 PMCID: PMC6337116 DOI: 10.3390/molecules24010137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/23/2018] [Accepted: 12/26/2018] [Indexed: 12/14/2022] Open
Abstract
Samento (extract from Uncaria tomentosa) and Banderol (extract from Otoba parvifolia) have been demonstrated to have anti-inflammatory and antimicrobial properties, e.g., against different morphological forms of Borrelia burgdorferi. However, there is hardly any data on the pharmacological safety of these two herbal medicines. This in vitro study aimed at scrutinizing their possible characteristics as perpetrators in pharmacokinetic herbal–drug interactions. Inhibition of cytochrome P450 enzymes (CYPs) was quantified by commercial kits and inhibition of drug transporters by use of fluorescent probe substrates. Induction was quantified by real-time RT-PCR and activation of pregnane x receptor (PXR) and aryl hydrocarbon receptor (AhR) by reporter gene assays. Organic anion transporting polypeptide 1B1 (OATP1B1) (IC50 = 0.49 ± 0.28%) and OATP1B3 (IC50 = 0.65 ± 0.29%) were potently inhibited by Banderol, but only weakly by Samento. CYP3A4 was inhibited about 40% at a Samento concentration of 1%. Samento significantly induced mRNA expression of CYP2J2, UGT1A3, UGT1A9, ABCB1, and SLCO1B1 and strongly activated PXR, but hardly AhR. In conclusion, the perpetrator profiles of Samento and Banderol for herb–drug interactions completely differ. Clinical studies are strongly recommended to clarify whether the effects observed in vitro are of clinical relevance.
Collapse
Affiliation(s)
- Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
18
|
Hosogi J, Ohashi R, Maeda H, Fujita K, Ushiki J, Kuwabara T, Yamamoto Y, Imamura T. An iminium ion metabolite hampers the production of the pharmacologically active metabolite of a multikinase inhibitor KW-2449 in primates: irreversible inhibition of aldehyde oxidase and covalent binding with endogenous proteins. Biopharm Drug Dispos 2018; 39:164-174. [DOI: 10.1002/bdd.2123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 02/01/2018] [Accepted: 02/06/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Jun Hosogi
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Rui Ohashi
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Hiroshi Maeda
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Kazuhiro Fujita
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Junko Ushiki
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Takashi Kuwabara
- Translational Research Unit; Research and Development Division, Kyowa Hakko Kirin Co., Ltd; 1188 Shimotogari, Nagaizumi-cho, Sunto-gun Shizuoka 411-8731 Japan
| | - Yorihiro Yamamoto
- School of Bioscience and Biotechnology; Tokyo University of Technology; 1404-1 Katakura-cho, Hachioji Tokyo 192-0983 Japan
| | - Toru Imamura
- School of Bioscience and Biotechnology; Tokyo University of Technology; 1404-1 Katakura-cho, Hachioji Tokyo 192-0983 Japan
| |
Collapse
|
19
|
Di (2-Ethylhexyl) Phthalate and Its Role in Developing Cholestasis: An In Vitro Study on Different Liver Cell Types. J Pediatr Gastroenterol Nutr 2018; 66:e28-e35. [PMID: 29095348 DOI: 10.1097/mpg.0000000000001813] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Di(2-ethylhexyl) phthalate (DEHP) is a plasticizer used in many polyvinylchloride medical devices and is washed out easily. Thereby critically ill infants can become exposed to DEHP concentrations significantly exceeding the recommended threshold. We suspect DEHP to play an important role in the development of intestinal failure-associated liver disease. The aim of this study was therefore to determine the direct influence of DEHP on different liver cell types. METHODS HepG2, human upcyte hepatocytes, primary murine hepatocytes, LX-2, human upcyte hepatic stellate cells, and liver organoids were cultured with DEHP (0.5-500 μmol/L) and parameters including cytotoxicity, cell-cell interactions, and expression of metabolizing enzymes were investigated. RESULTS DEHP modulated the expression of xenobiotic metabolizing enzymes, reduced the formation of bile canaliculi and cell polarity, and inhibited Cyp-activity in hepatocytes. DEHP had a toxic effect on LX-2 and induced the fibrogenic activation of hepatic stellate cells. The mode of action of DEHP was different in monolayer cultures compared to 3D-liver organoids, which were more sensitive to DEHP. CONCLUSIONS This study suggests that DEHP modulates expression and activity of drug-detoxifying liver enzymes in humans at a clinically relevant concentration. Furthermore, it may contribute to the development of cholestasis and fibrosis. These findings strongly support the opinion, that there is a significant potential for serious adverse effects of DEHP derived from medical devices on human health, especially in very young infants with immature livers.
Collapse
|
20
|
Chang HY, Chen CJ, Ma WC, Cheng WK, Lin YN, Lee YR, Chen JJ, Lim YP. Modulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) activation by ursolic acid (UA) attenuates rifampin-isoniazid cytotoxicity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 36:37-49. [PMID: 29157826 DOI: 10.1016/j.phymed.2017.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/30/2017] [Accepted: 09/24/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Interactions between transcriptional inducers of cytochrome P450 (CYP450) enzymes and therapeutic drugs may be prevented by antagonizing the activation of a nuclear receptor (NR), pregnane X receptor (PXR, NR1I2), thus improving therapeutic efficacy. PURPOSE In the present study, we aim to identify that ursolic acid (UA), a widely distributed pentacyclic triterpene, may act as an effective antagonist of PXR and its sister NR receptor, constitutive androstane receptor (CAR, NR1I3). METHODS The hepatocellular carcinoma cell line, HepG2, was used to evaluate the promoter activity of PXR and CAR target genes, CYP3A4 and CYP2B6, respectively. Catalytic activities, mRNA, and protein expression of CYP3A4 and CYP2B6 were evaluated in a differentiated HepaRG cell line. Coregulation of PXR with coregulators on CYP3A4 promoter response elements was also been characterized. RESULTS Transient transfection assays showed that UA effectively attenuated CYP3A4 and CYP2B6 promoter activities mediated by rifampin (RIF, human PXR agonist) and CITCO (human CAR agonist). These inhibitory effects were well correlated with the expression and catalytic activities of CYP3A4 and CYP2B6. Furthermore, the interaction of co-regulators with PXR and the transcriptional complexes in the CYP3A4 promoter activity and CYP3A4 promoter xenobiotic response element (everted repeat 6, ER6), respectively, were disrupted in the presence of UA. UA showed an antagonistic effect against PXR, and reversed the cytotoxic effects of isoniazid (INH) induced by RIF. Taken together, these results show that UA inhibits the transactivation effects of PXR and CAR, and reduces the expression and function of CYP3A4 and CYP2B6. CONCLUSION The present study suggests that UA could be a powerful agent for reducing potentially dangerous interactions between transcriptional inducers of CYP enzymes and therapeutic drugs.
Collapse
Affiliation(s)
- Hsiao-Yun Chang
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chao-Jung Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chih Ma
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Wai-Kok Cheng
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yen-Ning Lin
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Ying-Ray Lee
- Translational Medicine Research Center, Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Jih-Jung Chen
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming University, Taipei, Taiwan
| | - Yun-Ping Lim
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan; Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
21
|
Krämer M, Broecker S, Madea B, Hess C. Confirmation of metabolites of the neuroleptic drug prothipendyl using human liver microsomes, specific CYP enzymes and authentic forensic samples-Benefit for routine drug testing. J Pharm Biomed Anal 2017; 145:517-524. [PMID: 28756170 DOI: 10.1016/j.jpba.2017.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 11/29/2022]
Abstract
Metabolism of the tricyclic azaphenothiazine neuroleptic drug prothipendyl was investigated with in vitro studies using human liver microsomes but also specific isoforms of cytochrome P450 (CYP) enzymes. Identification and analysis of metabolites was done by liquid chromatography (LC) coupled with quadrupole time of flight mass spectrometry (LC-QTOF-MS) as well as triple quadrupole mass spectrometry (LC-QQQ-MS). Results of the herein presented study revealed the proof of various demethylated and oxidized metabolites (-CH2, -C2H4, four derivatives of prothipendyl +O and three derivatives of prothipendyl -CH2+O). Metabolic reactions of prothipendyl were mainly catalyzed by CYP enzymes CYP1A2, CYP2D6, CYP2C19 and CYP3A4. N-demethyl-prothipendyl was predominantly formed by isoforms CYP2C19 and CYP1A2, while particularly the CYP isoenzyme 3A4 was responsible for the formation of prothipendyl sulfoxide. To confirm the formation of previously identified metabolites in vivo, cardiac blood samples that were tested positive for prothipendyl during routine drug testing and serum and urine samples, collected after a voluntary intake of prothipendyl, were analyzed by LC-QQQ-MS. All metabolites of prothipendyl were proven in these authentic specimens. Neither in serum samples nor in urine samples, a prolonged detectability of metabolites in comparison to prothipendyl could be demonstrated.
Collapse
Affiliation(s)
- M Krämer
- University Bonn, Institute of Forensic Medicine, Department of Forensic Toxicology, Stiftsplatz 12, 53111 Bonn, Germany.
| | - S Broecker
- Broeckers Solutions - Consulting and Support, Dyrotzer Straße 8, 13583 Berlin, Germany.
| | - B Madea
- University Bonn, Institute of Forensic Medicine, Department of Forensic Toxicology, Stiftsplatz 12, 53111 Bonn, Germany.
| | - C Hess
- University Bonn, Institute of Forensic Medicine, Department of Forensic Toxicology, Stiftsplatz 12, 53111 Bonn, Germany.
| |
Collapse
|
22
|
AbdulHameed MDM, Ippolito DL, Wallqvist A. Predicting Rat and Human Pregnane X Receptor Activators Using Bayesian Classification Models. Chem Res Toxicol 2016; 29:1729-1740. [DOI: 10.1021/acs.chemrestox.6b00227] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mohamed Diwan M. AbdulHameed
- Department
of Defense Biotechnology High Performance Computing Software Applications
Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, 504 Scott Street, Fort Detrick, Maryland 21702, United States
| | - Danielle L. Ippolito
- U.S. Army Center for Environmental Health Research, 568 Doughten Drive, Fort
Detrick, Maryland 21702, United States
| | - Anders Wallqvist
- Department
of Defense Biotechnology High Performance Computing Software Applications
Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, 504 Scott Street, Fort Detrick, Maryland 21702, United States
| |
Collapse
|
23
|
Zhang YM, Chang MJ, Yang XS, Han X. In silico investigation of agonist activity of a structurally diverse set of drugs to hPXR using HM-BSM and HM-PNN. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2016; 36:463-468. [PMID: 27376821 DOI: 10.1007/s11596-016-1609-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/28/2016] [Indexed: 10/21/2022]
Abstract
The human pregnane X receptor (hPXR) plays a critical role in the metabolism, transport and clearance of xenobiotics in the liver and intestine. The hPXR can be activated by a structurally diverse of drugs to initiate clinically relevant drug-drug interactions. In this article, in silico investigation was performed on a structurally diverse set of drugs to identify critical structural features greatly related to their agonist activity towards hPXR. Heuristic method (HM)-Best Subset Modeling (BSM) and HM-Polynomial Neural Networks (PNN) were utilized to develop the linear and non-linear quantitative structure-activity relationship models. The applicability domain (AD) of the models was assessed by Williams plot. Statistically reliable models with good predictive power and explain were achieved (for HM-BSM, r (2)=0.881, q LOO (2) =0.797, q EXT (2) =0.674; for HM-PNN, r (2)=0.882, q LOO (2) =0.856, q EXT (2) =0.655). The developed models indicated that molecular aromatic and electric property, molecular weight and complexity may govern agonist activity of a structurally diverse set of drugs to hPXR.
Collapse
Affiliation(s)
- Yi-Ming Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China
| | - Mei-Jia Chang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xu-Shu Yang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiao Han
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
24
|
Prakash C, Zuniga B, Song CS, Jiang S, Cropper J, Park S, Chatterjee B. Nuclear Receptors in Drug Metabolism, Drug Response and Drug Interactions. NUCLEAR RECEPTOR RESEARCH 2015; 2:101178. [PMID: 27478824 PMCID: PMC4963026 DOI: 10.11131/2015/101178] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Orally delivered small-molecule therapeutics are metabolized in the liver and intestine by phase I and phase II drug-metabolizing enzymes (DMEs), and transport proteins coordinate drug influx (phase 0) and drug/drug-metabolite efflux (phase III). Genes involved in drug metabolism and disposition are induced by xenobiotic-activated nuclear receptors (NRs), i.e. PXR (pregnane X receptor) and CAR (constitutive androstane receptor), and by the 1α, 25-dihydroxy vitamin D3-activated vitamin D receptor (VDR), due to transactivation of xenobiotic-response elements (XREs) present in phase 0-III genes. Additional NRs, like HNF4-α, FXR, LXR-α play important roles in drug metabolism in certain settings, such as in relation to cholesterol and bile acid metabolism. The phase I enzymes CYP3A4/A5, CYP2D6, CYP2B6, CYP2C9, CYP2C19, CYP1A2, CYP2C8, CYP2A6, CYP2J2, and CYP2E1 metabolize >90% of all prescription drugs, and phase II conjugation of hydrophilic functional groups (with/without phase I modification) facilitates drug clearance. The conjugation step is mediated by broad-specificity transferases like UGTs, SULTs, GSTs. This review delves into our current understanding of PXR/CAR/VDR-mediated regulation of DME and transporter expression, as well as effects of single nucleotide polymorphism (SNP) and epigenome (specified by promoter methylation, histone modification, microRNAs, long non coding RNAs) on the expression of PXR/CAR/VDR and phase 0-III mediators, and their impacts on variable drug response. Therapeutic agents that target epigenetic regulation and the molecular basis and consequences (overdosing, underdosing, or beneficial outcome) of drug-drug/drug-food/drug-herb interactions are also discussed. Precision medicine requires understanding of a drug's impact on DME and transporter activity and their NR-regulated expression in order to achieve optimal drug efficacy without adverse drug reactions. In future drug screening, new tools such as humanized mouse models and microfluidic organs-on-chips, which mimic the physiology of a multicellular environment, will likely replace the current cell-based workflow.
Collapse
Affiliation(s)
- Chandra Prakash
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- William Carey University College of Osteopathic Medicine, 498 Tucsan Ave, Hattiesburg, Mississipi 39401
| | - Baltazar Zuniga
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- University of Texas at Austin, 2100 Comal Street, Austin, Texas 78712
| | - Chung Seog Song
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Shoulei Jiang
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Jodie Cropper
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Sulgi Park
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
| | - Bandana Chatterjee
- Department of Molecular Medicine/Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, Texas Research Park, 15355 Lambda Drive, San Antonio, Texas 78245
- South Texas Veterans Health Care System, Audie L Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, Texas 78229
| |
Collapse
|
25
|
Screening of a chemical library reveals novel PXR-activating pharmacologic compounds. Toxicol Lett 2015; 232:193-202. [DOI: 10.1016/j.toxlet.2014.10.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/02/2014] [Accepted: 10/06/2014] [Indexed: 11/21/2022]
|
26
|
Yu Y, Liu Y, Li Q, Sun J, Lin H, Liu G. Effects of Guanxinning injection on rat cytochrome P450 isoforms activities in vivo and in vitro. Xenobiotica 2014; 45:481-7. [PMID: 25495039 DOI: 10.3109/00498254.2014.993002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. We aimed to investigate the regulatory effects of Guanxinning injection (GXNI) on activities of cytochrome P1A2 (CYP1A2), CYP2C11, CYP2D1 and CYP3A1/2 by probe drugs in rats in vivo and in vitro. 2. GXNI-treated and blank control groups were administered GXNI and physiological saline by caudal vein for 14 days consecutively, then they were given the probe drugs of caffeine (10 mg/kg), tolbutamide (10 mg/kg), metoprolol (20 mg/kg) and dapsone (10 mg/kg) by intraperitoneal injection. The blood samples were collected at different times for ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) analysis. Changes of the pharmacokinetics parameters between the GXNI-treated and the blank control groups were used to evaluate the effects of GXNI on the four CYP450 isoforms in rats in vivo. After blood collection, the livers of rats were taken and made microsomes for in vitro tests. The relevant metabolites of phenacetin, tolbutamide, dextromethorphan and testosterone were analyzed quantitatively by high-performance liquid chromatography (HPLC) after microsome incubation. The statistical differences between the two groups were observed to detect the effects of GXNI on the four CYP450 isoforms in rats in vitro. 3. The in vivo and in vitro results demonstrated that GXNI could induce CYP1A2 activity in rats, but had no significant effects on CYP2C11, CYP2D1 and CYP3A1/2.
Collapse
Affiliation(s)
- Yue Yu
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University , Harbin , China
| | | | | | | | | | | |
Collapse
|
27
|
Liu G, Asanoma K, Takao T, Tsukimori K, Uchi H, Furue M, Kato K, Wake N. Aryl hydrocarbon receptor SNP -130 C/T associates with dioxins susceptibility through regulating its receptor activity and downstream effectors including interleukin 24. Toxicol Lett 2014; 232:384-92. [PMID: 25445724 DOI: 10.1016/j.toxlet.2014.11.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 12/25/2022]
Abstract
Dioxins are persistent environmental pollutants that cause multiple adverse health effects in humans, mainly through binding to the ligand-activated transcription factor, aryl hydrocarbon receptor (AhR). Genetic variation in AhR may modulate the susceptibility to dioxins. In this study, we aimed to evaluate the effects of the single nucleotide polymorphism (SNP) -130 C/T in the AhR promoter on dioxin-inducible gene transcription, and to investigate interleukin-24 (IL-24) and interleukin-1β (IL-1β) as proxies for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure. Using primary human chorionic stromal cells, we found that cells with the TT genotype showed higher AhR mRNA and protein levels than did those of the CC genotype. Microarray was carried out to analyze the gene expression profiles of cells (CC and TT genotype) after exposing the cells to TCDD. Several genes associated with human disorders were more highly up-regulated in cells of the TT genotype. Higher up-regulation of IL-24 and IL-1β mRNA in cells with the TT genotype was observed. Furthermore, blood samples from 64 Yusho patients who were accidentally exposed to high concentrations of dioxins were analyzed for the genotype, dioxins concentrations and serum levels of IL-24 and IL-1β. We observed higher serum IL-24 levels and lower serum IL-1β levels in Yusho patients with the TT genotype than in those with the CC genotype. AhR SNP -130 C/T affects serum IL-24 and IL-1β levels, independently of serum dioxins concentrations in Yusho patients. Our observations demonstrate that SNP -130 C/T modulates AhR expression and expression levels of IL-24 and IL-1β, and suggest an association of AhR SNP -130 C/T with the susceptibility to dioxins.
Collapse
Affiliation(s)
- Ge Liu
- Department of Genomic Epidemiology, Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuo Asanoma
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Tomoka Takao
- Department of Genomic Epidemiology, Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiyomi Tsukimori
- Department of Obstetrics, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Hiroshi Uchi
- Research and Clinical Center for Yusho and Dioxins, Kyushu University Hospital, Fukuoka, Japan; Department of Dermatology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Masutaka Furue
- Research and Clinical Center for Yusho and Dioxins, Kyushu University Hospital, Fukuoka, Japan; Department of Dermatology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Norio Wake
- Department of Genomic Epidemiology, Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
28
|
Banerjee M, Robbins D, Chen T. Targeting xenobiotic receptors PXR and CAR in human diseases. Drug Discov Today 2014; 20:618-28. [PMID: 25463033 DOI: 10.1016/j.drudis.2014.11.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/28/2014] [Accepted: 11/17/2014] [Indexed: 12/20/2022]
Abstract
Nuclear receptors such as the pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are xenobiotic receptors regulating not only drug metabolism and disposition but also various human diseases such as cancer, diabetes, inflammatory disease, metabolic disease and liver diseases, suggesting that PXR and CAR are promising targets for drug discovery. Consequently, there is an urgent need to discover and develop small molecules that target these PXR- and/or CAR-mediated human-disease-related pathways for relevant therapeutic applications. This review proposes approaches to target PXR and CAR, either individually or simultaneously, in the context of various human diseases, taking into consideration the structural differences between PXR and CAR.
Collapse
Affiliation(s)
- Monimoy Banerjee
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Delira Robbins
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
29
|
Banerjee M, Chen T. Thiazide-like diuretic drug metolazone activates human pregnane X receptor to induce cytochrome 3A4 and multidrug-resistance protein 1. Biochem Pharmacol 2014; 92:389-402. [PMID: 25181459 PMCID: PMC4252478 DOI: 10.1016/j.bcp.2014.08.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 01/14/2023]
Abstract
Human pregnane X receptor (hPXR) regulates the expression of drug-metabolizing enzyme cytochrome P450 3A4 (CYP3A4) and drug transporters such as multidrug-resistance protein 1 (MDR1). PXR can be modulated by small molecules, including Federal Drug Administration (FDA)-approved drugs, thus altering drug metabolism and causing drug-drug interactions. To determine the role of FDA-approved drugs in PXR-mediated regulation of drug metabolism and clearance, we screened 1481 FDA-approved small-molecule drugs by using a luciferase reporter assay in HEK293T cells and identified the diuretic drug metolazone as an activator of hPXR. Our data showed that metolazone activated hPXR-mediated expression of CYP3A4 and MDR1 in human hepatocytes and intestine cells and increased CYP3A4 promoter activity in various cell lines. Mammalian two-hybrid assays showed that hPXR recruits its co-activator SRC-1 upon metolazone binding in HepG2 cells, explaining the mechanism of hPXR activation. To understand the role of other commonly-used diuretics in hPXR activation and the structure-activity relationship of metolazone, thiazide and non-thiazide diuretics drugs were also tested but only metolazone activates hPXR. To understand the molecular mechanism, docking studies and mutational analysis were carried out and showed that metolazone binds in the ligand-binding pocket and interacts with mostly hydrophobic amino acid residues. This is the first report showing that metolazone activates hPXR. Because activation of hPXR might cause drug-drug interactions, metolazone should be used with caution for drug treatment in patients undergoing combination therapy.
Collapse
Affiliation(s)
- Monimoy Banerjee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Mail Stop 1000, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Mail Stop 1000, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| |
Collapse
|
30
|
Awortwe C, Manda VK, Avonto C, Khan SI, Khan IA, Walker LA, Bouic PJ, Rosenkranz B. Echinacea purpurea up-regulates CYP1A2, CYP3A4 and MDR1 gene expression by activation of pregnane X receptor pathway. Xenobiotica 2014; 45:218-29. [PMID: 25377539 DOI: 10.3109/00498254.2014.973930] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1.This study investigated the mechanism underlying Echinacea-mediated induction of CYP1A2, CYP3A4 and MDR1 in terms of human pregnane X receptor (PXR) activation. 2.Crude extracts and fractions of Echinacea purpurea were tested for PXR activation in HepG2 cells by a reporter gene assay. Quantitative real-time PCR was carried out to determine their effects on CYP1A2 and CYP3A4 mRNA expressions. Capsules and fractions were risk ranked as high, intermediate and remote risk of drug-metabolizing enzymes induction based on EC50 values determined for respective CYPs. 3. Fractions F1, F2 and capsule (2660) strongly activated PXR with 5-, 4- and 3.5-fold increase in activity, respectively. Echinacea preparations potentiated up-regulation of CYP1A2, CYP3A4 and MDR1 via PXR activation. 4.Thus E. purpurea preparations cause herb-drug interaction by up-regulating CYP1A2, CYP3A4 and P-gp via PXR activation.
Collapse
Affiliation(s)
- Charles Awortwe
- Division of Clinical Pharmacology, Faculty of Medicine and Health Sciences, University of Stellenbosch , Cape Town , South Africa
| | | | | | | | | | | | | | | |
Collapse
|
31
|
De Martin S, Gabbia D, Albertin G, Sfriso MM, Mescoli C, Albertoni L, Paliuri G, Bova S, Palatini P. Differential effect of liver cirrhosis on the pregnane X receptor-mediated induction of CYP3A1 and 3A2 in the rat. Drug Metab Dispos 2014; 42:1617-26. [PMID: 25030308 DOI: 10.1124/dmd.114.058511] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Conflicting results have been obtained by clinical studies investigating the effect of liver cirrhosis on enzyme induction. Because ethical concerns do not give consent for methodologically rigorous studies in humans, we addressed this question by examining the effect of the prototypical inducer dexamethasone (DEX) on the pregnane X receptor (PXR)-mediated induction of CYP3A1 and 3A2 in a validated animal model of liver cirrhosis obtained by exposure of rats to carbon tetrachloride. For this purpose, we assessed mRNA levels, protein expressions, and enzymatic activities of both CYP3A enzymes, as well as mRNA and protein expressions of PXR in rat populations rigorously stratified according to the severity of liver insufficiency. Constitutive mRNA and protein expressions of CYP3A1 and CYP3A2 and their basal enzyme activities were not affected by liver dysfunction. DEX treatment markedly increased steady-state mRNA level, protein content, and enzymatic activity of CYP3A1 in healthy and cirrhotic rats, irrespective of the degree of liver dysfunction. On the contrary, the inducing effect of DEX on gene and protein expressions and enzyme activity of CYP3A2 was preserved in moderate liver insufficiency, whereas it was greatly curtailed when liver insufficiency became severe. mRNA and protein expressions of PXR were neither reduced by liver dysfunction nor increased by DEX treatment. These results indicate that even the inducibility of cytochrome P450 isoforms under the transcriptional control of the same nuclear receptor may be differentially affected by cirrhosis and may partly explain why conflicting results were obtained by human studies.
Collapse
Affiliation(s)
- Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Giovanna Albertin
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Maria Martina Sfriso
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Claudia Mescoli
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Laura Albertoni
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Giovanna Paliuri
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Sergio Bova
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| | - Pietro Palatini
- Department of Pharmaceutical and Pharmacological Sciences (S.D.M., D.G., G.P., S.B., P.P.), Department of Molecular Medicine, Section of Anatomy (G.A., M.M.S.), and Department of Medicine, Surgical Pathology and Cytopathology Unit (C.M., L.A.), University of Padova, Padova, Italy
| |
Collapse
|
32
|
Chen H, Zhang X, Feng Y, Rui W, Shi Z, Wu L. Bioactive components of Glycyrrhiza uralensis mediate drug functions and properties through regulation of CYP450 enzymes. Mol Med Rep 2014; 10:1355-62. [PMID: 24939038 DOI: 10.3892/mmr.2014.2331] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 04/14/2014] [Indexed: 11/06/2022] Open
Abstract
Glycyrrhiza uralensis (G. uralensis) is a common medicinal plant that has mainly been used to modulate the pharmaceutical activity of herbal medicines. Although G. uralensis has been shown to affect the expression and activity of the key metabolic enzyme cytochrome P450 (CYP450), the detailed mechanism of this process has yet to be elucidated. The present study aimed to elucidate the effects of bioactive components of G. uralensis on different isoforms of CYP450 and determine the ability of these components to modulate drug properties. In the present study, mRNA levels of CYP1A2, CYP2D6, CYP2E1, and CYP3A4 were investigated by quantitative polymerase chain reaction (qPCR) in HepG2 cells following treatment with the major bioactive compounds of G. uralensis. The activity of CYP450 enzymes was investigated in human liver microsomes using the cocktail probe drug method, and the metabolites of specific probes were detected by UPLC‑MS/MS. The effects of G. uralensis on CYP450 were assessed using bioinformatics network analysis. Several compounds from G. uralensis had various effects on the expression and activity of multiple CYP450 isoforms. The majority of the compounds analysed the inhibited expression of CYP2D6 and CYP3A4. Several CYP isoforms were differentially modulated depending on the specific compound and dose tested. In conclusion, the present study suggested that G. uralensis influenced the expression and activity of CYP450 enzymes. Therefore, caution should be taken when G. uralensis is co‑administered with drugs that are known to be metabolized by CYP450. This study contributed to the knowledge of the mechanisms by which this medicinal plant, commonly known as licorice, modulates drug efficacy.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaomei Zhang
- Center Laboratory, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Yifan Feng
- Center Laboratory, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Wen Rui
- Center Laboratory, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Zhongfeng Shi
- Center Laboratory, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Lirong Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
33
|
Abstract
The pregnane X receptor (PXR) and constitutive androstane receptor (CAR), 2 closely related and liver-enriched members of the nuclear receptor superfamily, and aryl hydrocarbon receptor (AhR), a nonnuclear receptor transcription factor (TF), are major receptors/TFs regulating the expression of genes for the clearance and detoxification of xenobiotics. They are hence defined as "xenobiotic receptors". Recent studies have demonstrated that PXR, CAR and AhR also regulate the expression of key proteins involved in endobiotic responses such as the metabolic homeostasis of lipids, glucose, and bile acid, and inflammatory processes. It is suggested that the functions of PXR, CAR and AhR may be closely implicated in the pathogeneses of metabolic vascular diseases, such as hyperlipidemia, atherogenesis, and hypertension. Therefore, manipulation of the activities of these receptors may provide novel strategies for the treatment of vascular diseases. Here, we review the pathophysiological roles of PXR, CAR and AhR in the vascular system.
Collapse
Affiliation(s)
- Lei Xiao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University
| | | | | |
Collapse
|
34
|
Bissonnette L, Bergeron MG. Next revolution in the molecular theranostics of infectious diseases: microfabricated systems for personalized medicine. Expert Rev Mol Diagn 2014; 6:433-50. [PMID: 16706745 DOI: 10.1586/14737159.6.3.433] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The molecular diagnosis of infectious diseases is currently going through a revolution sustained by the regulatory approval of amplification tests that have been shown to be equivalent or superior to existing gold standard methods. The recent approval of a microarray system for the pharmacogenomic profiling of cytochrome P450-mediated drug metabolism is paving the way to novel, rapid, sensitive, robust and economical microfabricated systems for point-of-care diagnostics, which are utilized closer and closer to the patient's bedside. These systems will enable the multiparametric genetic evaluation of several medical conditions, including infectious diseases. This forecoming revolution will position molecular theranostics in a broader integrated view of personalized medicine, which exploits genetic information from microbes and human hosts to optimize patient management and disease treatment.
Collapse
Affiliation(s)
- Luc Bissonnette
- Département de Biologie Médicale (Microbiologie), Faculté de Médecine, Université Laval, Québec City, Canada.
| | | |
Collapse
|
35
|
Lux R, Wärntges S, Bergner S, Kütting B. [Improvement of medication safety by identification of genetically predisposed subjects. Personalized clinical strategies and regulatory advices]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2013; 56:1545-56. [PMID: 24170084 DOI: 10.1007/s00103-013-1827-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Because adverse drug events (ADEs) have a high socio-economic impact there is an urgent need for effective prevention. In addition to process-related avoidable errors personalised approaches for the prevention of ADEs should also focus on genetic polymorphisms as potential causative agents. AIM Using five case reports as examples therapeutic modalities are described to illustrate the clinical impact of prospective testing aimed at estimating the individual risk of susceptible subjects. MATERIAL AND METHODS The role of the HLA system, the cytochrome P450 family, other metabolic enzymes and transport proteins are described to illustrate the broad range of genetic susceptibility. It is shown, why, when and for whom pretherapeutic tests on genetic polymorphisms are recommended to reduce the risk of ADEs. RESULTS The determination of genetic susceptibility is already implemented in clinical practice prior to (1) carbamazepine therapy in south-east Asians and (2) treatment with abacavir independent of ethnicity. Before prescribing carbamazepine or abacavir, it is recommended that therapeutic decisions be based on these test results. CONCLUSION The broad application of personalised medicine used as an effective tool for minimizing ADE risks is limited by the evidence-based benefit for the patient on the one hand and the costs of the test on the other hand.
Collapse
Affiliation(s)
- R Lux
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Bonn, Deutschland
| | | | | | | |
Collapse
|
36
|
Floreani M, De Martin S, Gabbia D, Barbierato M, Nassi A, Mescoli C, Orlando R, Bova S, Angeli P, Gola E, Sticca A, Palatini P. Severe liver cirrhosis markedly reduces AhR-mediated induction of cytochrome P450 in rats by decreasing the transcription of target genes. PLoS One 2013; 8:e61983. [PMID: 23626760 PMCID: PMC3633963 DOI: 10.1371/journal.pone.0061983] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/15/2013] [Indexed: 12/13/2022] Open
Abstract
Although the induction of cytochrome P450 (CYP) has long been investigated in patients with cirrhosis, the question whether liver dysfunction impairs the response to CYP inducers still remains unresolved. Moreover, the mechanism underlying the possible effect of cirrhosis on induction has not been investigated. Since ethical constraints do not permit methodologically rigorous studies in humans, this question was addressed by investigating the effect of the prototypical inducer benzo[a]pyrene (BP) on CYP1A1 and CYP1A2 in cirrhotic rats stratified according to the severity of liver dysfunction. We simultaneously assessed mRNA level, protein expression and enzymatic activity of the CYP1A enzymes, as well as mRNA and protein expressions of the aryl hydrocarbon receptor (AhR), which mediates the BP effect. Basal mRNA and protein expressions of CYP1A1 were virtually absent in both healthy and cirrhotic rats. On the contrary, CYP1A2 mRNA, protein and enzyme activity were constitutively present in healthy rats and decreased significantly as liver function worsened. BP treatment markedly increased the concentrations of mRNA and immunodetectable protein, and the enzymatic activities of both CYP1A enzymes to similar levels in healthy and non-ascitic cirrhotic rats. Induced mRNA levels, protein expressions and enzymatic activities of both CYPs were much lower in ascitic rats and were proportionally reduced. Both constitutive and induced protein expressions of AhR were significantly lower in ascitic than in healthy rats. These results indicate that the inducibility of CYP1A enzymes is well preserved in compensated cirrhosis, whereas it is markedly reduced when liver dysfunction becomes severe. Induction appears to be impaired at the transcriptional level, due to the reduced expression of AhR, which controls the transcription of CYP1A genes.
Collapse
MESH Headings
- Animals
- Ascites/chemically induced
- Ascites/enzymology
- Ascites/genetics
- Ascites/pathology
- Benzo(a)pyrene
- Cytochrome P-450 CYP1A1/genetics
- Cytochrome P-450 CYP1A1/metabolism
- Cytochrome P-450 CYP1A2
- Cytochromes/genetics
- Cytochromes/metabolism
- Enzyme Induction/genetics
- Gene Expression Regulation
- Liver/enzymology
- Liver/pathology
- Liver Cirrhosis, Experimental/chemically induced
- Liver Cirrhosis, Experimental/enzymology
- Liver Cirrhosis, Experimental/genetics
- Liver Cirrhosis, Experimental/pathology
- Liver Function Tests
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Severity of Illness Index
- Transcription, Genetic
Collapse
Affiliation(s)
- Maura Floreani
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Alberto Nassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Claudia Mescoli
- Surgical Pathology and Cytopathology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Rocco Orlando
- Laboratory of Experimental Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Sergio Bova
- Laboratory of Experimental Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Paolo Angeli
- Laboratory of Experimental Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Elisabetta Gola
- Laboratory of Experimental Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Antonietta Sticca
- Laboratory of Experimental Hepatology, Department of Medicine, University of Padova, Padova, Italy
| | - Pietro Palatini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- * E-mail:
| |
Collapse
|
37
|
Floreani M, De Martin S, Gabbia D, Barbierato M, Nassi A, Mescoli C, Orlando R, Bova S, Angeli P, Gola E, Sticca A, Palatini P. Severe liver cirrhosis markedly reduces AhR-mediated induction of cytochrome P450 in rats by decreasing the transcription of target genes. PLoS One 2013. [PMID: 23626760 DOI: 10.1371/journal.pone.0061983;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although the induction of cytochrome P450 (CYP) has long been investigated in patients with cirrhosis, the question whether liver dysfunction impairs the response to CYP inducers still remains unresolved. Moreover, the mechanism underlying the possible effect of cirrhosis on induction has not been investigated. Since ethical constraints do not permit methodologically rigorous studies in humans, this question was addressed by investigating the effect of the prototypical inducer benzo[a]pyrene (BP) on CYP1A1 and CYP1A2 in cirrhotic rats stratified according to the severity of liver dysfunction. We simultaneously assessed mRNA level, protein expression and enzymatic activity of the CYP1A enzymes, as well as mRNA and protein expressions of the aryl hydrocarbon receptor (AhR), which mediates the BP effect. Basal mRNA and protein expressions of CYP1A1 were virtually absent in both healthy and cirrhotic rats. On the contrary, CYP1A2 mRNA, protein and enzyme activity were constitutively present in healthy rats and decreased significantly as liver function worsened. BP treatment markedly increased the concentrations of mRNA and immunodetectable protein, and the enzymatic activities of both CYP1A enzymes to similar levels in healthy and non-ascitic cirrhotic rats. Induced mRNA levels, protein expressions and enzymatic activities of both CYPs were much lower in ascitic rats and were proportionally reduced. Both constitutive and induced protein expressions of AhR were significantly lower in ascitic than in healthy rats. These results indicate that the inducibility of CYP1A enzymes is well preserved in compensated cirrhosis, whereas it is markedly reduced when liver dysfunction becomes severe. Induction appears to be impaired at the transcriptional level, due to the reduced expression of AhR, which controls the transcription of CYP1A genes.
Collapse
MESH Headings
- Animals
- Ascites/chemically induced
- Ascites/enzymology
- Ascites/genetics
- Ascites/pathology
- Benzo(a)pyrene
- Cytochrome P-450 CYP1A1/genetics
- Cytochrome P-450 CYP1A1/metabolism
- Cytochrome P-450 CYP1A2
- Cytochromes/genetics
- Cytochromes/metabolism
- Enzyme Induction/genetics
- Gene Expression Regulation
- Liver/enzymology
- Liver/pathology
- Liver Cirrhosis, Experimental/chemically induced
- Liver Cirrhosis, Experimental/enzymology
- Liver Cirrhosis, Experimental/genetics
- Liver Cirrhosis, Experimental/pathology
- Liver Function Tests
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Severity of Illness Index
- Transcription, Genetic
Collapse
Affiliation(s)
- Maura Floreani
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP, Miwa G, Ni L, Kumar G, McLeod J, Obach SR, Roberts S, Roe A, Shah A, Snikeris F, Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA. The Conduct of In Vitro and In Vivo Drug-Drug Interaction Studies: A PhRMA Perspective. J Clin Pharmacol 2013. [DOI: 10.1177/0091270003252519] [Citation(s) in RCA: 201] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
39
|
Zhou H, Tong Z, McLeod JF. “Cocktail” Approaches and Strategies in Drug Development: Valuable Tool or Flawed Science? J Clin Pharmacol 2013; 44:120-34. [PMID: 14747420 DOI: 10.1177/0091270003261333] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is an increasing interest in the simultaneous administration of several probe substrates to characterize the activity of multiple drug-metabolizing enzymes, the so-called "cocktail" approach. However, this method remains controversial and is being investigated more extensively. No general consensus has emerged on the applicability of this approach in clinical investigation and during drug development. The objective of the article is to review this important yet specialized technique, as well as its merits, drawbacks, and potential application in drug development. Among the two-, three-, four-, five-, and six-drug in vivo cocktails previously evaluated in humans, a variety of substrate probe combinations have been studied. Some probe combinations have been validated not to interact in vivo and have been useful in characterizing drug-drug interaction potential and metabolic enzyme induction in humans. For drug candidates that affect two or more in vitro pathways or are potential gene inducers, the use of a cocktail approach may facilitate the rapid delineation of the drug candidate's drug interaction potential. It may also offer the potential of providing clear guidance on safely conducting larger clinical studies and limiting comedication restrictions to only those likely to be clinically relevant.
Collapse
Affiliation(s)
- Honghui Zhou
- Clinical Pharmacology, Wyeth Research, 500 Arcola Road, Collegeville, PA 19426, USA
| | | | | |
Collapse
|
40
|
Flavonoids and Polymer Derivatives as CYP3A4 Inhibitors for Improved Oral Drug Bioavailability. J Pharm Sci 2013. [DOI: 10.1002/jps.23382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
41
|
Maganda B, Heudi O, Cortinovis A, Picard F, Kretz O, Minzi O. A fast and reliable reversed phase high performance liquid chromatography method for simultaneous determination of selected anti-retroviral and lumefantrine in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 919-920:52-60. [PMID: 23411019 DOI: 10.1016/j.jchromb.2013.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 01/10/2013] [Accepted: 01/13/2013] [Indexed: 10/27/2022]
Abstract
A fast and reliable high performance liquid chromatography (HPLC) method with UV diode array detection for simultaneous quantitative analysis of the anti-retroviral drugs, nevirapine (NVP) and efavirenz (EFV) and the anti-malarial, lumefantrine (LUM) in human plasma has been developed and validated. The sample preparation consisted of a plasma protein precipitation with 0.5% acetic acid acetonitrile solution containing the internal standard halofantrine (HALO) prior the LC-analysis. Chromatographic separation was carried out on a Acclaim Polar Advantage C(16), column (150 mm × 4.6 mm, particle size, 3 μm) using a gradient of mobile phase made of 0.01% TFA in 0.1M ammonium acetate (solvent A) and 0.1% TFA in acetonitrile (solvent B). The separation of NVP, EFV, LUM and HALO was achieved within 17 min at a flow rate of 1.0 mL/min and detections were initially performed at three wavelengths, 275 nm (NVP), 255 nm (EFV), and 300 nm (LUM). The method selectivity was demonstrated in six different human plasma batches. In addition, several concomitant drugs were analyzed under our experimental conditions and none of them co-eluted with EFV, NVP and LUM. This demonstrated that our method is highly selective. Calibration graphs plotted with seven concentrations in duplicate for each compound were linear between the selected ranges with a regression coefficient (R(2)) greater than 0.998. Absolute extraction recovery for NVP, EFV and LUM were 99%, 98.6 and 102%, respectively. Inter- and intra-day coefficients of variation for LUM, EFV and NVP were ≤10%. The lower limits of quantification were 0.125 μg/mL for LUM and 0.250 μg/mL for both EFV and NVP. Intra- and inter-assay relative standard deviation values were found to be less than 15% at the concentrations examined (0.125-10.0 μg/mL for LUM and 0.250-15.0 μg/mL for both EFV and NVP). The present method was successfully implemented in Tanzania and only one wavelength (255 nm) was used to measure samples of patients receiving either NVP or EFV in combination with LUM. The concentration found in human plasma samples for all three compounds were within the calibration range. This makes our method particularly applicable and useful to resource-limited settings.
Collapse
Affiliation(s)
- Betty Maganda
- Department of Pharmaceutics, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | | | | | | | | |
Collapse
|
42
|
Development of in silico filters to predict activation of the pregnane X receptor (PXR) by structurally diverse drug-like molecules. Bioorg Med Chem 2012; 20:5352-65. [PMID: 22560839 DOI: 10.1016/j.bmc.2012.04.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/28/2012] [Accepted: 04/07/2012] [Indexed: 01/22/2023]
Abstract
The pregnane X receptor (PXR), a member of the nuclear hormone superfamily, regulates the expression of several enzymes and transporters involved in metabolically relevant processes. The significant induction of CYP450 enzymes by PXR, in particular CYP3A4, might significantly alter the metabolism of prescribed drugs. In order to early identify molecules in drug discovery with a potential to activate PXR as antitarget, we developed fast and reliable in silico filters by ligand-based QSAR techniques. Two classification models were established on a diverse dataset of 434 drug-like molecules. A second augmented set allowed focusing on interesting regions in chemical space. These classifiers are based on decision trees combined with a genetic algorithm based variable selection to arrive at predictive models. The classifier for the first dataset on 29 descriptors showed good performance on a test set with a correct classification of both 100% for PXR activators and non-activators plus 87% for activators and 83% for non-activators in an external dataset. The second classifier then correctly predicts 97% activators and 91% non-activators in a test set and 94% for activators and 64% non-activators in an external set of 50 molecules, which still qualifies for application as a filter focusing on PXR activators. Finally a quantitative model for PXR activation for a subset of these molecules was derived using a regression-tree approach combined with GA variable selection. This final model shows a predictive r(2) of 0.774 for the test set and 0.452 for an external set of 33 molecules. Thus, the combination of these filters consistently provide guidelines for lowering PXR activation in novel candidate molecules.
Collapse
|
43
|
Pajares B, Torres E, Trigo JM, Sáez MI, Ribelles N, Jiménez B, Alba E. Tyrosine kinase inhibitors and drug interactions: a review with practical recommendations. Clin Transl Oncol 2012; 14:94-101. [DOI: 10.1007/s12094-012-0767-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochim Biophys Acta Mol Basis Dis 2011; 1812:956-63. [PMID: 21295138 DOI: 10.1016/j.bbadis.2011.01.014] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 12/14/2022]
Abstract
The pregnane X receptor (PXR, NR1I2) is a ligand activated transcription factor that belongs to the nuclear hormone receptor (NR) superfamily. PXR is highly expressed in the liver and intestine, but low levels of expression have also been found in many other tissues. PXR plays an integral role in xenobiotic and endobiotic metabolism by regulating the expression of drug-metabolizing enzymes and transporters, as well as genes implicated in the metabolism of endobiotics. PXR exerts its transcriptional regulation by binding to its DNA response elements as a heterodimer with the retinoid X receptor (RXR) and recruitment of a host of coactivators. The biological and physiological implications of PXR activation are broad, ranging from drug metabolism and drug-drug interactions to the homeostasis of numerous endobiotics, such as glucose, lipids, steroids, bile acids, bilirubin, retinoic acid, and bone minerals. The purpose of this article is to provide an overview on the transcriptional circuits and metabolic relevance controlled by PXR. This article is part of a Special Issue entitled: Translating Nuclear Receptors from Health to Disease.
Collapse
|
45
|
Lee S, Poet TS, Smith JN, Hjerpe AL, Gunawan R, Timchalk C. Impact of repeated nicotine and alcohol coexposure on in vitro and in vivo chlorpyrifos dosimetry and cholinesterase inhibition. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2011; 74:1334-1350. [PMID: 21899407 DOI: 10.1080/15287394.2011.567958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Chlorpyrifos (CPF) is an organophosphorus insecticide, and neurotoxicity results from inhibition of acetylcholinesterase (AChE) by its metabolite, chlorpyrifos-oxon. Routine consumption of alcohol and tobacco modifies metabolic and physiological processes impacting the metabolism and pharmacokinetics of other xenobiotics, including pesticides. This study evaluated the influence of repeated ethanol and nicotine coexposure on in vivo CPF dosimetry and cholinesterase (ChE) response (ChE- includes AChE and/or butyrylcholinesterase (BuChE)). Hepatic microsomes were prepared from groups of naive, ethanol-only (1 g/kg/d, 7 d, po), and ethanol + nicotine (1 mg/kg/d 7 d, sc)-treated rats, and the in vitro metabolism of CPF was evaluated. For in vivo studies, rats were treated with saline or ethanol (1 g/kg/d, po) + nicotine (1 mg/kg/d, sc) in addition to CPF (1 or 5 mg/kg/d, po) for 7 d. The major CPF metabolite, 3,5,6-trichloro-2-pyridinol (TCPy), in blood and urine and the plasma ChE and brain acetylcholinesterase (AChE) activities were measured in rats. There were differences in pharmacokinetics, with higher TCPy peak concentrations and increased blood TCPy AUC in ethanol + nicotine groups compared to CPF only (approximately 1.8- and 3.8-fold at 1 and 5 mg CPF doses, respectively). Brain AChE activities after ethanol + nicotine treatments showed significantly less inhibition following repeated 5 mg CPF/kg dosing compared to CPF only (96 ± 13 and 66 ± 7% of naive at 4 h post last CPF dosing, respectively). Although brain AChE activity was minimal inhibited for the 1-mg CPF/kg/d groups, the ethanol + nicotine pretreatment resulted in a similar trend (i.e., slightly less inhibition). No marked differences were observed in plasma ChE activities due to the alcohol + nicotine treatments. In vitro, CPF metabolism was not markedly affected by repeated ethanol or both ethanol + nicotine exposures. Compared with a previous study of nicotine and CPF exposure, there were no apparent additional exacerbating effects due to ethanol coexposure.
Collapse
Affiliation(s)
- S Lee
- Food and Drug Administration, Atlanta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
46
|
Mei N, Guo L, Fu PP, Fuscoe JC, Luan Y, Chen T. Metabolism, genotoxicity, and carcinogenicity of comfrey. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2010; 13:509-26. [PMID: 21170807 PMCID: PMC5894094 DOI: 10.1080/10937404.2010.509013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Comfrey has been consumed by humans as a vegetable and a tea and used as an herbal medicine for more than 2000 years. Comfrey, however, produces hepatotoxicity in livestock and humans and carcinogenicity in experimental animals. Comfrey contains as many as 14 pyrrolizidine alkaloids (PA), including 7-acetylintermedine, 7-acetyllycopsamine, echimidine, intermedine, lasiocarpine, lycopsamine, myoscorpine, symlandine, symphytine, and symviridine. The mechanisms underlying comfrey-induced genotoxicity and carcinogenicity are still not fully understood. The available evidence suggests that the active metabolites of PA in comfrey interact with DNA in liver endothelial cells and hepatocytes, resulting in DNA damage, mutation induction, and cancer development. Genotoxicities attributed to comfrey and riddelliine (a representative genotoxic PA and a proven rodent mutagen and carcinogen) are discussed in this review. Both of these compounds induced similar profiles of 6,7-dihydro-7-hydroxy-1-hydroxymethyl-5H-pyrrolizine (DHP)-derived DNA adducts and similar mutation spectra. Further, the two agents share common mechanisms of drug metabolism and carcinogenesis. Overall, comfrey is mutagenic in liver, and PA contained in comfrey appear to be responsible for comfrey-induced toxicity and tumor induction.
Collapse
Affiliation(s)
- Nan Mei
- Division of Genetic and Reproductive Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas 72079, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Candrilli SD, Manjunath R, Davis KL, Gidal BE. The association between antiepileptic drug and HMG-CoA reductase inhibitor co-medication and cholesterol management in patients with epilepsy. Epilepsy Res 2010; 91:260-6. [DOI: 10.1016/j.eplepsyres.2010.07.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/19/2010] [Accepted: 07/25/2010] [Indexed: 11/26/2022]
|
48
|
Mishra NK, Agarwal S, Raghava GPS. Prediction of cytochrome P450 isoform responsible for metabolizing a drug molecule. BMC Pharmacol 2010; 10:8. [PMID: 20637097 PMCID: PMC2912882 DOI: 10.1186/1471-2210-10-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 07/16/2010] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Different isoforms of Cytochrome P450 (CYP) metabolized different types of substrates (or drugs molecule) and make them soluble during biotransformation. Therefore, fate of any drug molecule depends on how they are treated or metabolized by CYP isoform. There is a need to develop models for predicting substrate specificity of major isoforms of P450, in order to understand whether a given drug will be metabolized or not. This paper describes an in-silico method for predicting the metabolizing capability of major isoforms (e.g. CYP 3A4, 2D6, 1A2, 2C9 and 2C19). RESULTS All models were trained and tested on 226 approved drug molecules. Firstly, 2392 molecular descriptors for each drug molecule were calculated using various softwares. Secondly, best 41 descriptors were selected using general and genetic algorithm. Thirdly, Support Vector Machine (SVM) based QSAR models were developed using 41 best descriptors and achieved an average accuracy of 86.02%, evaluated using fivefold cross-validation. We have also evaluated the performance of our model on an independent dataset of 146 drug molecules and achieved average accuracy 70.55%. In addition, SVM based models were developed using 26 Chemistry Development Kit (CDK) molecular descriptors and achieved an average accuracy of 86.60%. CONCLUSIONS This study demonstrates that SVM based QSAR model can predict substrate specificity of major CYP isoforms with high accuracy. These models can be used to predict isoform responsible for metabolizing a drug molecule. Thus these models can used to understand whether a molecule will be metabolized or not. This is possible to develop highly accurate models for predicting substrate specificity of major isoforms using CDK descriptors. A web server MetaPred has been developed for predicting metabolizing isoform of a drug molecule http://crdd.osdd.net/raghava/metapred/.
Collapse
Affiliation(s)
- Nitish K Mishra
- Bioinformatics Centre, Institute of Microbial Technology, Chandigarh, India
| | - Sandhya Agarwal
- Bioinformatics Centre, Institute of Microbial Technology, Chandigarh, India
| | | |
Collapse
|
49
|
Botts S, Ennulat D, Francke-Carroll S, Graham M, Maronpot RR, Mohutsky M. Introduction to Hepatic Drug Metabolizing Enzyme Induction in Drug Safety Evaluation Studies. Toxicol Pathol 2010; 38:796-8. [DOI: 10.1177/0192623310374330] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The following three articles represent the output of a combined effort initiated by the Scientific Regulatory Policy Committee of the Society of Toxicologic Pathology to provide a unified review of current scientific practices and relevant literature and provide suggestions regarding the recognition, interpretation, and risk assessment of hepatic drug metabolizing enzyme (DME) induction studies. The core objective was to provide a review that the scientific community including pathologists, regulatory scientists, toxicologists, investigative scientists, and others would find valuable for managing, designing, and interpreting toxicity studies supporting regulatory filings. Three working groups composed of scientists from industry, academia, and regulatory agencies were convened to review the available literature on important aspects of the interpretation and risk assessment of hepatic microsomal DME enzyme induction in three publications. The three reviews are as follows: “Effects of Hepatic Drug Metabolizing Enzyme Induction on Clinical Pathology Parameters in Animals and Man,” Toxicol Pathol “Hepatic Drug Metabolizing Enzyme Induction: Microscopic and Ultrastructural Appearance,” Toxicol Pathol “Hepatic Drug Metabolizing Enzyme Induction and Implications for Preclinical and Clinical Risk Assessment,” Toxicol Pathol The purpose of this introduction is not to summarize the articles but rather to frame the series and to provide a common mechanistic introduction.
Collapse
Affiliation(s)
- Suzanne Botts
- Hoffmann–La Roche Inc., Chair, SRPC Initiative, Nutley, NJ, USA
| | - Daniela Ennulat
- GlaxoSmithKline, Chair, Clinical Pathology Working Group, King of
Prussia, PA, USA
| | | | - Mark Graham
- AstraZeneca, Member, Clinical Pathology Working Group, Loughborough,
Leicestershire, UK
| | - Robert R. Maronpot
- Maronpot Consulting LLC, Chair, Anatomic and Ultrastructural Pathology
Working Group, Raleigh, NC, USA
| | - Michael Mohutsky
- Lilly Research Laboratories, Eli Lilly and Company, Member, Risk
Assessment Working Group, Indianapolis, IN, USA
| |
Collapse
|
50
|
Punyawudho B, Cloyd JC, Leppik IE, Ramsay RE, Marino SE, Pennell PB, White JR, Birnbaum AK. Characterization of the time course of carbamazepine deinduction by an enzyme turnover model. Clin Pharmacokinet 2010; 48:313-20. [PMID: 19566114 DOI: 10.2165/00003088-200948050-00003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Carbamazepine is a potent inducer of drug metabolizing enzymes, which results in a number of clinically significant drug-drug interactions. Deinduction occurs when long-term carbamazepine therapy is discontinued. The goal of this study was to develop a population pharmacokinetic model to describe the time course of carbamazepine deinduction. PATIENTS AND METHODS Stable-labelled carbamazepine was administered intravenously on three occasions during the deinduction period to 15 patients with epilepsy for whom carbamazepine therapy was being discontinued. Data were analysed using a nonlinear mixed-effects model (NONMEM). An enzyme turnover model consisting of a one-compartment model linked with a hypothetical enzyme compartment was applied to characterize the time course of carbamazepine deinduction. Model evaluation was performed using the bootstrap approach and a visual predictive check. RESULTS In the final model, the deinduction process was accomplished by decreasing the rate of enzyme synthesis, resulting in a decrease in the relative amount of enzymes. The estimated rate constant for enzyme degradation was 0.00805 h-1, corresponding to a half-life of the combined enzymes of 86.1 hours (3.6 days). CONCLUSION An enzyme turnover model adequately characterized the experimental data. Based on the predicted enzyme half-life from the final model, the deinduction process should be completed within 2 weeks after carbamazepine therapy is terminated.
Collapse
Affiliation(s)
- Baralee Punyawudho
- Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | | | | | | | | | | | | | | |
Collapse
|