1
|
Karlsson IB, Ekdahl A, Etchingham-Coll H, Li XQ, Ericsson C, Ahlqvist M, Samuelsson K. Investigation of Biotransformation Pathways in a Chimeric Mouse with a Humanized Liver. Int J Mol Sci 2025; 26:1141. [PMID: 39940909 PMCID: PMC11818726 DOI: 10.3390/ijms26031141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Xenobiotics, including drugs, undergo metabolism to facilitate detoxification and excretion. Predicting a compound's metabolic fate before clinical trials is crucial for efficacy and safety. The existing methods rely on in vitro systems and in vivo animal testing. In vitro systems do not replicate the complexity of in vivo systems, and differences in biotransformation pathways between humans and nonclinical species may occur; thus, accurate predictions of human-specific drug metabolism are not always achieved. The aim of this study was to evaluate whether a chimeric mouse with a humanized liver, specifically the PXB-mouse, can mimic human metabolic profiles. PXB-mice have livers engrafted with up to 95% human hepatocytes. The biotransformation of 12 different small-molecule drugs were evaluated in PXB-mice (through analysis of blood and urine) and compared with the metabolism by hepatocytes from humans and mice and, when available, literature reports on human in vivo metabolism. The detected metabolites included major Phase I and II transitions, such as hydroxylation, and N- and O-dealkylation and glucuronidation. The metabolic patterns of the PXB-mice closely matched human in vivo data. It is also worth noting that the human hepatocytes formed most of the circulating metabolites, indicating that hepatocytes provide reliable predictions of human metabolic pathways. Thus, for drugs with human biotransformation pathways that are not observed in nonclinical species, the PXB-mouse model can be valuable in predicting human-specific metabolism.
Collapse
Affiliation(s)
- Isabella B. Karlsson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden; (A.E.); (H.E.-C.); (X.-Q.L.); (C.E.); (M.A.)
| | | | | | | | | | | | - Kristin Samuelsson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden; (A.E.); (H.E.-C.); (X.-Q.L.); (C.E.); (M.A.)
| |
Collapse
|
2
|
Tang LWT, Lapham K, Goosen TC. UGT2B10 is the Major UDP-Glucuronosyltransferase 2B Isoform Involved in the Metabolism of Lamotrigine and is Implicated in the Drug-Drug Interaction with Valproic Acid. AAPS J 2024; 26:107. [PMID: 39322784 DOI: 10.1208/s12248-024-00978-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/08/2024] [Indexed: 09/27/2024] Open
Abstract
Lamotrigine is a phenyltriazine anticonvulsant that is primarily metabolized by phase II UDP-glucuronosyltransferases (UGT) to a quaternary N2-glucuronide, which accounts for ~ 90% of the excreted dose in humans. While there is consensus that UGT1A4 plays a predominant role in the formation of the N2-glucuronide, there is compelling evidence in the literature to suggest that the metabolism of lamotrigine is catalyzed by another UGT isoform. However, the exact identity of the UGT isoform that contribute to the formation of this glucuronide remains uncertain. In this study, we harnessed a robust reaction phenotyping strategy to delineate the identities and its associated fraction metabolized (fm) of the UGTs involved in lamotrigine N2-glucuronidation. Foremost, human recombinant UGT mapping experiments revealed that the N2-glucuronide is catalyzed by multiple UGT isoforms. (i.e., UGT1A1, 1A3, 1A4, 1A9, 2B4, 2B7, and 2B10). Thereafter, scaling the apparent intrinsic clearances obtained from the enzyme kinetic experiments with our in-house liver-derived relative expression factors (REF) and relative activity factors (RAF) revealed that, in addition to UGT1A4, UGT2B10 was involved in the N2-glucuronidation of lamotrigine. This was further confirmed via chemical inhibition in human liver microsomes with the UGT1A4-selective inhibitor hecogenin and the UGT2B10-selective inhibitor desloratadine. By integrating various orthogonal approaches (i.e., REF- and RAF-scaling, and chemical inhibition), we quantitatively determined that the fm for UGT1A4 and UGT2B10 ranged from 0.42 - 0.64 and 0.32 - 0.57, respectively. Finally, we also provided nascent evidence that the pharmacokinetic interaction between lamotrigine and valproic acid likely arose from the in vivo inhibition of its UGT2B10-mediated pathway.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA.
| | - Kimberly Lapham
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA
| | - Theunis C Goosen
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Research and Development, Pfizer Inc., 445 Eastern Point Rd, Groton, CT, 06340, USA
| |
Collapse
|
3
|
Shanu-Wilson J, Coe S, Evans L, Steele J, Wrigley S. Small molecule drug metabolite synthesis and identification: why, when and how? Drug Discov Today 2024; 29:103943. [PMID: 38452922 DOI: 10.1016/j.drudis.2024.103943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
The drug discovery and development process encompasses the interrogation of metabolites arising from the biotransformation of drugs. Here we look at why, when and how metabolites of small-molecule drugs are synthesised from the perspective of a specialist contract research organisation, with particular attention paid to projects for which regulatory oversight is relevant during this journey. To illustrate important aspects, we look at recent case studies, trends and learnings from our experience of making and identifying metabolites over the past ten years, along with with selected examples from the literature.
Collapse
Affiliation(s)
- Julia Shanu-Wilson
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK.
| | - Samuel Coe
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Liam Evans
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Jonathan Steele
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Stephen Wrigley
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| |
Collapse
|
4
|
Kanamori T, Okada Y, Segawa H, Yamamuro T, Kuwayama K, Tsujikawa K, Iwata YT. Preparation of glucuronides using liver microsomes and their characterization by 1D/2D NMR spectroscopy and mass spectrometry: Application to fentanyl metabolites. Drug Test Anal 2024; 16:447-456. [PMID: 37583076 DOI: 10.1002/dta.3564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
A simple, low-cost method for preparing glucuronic acid-conjugated metabolites was developed using fentanyl, a potent synthetic opioid, as a model drug. Five glucuronic acid-conjugated metabolites of fentanyl were measured in the culture medium of fresh human hepatocytes incubated with fentanyl. These glucuronides were also formed by incubation of their corresponding substrates (e.g., 4'-hydroxy-fentanyl and β-hydroxy-fentanyl) with uridine 5'-diphosphoglucuronic acid and human liver microsomes (HLM). Experiments using liver microsomes of several animals revealed that significant species differences exist in the glucuronide formation patterns; fentanyl glucuronide was only formed in HLM, and 4'-hydroxy-fentanyl glucuronide was formed much more in rat liver microsomes (RLM) than HLM and dog liver microsomes. Furthermore, surprisingly, HLM and RLM showed opposite substrate selectivity for the enantiomers of β-hydroxy-fentanyl. Submilligram amounts of three of these metabolites, namely, 4'-hydroxy-fentanyl glucuronide and two glucuronides of β-hydroxy-fentanyl, were prepared by using HLM or RLM. The products were readily purified with a reversed-phase/anion-exchange mixed-mode solid-phase extraction cartridge, and then, their chemical structures were confirmed by 1D/2D nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry data. In addition, the products were quantitated by quantitative NMR, and the yields were 3.6-69%.
Collapse
Affiliation(s)
| | - Yuki Okada
- National Research Institute of Police Science, Kashiwa, Japan
| | - Hiroki Segawa
- National Research Institute of Police Science, Kashiwa, Japan
| | | | - Kenji Kuwayama
- National Research Institute of Police Science, Kashiwa, Japan
| | - Kenji Tsujikawa
- National Research Institute of Police Science, Kashiwa, Japan
| | - Yuko T Iwata
- National Research Institute of Police Science, Kashiwa, Japan
| |
Collapse
|
5
|
Thomann J, Kolaczynska KE, Stoeckmann OV, Rudin D, Vizeli P, Hoener MC, Pryce CR, Vollenweider FX, Liechti ME, Duthaler U. In vitro and in vivo metabolism of psilocybin's active metabolite psilocin. Front Pharmacol 2024; 15:1391689. [PMID: 38741590 PMCID: PMC11089204 DOI: 10.3389/fphar.2024.1391689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
In vivo, psilocybin is rapidly dephosphorylated to psilocin which induces psychedelic effects by interacting with the 5-HT2A receptor. Psilocin primarily undergoes glucuronidation or conversion to 4-hydroxyindole-3-acetic acid (4-HIAA). Herein, we investigated psilocybin's metabolic pathways in vitro and in vivo, conducting a thorough analysis of the enzymes involved. Metabolism studies were performed using human liver microsomes (HLM), cytochrome P450 (CYP) enzymes, monoamine oxidase (MAO), and UDP-glucuronosyltransferase (UGT). In vivo, metabolism was examined using male C57BL/6J mice and human plasma samples. Approximately 29% of psilocin was metabolized by HLM, while recombinant CYP2D6 and CYP3A4 enzymes metabolized nearly 100% and 40% of psilocin, respectively. Notably, 4-HIAA and 4-hydroxytryptophol (4-HTP) were detected with HLM but not with recombinant CYPs. MAO-A transformed psilocin into minimal amounts of 4-HIAA and 4-HTP. 4-HTP was only present in vitro. Neither 4-HIAA nor 4-HTP showed relevant interactions at assessed 5-HT receptors. In contrast to in vivo data, UGT1A10 did not extensively metabolize psilocin in vitro. Furthermore, two putative metabolites were observed. N-methyl-4-hydroxytryptamine (norpsilocin) was identified in vitro (CYP2D6) and in mice, while an oxidized metabolite was detected in vitro (CYP2D6) and in humans. However, the CYP2D6 genotype did not influence psilocin plasma concentrations in the investigated study population. In conclusion, MAO-A, CYP2D6, and CYP3A4 are involved in psilocin's metabolism. The discovery of putative norpsilocin in mice and oxidized psilocin in humans further unravels psilocin's metabolism. Despite limitations in replicating phase II metabolism in vitro, these findings hold significance for studying drug-drug interactions and advancing research on psilocybin as a therapeutic agent.
Collapse
Affiliation(s)
- Jan Thomann
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Karolina E. Kolaczynska
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Oliver V. Stoeckmann
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Patrick Vizeli
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Marius C. Hoener
- Neuroscience Research, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christopher R. Pryce
- Department of Psychiatry, Psychotherapy and Psychosomatics, Preclinical Laboratory for Translational Research Into Affective Disorders, University of Zurich, Zurich, Switzerland
| | - Franz X. Vollenweider
- Department of Psychiatry, Psychotherapy and Psychosomatics, Neurophenomenology and Consciousness, University of Zurich, Zurich, Switzerland
| | - Matthias E. Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Urs Duthaler
- Division of Clinical Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Institute of Forensic Medicine, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
- Institute of Forensic Medicine, Health Department Basel-Stadt, Basel, Switzerland
| |
Collapse
|
6
|
Papp R, Trimble L, Fretland AJ, Manohar R, Phipps R, Kvaerno L, Perryman AL, Reynolds G, Black WC. Identification and Biosynthesis of an N-Glucuronide Metabolite of Camonsertib. Drug Metab Dispos 2024; 52:DMD-AR-2023-001611. [PMID: 38378703 DOI: 10.1124/dmd.123.001611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024] Open
Abstract
Camonsertib is a novel ATR kinase inhibitor in clinical development for advanced cancers targeting sensitizing mutations. This article describes the identification and biosynthesis of an N-glucuronide metabolite of camonsertib. This metabolite was first observed in human hepatocyte incubations and was subsequently isolated to determine the structure, evaluate its stability as part of bioanalytical method development and for use as a standard for estimating its concentration in Phase I samples. The N-glucuronide was scaled-up using a purified bacterial culture preparation and was subsequently isolated using preparative chromatography. The bacterial culture generated sufficient material of the glucuronide to allow for one- and two-dimensional 1H and 13C NMR structural elucidation and further bioanalytical characterization. The NOE data combined with the gradient HMBC experiment and molecular modeling, strongly suggests that the point of attachment of the glucuronide results in the formation of (2S,3S,4S,5R,6R)-3,4,5-trihydroxy-6-(5-(4-((1R,3r,5S)-3-hydroxy-8-oxabicyclo[3.2.1]octan-3-yl)-6-((R)-3-methylmorpholino)-1H-pyrazolo[3,4-b]pyridin-1-yl)-1H-pyrazol-1-yl)tetrahydro-2H-pyran-2-carboxylic acid. Significance Statement This is the first report of a glucuronide metabolite of camonsertib formed by human hepatocyte incubations. This study reveals the structure of an N-glucuronide metabolite of camonsertib using detailed elucidation by one- and two-dimensional NMR after scale-up using a novel bacterial culture approach yielding significant quantities of a purified metabolite.
Collapse
Affiliation(s)
- Robert Papp
- Drug Metabolism and Pharmacokinetics, Repare Therapeutics, Inc., Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zhang C, Su D, Choo EF, Liu L, Bobba S, Jorski JD, Ho Q, Wang J, Kenny JR, Khojasteh SC, Zhang D. Identification of a Discrete Diglucuronide of GDC-0810 in Human Plasma after Oral Administration. Drug Metab Dispos 2023; 51:1284-1294. [PMID: 37349116 DOI: 10.1124/dmd.122.001071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
GDC-0810 is a small molecule therapeutic agent having potential to treat breast cancer. In plasma of the first-in-human study, metabolite M2, accounting for 20.7% of total drug-related materials, was identified as a discrete diglucuronide that was absent in rats. Acyl glucuronide M6 and N-glucuronide M4 were also identified as prominent metabolites in human plasma. Several in vitro studies were conducted in incubations of [14C]GDC-0810, synthetic M6 and M4 with liver microsomes, intestinal microsomes, and hepatocytes of different species as well as recombinant UDP-glucuronosyltransferase (UGT) enzymes to further understand the formation of M2. The results suggested that 1) M2 was more efficiently formed from M6 than from M4, and 2) acyl glucuronidation was mainly catalyzed by UGT1A8/7/1 that is highly expressed in the intestines whereas N-glucuronidation was mainly catalyzed by UGT1A4 that is expressed in the human liver. This complicated mechanism presented challenges in predicting M2 formation using human in vitro systems. The absence of M2 and M4 in rats can be explained by low to no expression of UGT1A4 in rodents. M2 could be the first discrete diglucuronide that was formed from both acyl- and N-glucuronidation on a molecule identified in human plasma. SIGNIFICANCE STATEMENT: A discrete diglucuronidation metabolite of GDC-0810, a breast cancer drug candidate, was characterized as a unique circulating metabolite in humans that was not observed in rats or little formed in human in vitro system.
Collapse
Affiliation(s)
- Chenghong Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Dian Su
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Edna F Choo
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Lichuan Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Sudheer Bobba
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Jamie D Jorski
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Quynh Ho
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Jing Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Jane R Kenny
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (E.F.C., S.B., J.D.J., J.W., J.R.K., S.C.K., D.Z.); Pfizer, South San Francisco, California (C.Z.); Bicycle Therapeutics, Cambridge, Massachusetts (D.S.); Innovative Research BU, Yifan Pharmaceutical, Hangzhou, China (L.L.); and Abbvie Biotherapeutics Inc., South San Francisco, California (Q.H.)
| |
Collapse
|
8
|
Ivanova L, Fæste CK, Solhaug A. Atlantic Salmon Gill Epithelial Cell Line (ASG-10) as a Suitable Model for Xenobiotic Biotransformation. Metabolites 2023; 13:771. [PMID: 37367928 PMCID: PMC10303838 DOI: 10.3390/metabo13060771] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
Fish are exposed to xenobiotics in the water. Uptake occurs mainly through the gills, which function as an exchange point with the environment. The gills' ability to detoxify harmful compounds by biotransformation is an essential protection mechanism. The enormous numbers of waterborne xenobiotics requiring ecotoxicological assessment makes it necessary to replace in vivo fish studies with predictive in vitro models. Here, we have characterized the metabolic capacity of the ASG-10 gill epithelial cell line from Atlantic salmon. Inducible CYP1A expression was confirmed by enzymatic assays and immunoblotting. The activities of important cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT) enzymes were established using specific substrates and metabolite analysis by liquid chromatography (LC) triple quadrupole mass spectrometry (TQMS). Metabolism of the fish anesthetic benzocaine (BZ) in ASG-10 confirmed esterase and acetyl transferase activities through the production of N-acetylbenzocaine (AcBZ), p-aminobenzoic acid (PABA) and p-acetaminobenzoic acid (AcPABA). Moreover, we were able to determine hydroxylamine benzocaine (BZOH), benzocaine glucuronide (BZGlcA) and hydroxylamine benzocaine glucuronide (BZ(O)GlcA) by LC high-resolution tandem mass spectrometry (HRMS/MS) fragment pattern analysis for the first time. Comparison to metabolite profiles in hepatic fractions, and in plasma of BZ-euthanized salmon, confirmed the suitability of the ASG-10 cell line for investigating biotransformation in gills.
Collapse
Affiliation(s)
- Lada Ivanova
- Chemistry and Toxinology Research Group, Norwegian Veterinary Institute, 1433 Ås, Norway; (C.K.F.); (A.S.)
| | | | | |
Collapse
|
9
|
Lv X, Wang Z, Wang Z, Yin H, Xia Y, Jiang L, Liu Y. Inhibition of human UDP-glucuronosyltransferase enzyme by ripretinib: Implications for drug-drug interactions. Toxicol Appl Pharmacol 2023; 466:116490. [PMID: 36963523 DOI: 10.1016/j.taap.2023.116490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
Ripretinib, a tyrosine kinase inhibitor (TKI), is the first FDA approved fourth-line therapy for adults with advanced gastrointestinal stromal tumor (GIST). Studies have shown that several TKIs for treating GIST were potent inhibitors of human UDP- glucosyltransferase (UGTs) enzymes. However, whether ripretinib affects the activity of UGTs remains unclear. The aim of this study was to investigate the effects of ripretinib on major UGT isoforms, as well as to evaluate its potential drug-drug interactions (DDIs) risk caused by the inhibition of UGTs activities. The inhibitory effects and inhibition modes of ripretinib on UGTs were systematically evaluated using high-performance liquid chromatography (HPLC) and enzyme kinetic studies, respectively. Our data showed that ripretinib exhibited potent inhibition against UGT1A1, UGT1A3, UGT1A4, UGT1A7 and UGT1A8. Enzyme kinetic studies indicated that ripretinib was not only a competitive inhibitor of UGT1A1, UGT1A4 and UGT1A7, but also a noncompetitive inhibitor of UGT1A3, as well as a mixed inhibitor of UGT1A8. The prediction results of in vitro-in vivo extrapolation (IVIVE) demonstrated that ripretinib might bring the potential risk of DDIs when combined with substrates of UGT1A1, UGT1A3, UGT1A4, UGT1A7 or UGT1A8. Therefore, special attention should be paid when ripretinib is used in conjunction with other drugs metabolized by UGTs to avoid risk of DDIs in clinic.
Collapse
Affiliation(s)
- Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China; Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
10
|
Gadgoli UB, Sunil Kumar YC, Kumar D. An Insight into the Metabolism of 2,5-Disubstituted Monotetrazole Bearing Bisphenol Structures: Emerging Bisphenol A Structural Congeners. Molecules 2023; 28:molecules28031465. [PMID: 36771130 PMCID: PMC9921896 DOI: 10.3390/molecules28031465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
The non-estrogenic 2,5-disubstituted tetrazole core-bearing bisphenol structures (TbB) are being researched as emerging structural congeners of Bisphenol A, an established industrial endocrine disruptor. However, there is no understanding of TbB's adverse effects elicited via metabolic activation. Therefore, the current study aimed to investigate the metabolism of TbB ligands, with in silico results serving as a guide for in vitro studies. The Cytochrome P450 enzymes (CYP) inhibitory assay of TbB ligands on the seven human liver CYP isoforms (i.e., 1A2, 2A6, 2D6, 2C9, 2C8, 2C19, and 3A4) using human liver microsomes (HLM) revealed TbB ligand 223-3 to have a 50% inhibitory effect on all the CYP isoforms at a 10 μM concentration, except 1A2. The TbB ligand 223-10 inhibited 2B6 and 2C8, whereas the TbB ligand 223-2 inhibited only 2C9. The first-order inactivity rate constant (Kobs) studies indicated TbB ligands 223-3, 223-10 to be time-dependent (TD) inhibitors, whereas the TbB 223-2 ligand did not show such a significant effect. The 223-3 exhibited a TD inhibition for 2C9, 2C19, and 1A2 with Kobs values of 0.0748, 0.0306, and 0.0333 min-1, respectively. On the other hand, the TbB ligand 223-10 inhibited 2C9 in a TD inhibition manner with Kobs value 0.0748 min-1. However, the TbB ligand 223-2 showed no significant TD inhibition effect on the CYPs. The 223-2 ligand biotransformation pathway by in vitro studies in cryopreserved human hepatocytes suggested the clearance via glucuronidation with the predominant detection of only 223-2 derived mono glucuronide as a potential inactive metabolite. The present study demonstrated that the 223-2 ligand did not elicit any significant adverse effect via metabolic activation, thus paving the way for its in vivo drug-drug interactions (DDI) studies.
Collapse
Affiliation(s)
- Umesh B. Gadgoli
- Department of Chemistry, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
- Correspondence:
| | - Yelekere C. Sunil Kumar
- Dayanada Sagar Academy of Technology and Management, Kanakapura Rd, Opp. Art of Living International Centre, Udaypura, Bengaluru 560082, Karnataka, India
| | - Deepak Kumar
- Department of Chemistry, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| |
Collapse
|
11
|
Identification of Human UDP-Glucuronosyltransferase Involved in Gypensapogenin C Glucuronidation and Species Differences. Int J Mol Sci 2023; 24:ijms24021454. [PMID: 36674970 PMCID: PMC9865363 DOI: 10.3390/ijms24021454] [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: 12/03/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Gypensapogenin C (GPC) is one of the important aglycones of Gynostemma pentaphyllum (GP), which is structurally glucuronidated and is highly likely to bind to UGT enzymes in vivo. Due to the important role of glucuronidation in the metabolism of GPC, the UDP-glucuronosyltransferase metabolic pathway of GPC in human and other species' liver microsomes is investigated in this study. In the present study, metabolites were detected using high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). The results show that GPC could generate a metabolite through glucuronidation in the human liver microsomes (HLMs). Additionally, chemical inhibitors combined with recombinant human UGT enzymes clarified that UGT1A4 is the primary metabolic enzyme for GPC glucuronidation in HLMs according to the kinetic analysis of the enzyme. Metabolic differential analysis in seven other species indicated that rats exhibited the most similar metabolic rate to that of humans. In conclusion, UGT1A4 is a major enzyme responsible for the glucuronidation of GPC in HLMs, and rats may be an appropriate animal model to evaluate the GPC metabolism.
Collapse
|
12
|
He YF, Liu Y, Yu JH, Cheng H, Odilov A, Yang FP, Tian GH, Yao XM, Duan HQ, Yu CY, Yu C, Liu YM, Liu GY, Shen JS, Wang Z, Diao XX. Pharmacokinetics, mass balance, and metabolism of [ 14C]TPN171, a novel PDE5 inhibitor, in humans for the treatment of pulmonary arterial hypertension. Acta Pharmacol Sin 2023; 44:221-233. [PMID: 35676531 DOI: 10.1038/s41401-022-00922-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/11/2022] [Indexed: 01/18/2023]
Abstract
TPN171 is a novel phosphodiesterase-5 (PDE5) inhibitor used to treat pulmonary arterial hypertension (PAH) and erectile dysfunction (ED), which currently is undergoing phase II clinical trials in China. In this single-center, single-dose, nonrandomized, and open design study, radiolabeled [14C]TPN171 was used to investigate the metabolic mechanism, pharmacokinetic characteristics, and clearance pathways of TPN171 in 6 healthy Chinese male volunteers. Each volunteer was administered a single oral suspension of 10 mg (100 μCi) of [14C]TPN171. We found that TPN171 was absorbed rapidly in humans with a peak time (Tmax) of 0.667 h and a half-life (t1/2) of approximately 9.89 h in plasma. Excretion of radiopharmaceutical-related components was collected 216 h after administration, accounting for 95.21% of the dose (46.61% in urine and 48.60% in feces). TPN171 underwent extensive metabolism in humans. Twenty-two metabolites were detected in human plasma, urine, and feces using a radioactive detector combined with a high-resolution mass spectrometer. According to radiochromatograms, a glucuronide metabolite of O-dealkylated TPN171 exceeded 10% of the total drug-related components in human plasma. However, according to the Food and Drug Administration (FDA) guidelines, no further tests are needed to evaluate the safety of this metabolite because it is a phase II metabolite, but the compound is still worthy of attention. The main metabolic biotransformation of TPN171 was mono-oxidation (hydroxylation and N-oxidation), dehydrogenation, N-dealkylation, O-dealkylation, amide hydrolysis, glucuronidation, and acetylation. Cytochrome P450 3A4 (CYP3A4) mainly catalyzed the formation of metabolites, and CYP2E1 and CYP2D6 were involved in the oxidative metabolism of TPN171 to a lesser extent. According to the incubation data, M1 was mainly metabolized to M1G by UDP-glucuronosyltransferase 1A9 (UGT1A9), followed by UGT1A7 and UGT1A10.
Collapse
Affiliation(s)
- Yi-Fei He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yin Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing-Hua Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huan Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Ji-nan, 250355, China
| | - Abdullajon Odilov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei-Pu Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | | - Xiu-Mei Yao
- Vigonvita Life Sciences Co., Ltd, Suzhou, 215000, China
| | - Hua-Qing Duan
- Vigonvita Life Sciences Co., Ltd, Suzhou, 215000, China
| | - Cheng-Yin Yu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Chen Yu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Yan-Mei Liu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Gang-Yi Liu
- Shanghai Xuhui Central Hospital, Shanghai, 200030, China
| | - Jing-Shan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,Lingang Laboratory, Shanghai, 201602, China.
| | - Xing-Xing Diao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Machine learning and structure-based modeling for the prediction of UDP-glucuronosyltransferase inhibition. iScience 2022; 25:105290. [PMID: 36304105 PMCID: PMC9593791 DOI: 10.1016/j.isci.2022.105290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/05/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are responsible for 35% of the phase II drug metabolism. In this study, we focused on UGT1A1, which is a key UGT isoform. Strong inhibition of UGT1A1 may trigger adverse drug/herb-drug interactions, or result in disorders of endobiotic metabolism. Most of the current machine learning methods predicting the inhibition of drug metabolizing enzymes neglect protein structure and dynamics, both being essential for the recognition of various substrates and inhibitors. We performed molecular dynamics simulations on a homology model of the human UGT1A1 structure containing both the cofactor- (UDP-glucuronic acid) and substrate-binding domains to explore UGT conformational changes. Then, we created models for the prediction of UGT1A1 inhibitors by integrating information on UGT1A1 structure and dynamics, interactions with diverse ligands, and machine learning. These models can be helpful for further prediction of drug-drug interactions of drug candidates and safety treatments. UGTs are responsible for 35% of the phase II drug metabolism reactions We created machine learning models for prediction of UGT1A1 inhibitors Our simulations suggested key residues of UGT1A1 involved in the substrate binding
Collapse
|
14
|
Gorrochategui-Ortega J, Muñoz-Colmenero M, Kovačić M, Filipi J, Puškadija Z, Kezić N, Parejo M, Büchler R, Estonba A, Zarraonaindia I. A short exposure to a semi-natural habitat alleviates the honey bee hive microbial imbalance caused by agricultural stress. Sci Rep 2022; 12:18832. [PMID: 36336704 PMCID: PMC9637708 DOI: 10.1038/s41598-022-23287-6] [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: 01/18/2022] [Accepted: 10/28/2022] [Indexed: 11/08/2022] Open
Abstract
Honeybee health and the species' gut microbiota are interconnected. Also noteworthy are the multiple niches present within hives, each with distinct microbiotas and all coexisting, which we termed "apibiome". External stressors (e.g. anthropization) can compromise microbial balance and bee resilience. We hypothesised that (1) the bacterial communities of hives located in areas with different degrees of anthropization differ in composition, and (2) due to interactions between the multiple microbiomes within the apibiome, changes in the community of a niche would impact the bacteria present in other hive sections. We characterised the bacterial consortia of different niches (bee gut, bee bread, hive entrance and internal hive air) of 43 hives from 3 different environments (agricultural, semi-natural and natural) through 16S rRNA amplicon sequencing. Agricultural samples presented lower community evenness, depletion of beneficial bacteria, and increased recruitment of stress related pathways (predicted via PICRUSt2). The taxonomic and functional composition of gut and hive entrance followed an environmental gradient. Arsenophonus emerged as a possible indicator of anthropization, gradually decreasing in abundance from agriculture to the natural environment in multiple niches. Importantly, after 16 days of exposure to a semi-natural landscape hives showed intermediate profiles, suggesting alleviation of microbial dysbiosis through reduction of anthropization.
Collapse
Affiliation(s)
- June Gorrochategui-Ortega
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Marta Muñoz-Colmenero
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain ,grid.419099.c0000 0001 1945 7711Instituto de Investigaciones Marinas (CSIC)/Institute of Marine Research, Eduardo Cabello 6, 36208 Vigo, Pontevedra Spain
| | - Marin Kovačić
- grid.412680.90000 0001 1015 399XFaculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek, V.Preloga 1, 31000 Osijek, Croatia
| | - Janja Filipi
- grid.424739.f0000 0001 2159 1688Department of Ecology, Agronomy and Aquaculture, University of Zadar, Trg Kneza Višeslava 9, 23000 Zadar, Croatia
| | - Zlatko Puškadija
- grid.412680.90000 0001 1015 399XFaculty of Agrobiotechnical Sciences Osijek, Josip Juraj Strossmayer University of Osijek, V.Preloga 1, 31000 Osijek, Croatia
| | - Nikola Kezić
- grid.4808.40000 0001 0657 4636Department of Fisheries, Apiculture and Special Zoology, Faculty of Agriculture, University of Zagreb, Svetošimunska Cesta 25, 10000 Zagreb, Croatia
| | - Melanie Parejo
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Ralph Büchler
- grid.506460.10000 0004 4679 6788Landesbetrieb Landwirtschaft Hessen (LLH), Bieneninstitut, Erlenstraße 9, 35274 Kirchhain, Germany
| | - Andone Estonba
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Iratxe Zarraonaindia
- grid.11480.3c0000000121671098Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain ,grid.424810.b0000 0004 0467 2314IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
15
|
Validated single urinary assay designed for exposomic multi-class biomarkers of common environmental exposures. Anal Bioanal Chem 2022; 414:5943-5966. [PMID: 35754089 PMCID: PMC9326253 DOI: 10.1007/s00216-022-04159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 11/01/2022]
Abstract
Epidemiological studies often call for analytical methods that use a small biospecimen volume to quantify trace level exposures to environmental chemical mixtures. Currently, as many as 150 polar metabolites of environmental chemicals have been found in urine. Therefore, we developed a multi-class method for quantitation of biomarkers in urine. A single sample preparation followed by three LC injections was optimized in a proof-of-approach for a multi-class method. The assay was validated to quantify 50 biomarkers of exposure in urine, belonging to 7 chemical classes and 16 sub-classes. The classes represent metabolites of 12 personal care and consumer product chemicals (PCPs), 5 polycyclic aromatic hydrocarbons (PAHs), 5 organophosphate flame retardants (OPFRs), 18 pesticides, 5 volatile organic compounds (VOCs), 4 tobacco alkaloids, and 1 drug of abuse. Human urine (0.2 mL) was spiked with isotope-labeled internal standards, enzymatically deconjugated, extracted by solid-phase extraction, and analyzed using high-performance liquid chromatography-tandem mass spectrometry. The methanol eluate from the cleanup was split in half and the first half analyzed for PCPs, PAH, and OPFR on a Betasil C18 column; and pesticides and VOC on a Hypersil Gold AQ column. The second half was analyzed for tobacco smoke metabolites and a drug of abuse on a Synergi Polar RP column. Limits of detection ranged from 0.01 to 1.0 ng/mL of urine, with the majority ≤0.5 ng/mL (42/50). Analytical precision, estimated as relative standard deviation of intra- and inter-batch uncertainty, variabilities, was <20%. Extraction recoveries ranged from 83 to 109%. Results from the optimized multi-class method were qualified in formal international proficiency testing programs. Further method customization options were explored and method expansion was demonstrated by inclusion of up to 101 analytes of endo- and exogenous chemicals. This exposome-scale assay is being used for population studies with savings of assay costs and biospecimens, providing both quantitative results and the discovery of unexpected exposures.
Collapse
|
16
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
17
|
Janssen W, Schwarz T, Bütehorn U, Steinke W, Sandmann S, Lang D, Kern A, Hucke F, Gerisch M. Pharmacokinetics and mass balance of vericiguat in rats and dogs and distribution in rats. Xenobiotica 2022; 52:453-462. [PMID: 35616579 DOI: 10.1080/00498254.2022.2082899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Vericiguat is a soluble guanylate cyclase stimulator. The pharmacokinetics, absorption, metabolism, and excretion properties of vericiguat in rats and dogs and the distribution in rats are reported. [14C]-labelled vericiguat was studied in intact and bile duct-cannulated rats (oral and intravenous administration), and dogs (oral administration).Vericiguat reached maximum plasma concentrations at 1-3 h after oral administration. Absolute bioavailability was moderate in rats and high in dogs. Vericiguat was the most abundant component in plasma of rats and dogs.After oral administration to rats, radioactivity was widely distributed. Penetration into the brain was minimal. Elimination was rapid from most tissues in rats. Most of the radioactivity was excreted in faeces (rat: 81%, dog: 89%), while low amounts were excreted in urine (rat: 11%, dog: 4%). Clearance routes in both species were unchanged excretion and metabolism via glucuronidation and oxidative reactions. After intravenous administration to bile duct cannulated rats, a relevant proportion of the dose (30%) underwent direct excretion into the gastrointestinal tract as unchanged vericiguat.
Collapse
Affiliation(s)
- Wiebke Janssen
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | - Thomas Schwarz
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | - Ulf Bütehorn
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | - Wolfram Steinke
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany.,Pharmacokinetic Imaging Consulting & Autoradiography Services, 58553 Halver, Germany
| | | | - Dieter Lang
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | - Armin Kern
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | - Frank Hucke
- Bayer Aktiengesellschaft, DMPK, 42096 Wuppertal, Germany
| | | |
Collapse
|
18
|
Hu DG, Mackenzie PI, Hulin JA, McKinnon RA, Meech R. Regulation of human UDP-glycosyltransferase ( UGT) genes by miRNAs. Drug Metab Rev 2022; 54:120-140. [PMID: 35275773 DOI: 10.1080/03602532.2022.2048846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The human UGT gene superfamily is divided into four subfamilies (UGT1, UGT2, UGT3 and UGT8) that encodes 22 functional enzymes. UGTs are critical for the metabolism and clearance of numerous endogenous and exogenous compounds, including steroid hormones, bile acids, bilirubin, fatty acids, carcinogens, and therapeutic drugs. Therefore, the expression and activities of UGTs are tightly regulated by multiple processes at the transcriptional, post-transcriptional and post-translational levels. During recent years, nearly twenty studies have investigated the post-transcriptional regulation of UGT genes by miRNAs using human cancer cell lines (predominantly liver cancer). Overall, 14 of the 22 UGT mRNAs (1A1, 1A3, 1A4, 1A6, 1A8, 1A9, 1A10, 2A1, 2B4, 2B7, 2B10, 2B15, 2B17, UGT8) have been shown to be regulated by various miRNAs through binding to their respective 3' untranslated regions (3'UTRs). Three 3'UTRs (UGT1A, UGT2B7 and UGT2B15) contain the largest number of functional miRNA target sites; in particular, the UGT1A 3'UTR contains binding sites for 12 miRNAs (548d-5p, 183-5p, 214-5p, 486-3p, 200a-3p, 491-3p, 141-3p, 298, 103b, 376b-3p, 21-3p, 1286). Although all nine UGT1A family members have the same 3'UTR, these miRNA target sites appear to be functional in an isoform-specific and cellular context-dependent manner. Collectively, these observations demonstrate that miRNAs represent important post-transcriptional regulators of the UGT gene superfamily. In this article, we present a comprehensive review of reported UGT/miRNA regulation studies, describe polymorphisms within functional miRNA target sites that may affect their functionalities, and discuss potential cooperative and competitive regulation of UGT mRNAs by miRNAs through adjacently located miRNA target sites.
Collapse
Affiliation(s)
- Dong Gui Hu
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Peter I Mackenzie
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Ross A McKinnon
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| |
Collapse
|
19
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Zhang G, Xiao L, Qi L. Metabolite Profiling of Meridianin C In Vivo of Rat by UHPLC/Q-TOF MS. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2021; 2021:1382421. [PMID: 34721922 PMCID: PMC8553504 DOI: 10.1155/2021/1382421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/23/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Meridianin C (MC), as a marine alkaloid, is a potent protein kinase inhibitor which exhibits good anticancer activity. However, the in vivo metabolism of MC has not been described to date. In this study, an ultra-high performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UHPLC/Q-TOF MS) method is employed to investigate the in vivo metabolites of MC in rats. Plasma, bile, urine, and feces are collected after a single oral dose of MC. Protein precipitation, solid phase extraction (SPE), and ultrasonic extraction methods are used to prepare samples. Based on the mass spectral fragmentation patterns, elution order, and retrieving literatures, a total of 13 metabolites of MC were detected and tentatively identified, utilizing MetaboLynx software. The metabolic pathways of MC in rats include N- or O-glucuronidation, O-sulfation, N-hydroxylation, dihydroxylation, and trihydroxylation. The relative content of the metabolites in each kinds of biological samples is also evaluated. This study will help to understand the in vivo properties of MC for the future usage.
Collapse
Affiliation(s)
- Guozhe Zhang
- Department of Translational Medicine, Jiangsu Vocational College of Medicine, 283 South of Republic Road, Yancheng 224005, China
| | - Linxia Xiao
- Department of Translational Medicine, Jiangsu Vocational College of Medicine, 283 South of Republic Road, Yancheng 224005, China
| | - Liang Qi
- Department of Translational Medicine, Jiangsu Vocational College of Medicine, 283 South of Republic Road, Yancheng 224005, China
| |
Collapse
|
21
|
Taavitsainen P, Prien O, Kähkönen M, Niehues M, Korjamo T, Denner K, Nykänen P, Vuorela A, Jungmann NA, von Bühler CJ, Koskinen M, Zurth C, Gieschen H. Metabolism and Mass Balance of the Novel Nonsteroidal Androgen Receptor Inhibitor Darolutamide in Humans. Drug Metab Dispos 2021; 49:420-433. [PMID: 33785516 DOI: 10.1124/dmd.120.000309] [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: 11/10/2020] [Accepted: 03/10/2021] [Indexed: 12/28/2022] Open
Abstract
The biotransformation and excretion of darolutamide were investigated in a phase I study. Six healthy male volunteers received a single dose of 300 mg 14C-darolutamide as an oral solution in the fasted state. Plasma, urine, and feces samples were analyzed for mass balance evaluation by liquid scintillation counting (LSC). Metabolite profiling and identification were determined using liquid chromatography mass-spectrometry with off-line radioactivity detection using LSC. Complete mass balance was achieved, with mean radioactivity recovery of 95.9% within 168 hours (63.4% in urine, 32.4% in feces). The administered 1:1 ratio of (S,R)- and (S,S)-darolutamide changed to approximately 1:5, respectively, in plasma. Darolutamide and the oxidation product, keto-darolutamide, were the only components quantifiable by LSC in plasma, accounting for 87.4% of total radioactivity, with a 2.1-fold higher plasma exposure for keto-darolutamide. Aside from darolutamide, the most prominent metabolites in urine were O-glucoronide (M-7a/b) and N-glucuronide (M-15a/b), as well as pyrazole sulfates (M-29, M-24) and glucuronides (M-21, M-22) resulting from oxidative cleavage of the parent. The darolutamide diastereomers were mainly detected in feces. In vitro assays showed that darolutamide metabolism involves a complex interplay between oxidation and reduction, as well as glucuronidation. Interconversion of the diastereomers involves oxidation to keto-darolutamide, primarily mediated by CYP3A4, followed by reduction predominantly catalyzed by cytosolic reductase(s), with aldo-keto reductase 1C3 playing the major role. The latter reaction showed stereoselectivity with preferential formation of (S,S)-darolutamide. SIGNIFICANCE STATEMENT: The metabolism and excretion of darolutamide in humans revealed that oxidation (CYP3A4) and glucuronidation (UGT1A9, UGT1A1) were the main metabolic routes of elimination. Direct excretion also contributed to overall clearance. The two pharmacologically equipotent diastereomers of darolutamide interconvert primarily via oxidation to the active metabolite keto-darolutamide, followed by reduction predominantly by cytosolic reductase(s). The latter reaction showed stereoselectivity with preferential formation of (S,S)-darolutamide. Data indicate a low drug-drug interaction potential of darolutamide with inducers or inhibitors of metabolizing enzymes.
Collapse
Affiliation(s)
- Päivi Taavitsainen
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Olaf Prien
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Marja Kähkönen
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Michael Niehues
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Timo Korjamo
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Karsten Denner
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Pirjo Nykänen
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Annamari Vuorela
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Natalia A Jungmann
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Clemens-Jeremias von Bühler
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Mikko Koskinen
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Christian Zurth
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| | - Hille Gieschen
- Orion Corporation Orion Pharma, Turku, Finland (P.T.); Bayer AG, Berlin, Germany (O.P., M.N., K.D., C.Z., H.G.); Orion Corporation Orion Pharma, Espoo, Finland (M.K., T.K., P.N., A.V., M.K.); and Bayer AG, Wuppertal, Germany (N.A.J., C.-J.v.B.)
| |
Collapse
|
22
|
Zhou QH, Qin WW, Finel M, He QQ, Tu DZ, Wang CR, Ge GB. A broad-spectrum substrate for the human UDP-glucuronosyltransferases and its use for investigating glucuronidation inhibitors. Int J Biol Macromol 2021; 180:252-261. [PMID: 33741369 DOI: 10.1016/j.ijbiomac.2021.03.073] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 02/07/2023]
Abstract
Strong inhibition of the human UDP-glucuronosyltransferase enzymes (UGTs) may lead to undesirable effects, including hyperbilirubinaemia and drug/herb-drug interactions. Currently, there is no good way to examine the inhibitory effects and specificities of compounds toward all the important human UGTs, side-by-side and under identical conditions. Herein, we report a new, broad-spectrum substrate for human UGTs and its uses in screening and characterizing of UGT inhibitors. Following screening a variety of phenolic compound(s), we have found that methylophiopogonanone A (MOA) can be readily O-glucuronidated by all tested human UGTs, including the typical N-glucuronidating enzymes UGT1A4 and UGT2B10. MOA-O-glucuronidation yielded a single mono-O-glucuronide that was biosynthesized and purified for structural characterization and for constructing an LC-UV based MOA-O-glucuronidation activity assay, which was then used for investigating MOA-O-glucuronidation kinetics in recombinant human UGTs. The derived Km values were crucial for selecting the most suitable assay conditions for assessing inhibitory potentials and specificity of test compound(s). Furthermore, the inhibitory effects and specificities of four known UGT inhibitors were reinvestigated by using MOA as the substrate for all tested UGTs. Collectively, MOA is a broad-spectrum substrate for the human UGTs, which offers a new and practical tool for assessing inhibitory effects and specificities of UGT inhibitors.
Collapse
Affiliation(s)
- Qi-Hang Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Wei Qin
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014, Finland
| | - Qing-Qing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dong-Zhu Tu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chao-Ran Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
23
|
Huang K, Que L, Ding Y, Chu N, Qian Z, Qin W, Chen Y, Zhang J, He Q. Identification of human uridine diphosphate-glucuronosyltransferase isoforms responsible for the glucuronidation of 10,11-dihydro-10-hydroxy-carbazepine. J Pharm Pharmacol 2021; 73:388-397. [PMID: 33793880 DOI: 10.1093/jpp/rgaa059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To determine the kinetics of the formation of 10,11-dihydro-10-hydroxy-carbazepine (MHD)-O-glucuronide in human liver microsomes (HLMs), human intestine microsomes (HIMs), human kidney microsomes (HKMs) and recombinant human UDP-glucuronosyltransferase (UGTs), and identify the primary UGT isoforms catalyzing the glucuronidation of MHD. METHODS The kinetics of the glucuronidation of MHD was determined in HLMs, HIMs as well as HKMs. Screening assays with 13 recombinant human UGTs, inhibition studies and correlation analysis were performed to identify the main UGTs involved in the glucuronidation of MHD. KEY FINDINGS MHD-O-glucuronide was formed in HLMs, HIMs as well as HKMs, HLMs showed the highest intrinsic clearance of MHD. Among 13 recombinant human UGTs, UGT2B7 and UGT1A9 were identified to be the principal UGT isoforms mediating the glucuronidation of MHD, while UGT1A4 played a partial role. In addition, inhibition studies and correlation analysis further confirmed that UGT2B7 and UGT1A9 participated in the formation of MHD-O-glucuronide. CONCLUSIONS MHD could be metabolized by UGTs in the liver, intestine and kidney, and the hepatic glucuronidation was the critical metabolic pathway. UGT2B7 and UGT1A9 were the primary UGT isoforms mediating the formation of MHD-O-glucuronide in the liver.
Collapse
Affiliation(s)
- Kai Huang
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Linling Que
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Ying Ding
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Nannan Chu
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhenzhong Qian
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Wei Qin
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yuanxing Chen
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jisheng Zhang
- Drug Clinical Trial Institution, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | | |
Collapse
|
24
|
Perez-Paramo YX, Lazarus P. Pharmacogenetics factors influencing smoking cessation success; the importance of nicotine metabolism. Expert Opin Drug Metab Toxicol 2021; 17:333-349. [PMID: 33322962 PMCID: PMC8049967 DOI: 10.1080/17425255.2021.1863948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/10/2020] [Indexed: 01/12/2023]
Abstract
Introduction: Smoking remains a worldwide epidemic, and despite an increase in public acceptance of the harms of tobacco use, it remains the leading cause of preventable death. It is estimated that up to 70% of all smokers express a desire to quit, but only 3-5% of them are successful.Areas covered: The goal of this review was to evaluate the current status of smoking cessation treatments and the feasibility of implementing personalized-medicine approaches to these pharmacotherapies. We evaluated the genetics associated with higher levels of nicotine addiction and follow with an analysis of the genetic variants that affect the nicotine metabolic ratio (NMR) and the FDA approved treatments for smoking cessation. We also highlighted the gaps in the process of translating current laboratory understanding into clinical practice, and the benefits of personalized treatment approaches for a successful smoking cessation strategy.Expert opinion: Evidence supports the use of tailored therapies to ensure that the most efficient treatments are utilized in an individual's smoking cessation efforts. An understanding of the genetic effects on the efficacy of individualized smoking cessation pharmacotherapies is key to smoking cessation, ideally utilizing a polygenetic risk score that considers all genetic variation.
Collapse
Affiliation(s)
- Yadira X. Perez-Paramo
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| |
Collapse
|
25
|
Raju B, Choudhary S, Narendra G, Verma H, Silakari O. Molecular modeling approaches to address drug-metabolizing enzymes (DMEs) mediated chemoresistance: a review. Drug Metab Rev 2021; 53:45-75. [PMID: 33535824 DOI: 10.1080/03602532.2021.1874406] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Resistance against clinically approved anticancer drugs is the main roadblock in cancer treatment. Drug metabolizing enzymes (DMEs) that are capable of metabolizing a variety of xenobiotic get overexpressed in malignant cells, therefore, catalyzing drug inactivation. As evident from the literature reports, the levels of DMEs increase in cancer cells that ultimately lead to drug inactivation followed by drug resistance. To puzzle out this issue, several strategies inclusive of analog designing, prodrug designing, and inhibitor designing have been forged. On that front, the implementation of computational tools can be considered a fascinating approach to address the problem of chemoresistance. Various research groups have adopted different molecular modeling tools for the investigation of DMEs mediated toxicity problems. However, the utilization of these in-silico tools in maneuvering the DME mediated chemoresistance is least considered and yet to be explored. These tools can be employed in the designing of such chemotherapeutic agents that are devoid of the resistance problem. The current review canvasses various molecular modeling approaches that can be implemented to address this issue. Special focus was laid on the development of specific inhibitors of DMEs. Additionally, the strategies to bypass the DMEs mediated drug metabolism were also contemplated in this report that includes analogs and pro-drugs designing. Different strategies discussed in the review will be beneficial in designing novel chemotherapeutic agents that depreciate the resistance problem.
Collapse
Affiliation(s)
- Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
26
|
Israni B, Wouters FC, Luck K, Seibel E, Ahn SJ, Paetz C, Reinert M, Vogel H, Erb M, Heckel DG, Gershenzon J, Vassão DG. The Fall Armyworm Spodoptera frugiperda Utilizes Specific UDP-Glycosyltransferases to Inactivate Maize Defensive Benzoxazinoids. Front Physiol 2020; 11:604754. [PMID: 33408643 PMCID: PMC7781194 DOI: 10.3389/fphys.2020.604754] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
The relationship between plants and insects is continuously evolving, and many insects rely on biochemical strategies to mitigate the effects of toxic chemicals in their food plants, allowing them to feed on well-defended plants. Spodoptera frugiperda, the fall armyworm (FAW), accepts a number of plants as hosts, and has particular success on plants of the Poaceae family such as maize, despite their benzoxazinoid (BXD) defenses. BXDs stored as inert glucosides are converted into toxic aglucones by plant glucosidases upon herbivory. DIMBOA, the main BXD aglucone released by maize leaves, can be stereoselectively re-glucosylated by UDP-glycosyltransferases (UGTs) in the insect gut, rendering it non-toxic. Here, we identify UGTs involved in BXD detoxification by FAW larvae and examine how RNAi-mediated manipulation of the larval glucosylation capacity toward the major maize BXD, DIMBOA, affects larval growth. Our findings highlight the involvement of members of two major UGT families, UGT33 and UGT40, in the glycosylation of BXDs. Most of the BXD excretion in the frass occurs in the form of glucosylated products. Furthermore, the DIMBOA-associated activity was enriched in the gut tissue, with a single conserved UGT33 enzyme (SfUGT33F28) being dedicated to DIMBOA re-glucosylation in the FAW gut. The knock-down of its encoding gene reduces larval performance in a strain-specific manner. This study thus reveals that a single UGT enzyme is responsible for detoxification of the major maize-defensive BXD in this pest insect.
Collapse
Affiliation(s)
- Bhawana Israni
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Felipe C Wouters
- Max Planck Institute for Chemical Ecology, Jena, Germany.,Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Katrin Luck
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Elena Seibel
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Seung-Joon Ahn
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Mississippi State, MS, United States
| | | | | | - Heiko Vogel
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Matthias Erb
- Institute of Plant Sciences, University of Bern, Bern, Switzerland
| | - David G Heckel
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | | | | |
Collapse
|
27
|
Miners JO, Rowland A, Novak JJ, Lapham K, Goosen TC. Evidence-based strategies for the characterisation of human drug and chemical glucuronidation in vitro and UDP-glucuronosyltransferase reaction phenotyping. Pharmacol Ther 2020; 218:107689. [PMID: 32980440 DOI: 10.1016/j.pharmthera.2020.107689] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022]
Abstract
Enzymes of the UDP-glucuronosyltransferase (UGT) superfamily contribute to the elimination of drugs from almost all therapeutic classes. Awareness of the importance of glucuronidation as a drug clearance mechanism along with increased knowledge of the enzymology of drug and chemical metabolism has stimulated interest in the development and application of approaches for the characterisation of human drug glucuronidation in vitro, in particular reaction phenotyping (the fractional contribution of the individual UGT enzymes responsible for the glucuronidation of a given drug), assessment of metabolic stability, and UGT enzyme inhibition by drugs and other xenobiotics. In turn, this has permitted the implementation of in vitro - in vivo extrapolation approaches for the prediction of drug metabolic clearance, intestinal availability, and drug-drug interaction liability, all of which are of considerable importance in pre-clinical drug development. Indeed, regulatory agencies (FDA and EMA) require UGT reaction phenotyping for new chemical entities if glucuronidation accounts for ≥25% of total metabolism. In vitro studies are most commonly performed with recombinant UGT enzymes and human liver microsomes (HLM) as the enzyme sources. Despite the widespread use of in vitro approaches for the characterisation of drug and chemical glucuronidation by HLM and recombinant enzymes, evidence-based guidelines relating to experimental approaches are lacking. Here we present evidence-based strategies for the characterisation of drug and chemical glucuronidation in vitro, and for UGT reaction phenotyping. We anticipate that the strategies will inform practice, encourage development of standardised experimental procedures where feasible, and guide ongoing research in the field.
Collapse
Affiliation(s)
- John O Miners
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | | | | | | |
Collapse
|
28
|
Hindley G, Beck K, Borgan F, Ginestet CE, McCutcheon R, Kleinloog D, Ganesh S, Radhakrishnan R, D'Souza DC, Howes OD. Psychiatric symptoms caused by cannabis constituents: a systematic review and meta-analysis. Lancet Psychiatry 2020; 7:344-353. [PMID: 32197092 PMCID: PMC7738353 DOI: 10.1016/s2215-0366(20)30074-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Approximately 188 million people use cannabis yearly worldwide, and it has recently been legalised in 11 US states, Canada, and Uruguay for recreational use. The potential for increased cannabis use highlights the need to better understand its risks, including the acute induction of psychotic and other psychiatric symptoms. We aimed to investigate the effect of the cannabis constituent Δ9-tetrahydrocannabinol (THC) alone and in combination with cannabidiol (CBD) compared with placebo on psychiatric symptoms in healthy people. METHODS In this systematic review and meta-analysis, we searched MEDLINE, Embase, and PsycINFO for studies published in English between database inception and May 21, 2019, with a within-person, crossover design. Inclusion criteria were studies reporting symptoms using psychiatric scales (the Brief Psychiatric Rating Scale [BPRS] and the Positive and Negative Syndrome Scale [PANSS]) following the acute administration of intravenous, oral, or nasal THC, CBD, and placebo in healthy participants, and presenting data that allowed calculation of standardised mean change (SMC) scores for positive (including delusions and hallucinations), negative (such as blunted affect and amotivation), and general (including depression and anxiety) symptoms. We did a random-effects meta-analysis to assess the main outcomes of the effect sizes for total, positive, and negative PANSS and BPRS scores measured in healthy participants following THC administration versus placebo. Because the number of studies to do a meta-analysis on CBD's moderating effects was insufficient, this outcome was only systematically reviewed. This study is registered with PROSPERO, CRD42019136674. FINDINGS 15 eligible studies involving the acute administration of THC and four studies on CBD plus THC administration were identified. Compared with placebo, THC significantly increased total symptom severity with a large effect size (assessed in nine studies, with ten independent samples, involving 196 participants: SMC 1·10 [95% CI 0·92-1·28], p<0·0001); positive symptom severity (assessed in 14 studies, with 15 independent samples, involving 324 participants: SMC 0·91 [95% CI 0·68-1·14], p<0·0001); and negative symptom severity with a large effect size (assessed in 12 studies, with 13 independent samples, involving 267 participants: SMC 0·78 [95% CI 0·59-0·97], p<0·0001). In the systematic review, of the four studies evaluating CBD's effects on THC-induced symptoms, only one identified a significant reduction in symptoms. INTERPRETATION A single THC administration induces psychotic, negative, and other psychiatric symptoms with large effect sizes. There is no consistent evidence that CBD induces symptoms or moderates the effects of THC. These findings highlight the potential risks associated with the use of cannabis and other cannabinoids that contain THC for recreational or therapeutic purposes. FUNDING UK Medical Research Council, Maudsley Charity, Brain and Behavior Research Foundation, Wellcome Trust, and the UK National Institute for Health Research.
Collapse
Affiliation(s)
- Guy Hindley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Katherine Beck
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; South London and the Maudsley NHS Foundation Trust, London, UK; MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Cedric E Ginestet
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robert McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniel Kleinloog
- Department of Intensive Care Medicine, Leiden University Medical Hospital, Leiden, Netherlands
| | - Suhas Ganesh
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Rajiv Radhakrishnan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Deepak Cyril D'Souza
- Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; South London and the Maudsley NHS Foundation Trust, London, UK; MRC London Institute of Medical Sciences, Hammersmith Hospital Campus, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
29
|
Milani N, Qiu N, Molitor B, Badée J, Cruciani G, Fowler S. Use of Phenotypically Poor Metabolizer Individual Donor Human Liver Microsomes To Identify Selective Substrates of UGT2B10. Drug Metab Dispos 2020; 48:176-186. [PMID: 31839590 PMCID: PMC11022891 DOI: 10.1124/dmd.119.089482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/02/2019] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferase (UGT)1A4 and UGT2B10 are the human UGT isoforms most frequently involved in N-glucuronidation of drugs. UGT2B10 exhibits higher affinity than UGT1A4 for numerous substrates, making it potentially the more important enzyme for metabolism of these compounds in vivo. Clinically relevant UGT2B10 polymorphisms, including a null activity splice site mutation common in African populations, can lead to large exposure differences for UGT2B10 substrates that may limit their developability as marketed drugs. UGT phenotyping approaches using recombinantly expressed UGTs are limited by low enzyme activity and lack of validation of scaling to in vivo. In this study, we describe the use of an efficient experimental protocol for identification of UGT2B10-selective substrates (i.e., those with high fraction metabolized by UGT2B10), which exploits the activity difference between pooled human liver microsomes (HLM) and HLM from a phenotypically UGT2B10 poor metabolizer donor. Following characterization of the approach with eight known UGT2B10 substrates, we used ligand-based virtual screening and literature precedents to select 24 potential UGT2B10 substrates of 140 UGT-metabolized drugs for testing. Of these, dothiepin, cidoxepin, cyclobenzaprine, azatadine, cyproheptadine, bifonazole, and asenapine were indicated to be selective UGT2B10 substrates that have not previously been described. UGT phenotyping experiments and tests comparing conjugative and oxidative clearance were then used to confirm these findings. These approaches provide rapid and sensitive ways to evaluate whether a potential drug candidate cleared via glucuronidation will be sensitive to UGT2B10 polymorphisms in vivo. SIGNIFICANCE STATEMENT: The role of highly polymorphic UDP-glucuronosyltransferase (UGT)2B10 is likely to be underestimated currently for many compounds cleared via N-glucuronidation due to high test concentrations often used in vitro and low activity of UGT2B10 preparations. The methodology described in this study can be combined with the assessment of UGT versus oxidative in vitro metabolism to rapidly identify compounds likely to be sensitive to UGT2B10 polymorphism (high fraction metabolized by UGT2B10), enabling either chemical modification or polymorphism risk assessment before candidate selection.
Collapse
Affiliation(s)
- Nicolo Milani
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| | - NaHong Qiu
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| | - Birgit Molitor
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| | - Justine Badée
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| | - Gabriele Cruciani
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.M., N.Q., B.M., S.F.); Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy (N.M., G.C.); and Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B.)
| |
Collapse
|
30
|
Li Z, Gao Y, Yang C, Xiang Y, Zhang W, Zhang T, Su R, Lu C, Zhuang X. Assessment and Confirmation of Species Difference in Nonlinear Pharmacokinetics of Atipamezole with Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2020; 48:41-51. [PMID: 31699808 DOI: 10.1124/dmd.119.089151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/24/2019] [Indexed: 11/22/2022] Open
Abstract
Atipamezole, an α 2-adrenoceptor antagonist, displayed nonlinear pharmacokinetics (PK) in rats. The aim of this study was to understand the underlying mechanisms of nonlinear PK in rats and linear PK in humans and develop physiologically based PK models (PBPK) to capture and validate this phenomenon. In vitro and in vivo data were generated to show that metabolism is the main clearance pathway of atipamezole and species differences exist. Where cytochrome P450 (P450) was responsible for the metabolism in rats with a low Michaelis constant, human-specific UDP-glucuronosyltransferase 2B10- and 1A4-mediated N-glucuronidation was identified as the leading contributor to metabolism in humans with a high V max capacity. Saturation of metabolism was observed in rats at pharmacologically relevant doses, but not in humans at clinically relevant doses. PBPK models were developed using GastroPlus software to predict the PK profile of atipamezole in rats after intravenous or intramuscular administration of 0.1 to 3 mg/kg doses. The model predicted the nonlinear PK of atipamezole in rats and predicted observed exposures within 2-fold across dose levels. Under the same model structure, a human PBPK model was developed using human in vitro metabolism data. The PBPK model well described human concentration-time profiles at 10-100 mg doses showing dose-proportional increases in exposure. This study demonstrated that PBPK is a useful tool to predict human PK when interspecies extrapolation is not applicable. The nonlinear PK in rat and linear PK in human were characterized in vitro and allowed the prospective human PK via intramuscular dosing to be predicted at the preclinical stage. SIGNIFICANCE STATEMENT: This study demonstrated that PBPK is a useful tool for predicting human PK when interspecies extrapolation is not applicable due to species unique metabolism. Atipamezole, for example, is metabolized by P450 in rats and by N-glucuronidation in humans that were hypothesized to be the underlying reasons for a nonlinear PK in rats and linear PK in humans. This was testified by PBPK simulation in this study.
Collapse
Affiliation(s)
- Zheng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - You Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Chunmiao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Yanan Xiang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Tianhong Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Chuang Lu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China (Z.L., Y.G., C.Y., Y.X., W.Z., T.Z., R.S., X.Z.); and Department of DMPK, Sanofi Company, Waltham, Massachusetts (C.L.)
| |
Collapse
|
31
|
Chen X, Jia W, Zhu L, Mao L, Zhang Y. Recent advances in heterocyclic aromatic amines: An update on food safety and hazardous control from food processing to dietary intake. Compr Rev Food Sci Food Saf 2019; 19:124-148. [DOI: 10.1111/1541-4337.12511] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/17/2019] [Accepted: 10/30/2019] [Indexed: 01/14/2023]
Affiliation(s)
- Xiaoqian Chen
- Zhejiang Key Laboratory for Agro‐Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, College of Biosystems Engineering and Food ScienceZhejiang University Hangzhou China
| | - Wei Jia
- Zhejiang Key Laboratory for Agro‐Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, College of Biosystems Engineering and Food ScienceZhejiang University Hangzhou China
| | - Li Zhu
- Zhejiang Key Laboratory for Agro‐Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, College of Biosystems Engineering and Food ScienceZhejiang University Hangzhou China
| | - Lei Mao
- Department of NutritionSchool of Public Health, Zhejiang University School of Medicine Hangzhou China
| | - Yu Zhang
- Zhejiang Key Laboratory for Agro‐Food Processing, National Engineering Laboratory of Intelligent Food Technology and Equipment, College of Biosystems Engineering and Food ScienceZhejiang University Hangzhou China
| |
Collapse
|
32
|
Li Y, Meng Q, Yang M, Liu D, Hou X, Tang L, Wang X, Lyu Y, Chen X, Liu K, Yu AM, Zuo Z, Bi H. Current trends in drug metabolism and pharmacokinetics. Acta Pharm Sin B 2019; 9:1113-1144. [PMID: 31867160 PMCID: PMC6900561 DOI: 10.1016/j.apsb.2019.10.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Pharmacokinetics (PK) is the study of the absorption, distribution, metabolism, and excretion (ADME) processes of a drug. Understanding PK properties is essential for drug development and precision medication. In this review we provided an overview of recent research on PK with focus on the following aspects: (1) an update on drug-metabolizing enzymes and transporters in the determination of PK, as well as advances in xenobiotic receptors and noncoding RNAs (ncRNAs) in the modulation of PK, providing new understanding of the transcriptional and posttranscriptional regulatory mechanisms that result in inter-individual variations in pharmacotherapy; (2) current status and trends in assessing drug-drug interactions, especially interactions between drugs and herbs, between drugs and therapeutic biologics, and microbiota-mediated interactions; (3) advances in understanding the effects of diseases on PK, particularly changes in metabolizing enzymes and transporters with disease progression; (4) trends in mathematical modeling including physiologically-based PK modeling and novel animal models such as CRISPR/Cas9-based animal models for DMPK studies; (5) emerging non-classical xenobiotic metabolic pathways and the involvement of novel metabolic enzymes, especially non-P450s. Existing challenges and perspectives on future directions are discussed, and may stimulate the development of new research models, technologies, and strategies towards the development of better drugs and improved clinical practice.
Collapse
Affiliation(s)
- Yuhua Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
- The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Mengbi Yang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Hou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Tang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Wang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanfeng Lyu
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ai-Ming Yu
- UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Zhong Zuo
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong, China
| | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
33
|
Juvonen RO, Heikkinen AT, Kärkkäinen O, Jehangir R, Huuskonen J, Troberg J, Raunio H, Pentikäinen OT, Finel M. In vitro glucuronidation of 7-hydroxycoumarin derivatives in intestine and liver microsomes of Beagle dogs. Eur J Pharm Sci 2019; 141:105118. [PMID: 31669387 DOI: 10.1016/j.ejps.2019.105118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 12/31/2022]
Abstract
Beagle dog is a standard animal model for evaluating nonclinical pharmacokinetics of new drug candidates. Glucuronidation in intestine and liver is an important first-pass drug metabolic pathway, especially for phenolic compounds. This study evaluated the glucuronidation characteristics of several 7-hydroxycoumarin derivatives in beagle dog's intestine and liver in vitro. To this end, glucuronidation rates of 7-hydroxycoumarin (compound 1), 7-hydroxy-4-trifluoromethylcoumarin (2), 6-methoxy-7-hydroxycoumarin (3), 7-hydroxy-3-(4-tolyl)coumarin (4), 3-(4-fluorophenyl)coumarin (5), 7-hydroxy-3-(4-hydroxyphenyl)coumarin (6), 7-hydroxy-3-(4-methoxyphenyl)coumarin (7), and 7-hydroxy-3-(1H-1,2,4-tirazole)coumarin (8) were determined in dog's intestine and liver microsomes, as well as recombinant dog UGT1A enzymes. The glucuronidation rates of 1, 2 and 3 were 3-10 times higher in liver than in small intestine microsomes, whereas glucuronidation rates of 5, 6, 7 and 8 were similar in microsomes from both tissues. In the colon, glucuronidation of 1 and 2 was 3-5 times faster than in small intestine. dUGT1A11 glucuronidated efficiently all the substrates and was more efficient catalyst for 8 than any other dUGT1A. Other active enzymes were dUGT1A2 that glucuronidated efficiently 2, 3, 4, 5, 6 and 7, while dUGT1A10 glucuronidated efficiently 1, 2, 3, 4, 5 and 7. Kinetic analyses revealed that the compounds' Km values varied between 1.1 (dUGT1A10 and 2) and 250 µM (dUGT1A7 and 4). The results further strengthen the concept that dog intestine has high capacity for glucuronidation, and that different dUGT1As mediate glucuronidation with distinct substrates selectivity in dog and human.
Collapse
Affiliation(s)
- Risto O Juvonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Box 1627, FI-70211 Kuopio, Finland.
| | | | - Olli Kärkkäinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Box 1627, FI-70211 Kuopio, Finland
| | - Rabia Jehangir
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Box 1627, FI-70211 Kuopio, Finland
| | - Juhani Huuskonen
- University of Jyvaskyla, Department of Chemistry, P.O. Box 35, FI-40014 University of Jyvaskyla, Finland
| | - Johanna Troberg
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Hannu Raunio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Box 1627, FI-70211 Kuopio, Finland
| | - Olli T Pentikäinen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, FI-20014 University of Turku, Finland
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| |
Collapse
|
34
|
Stepanek O, Hin N, Thomas AG, Dash R, Alt J, Rais R, Rojas C, Slusher BS, Tsukamoto T. Neutral sphingomyelinase 2 inhibitors based on the 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol scaffold. Eur J Med Chem 2019; 170:276-289. [PMID: 30921693 PMCID: PMC9850959 DOI: 10.1016/j.ejmech.2019.03.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 01/21/2023]
Abstract
Neutral sphingomyelinase 2 (nSMase2), a key enzyme in ceramide biosynthesis, is a new therapeutic target for the treatment of neurological disorders and cancer. Using 2,6-dimethoxy-4-[4-phenyl-5-(2-thienyl)-1H-imidazol-2-yl]phenol (DPTIP), our initial hit compound (IC50 = 30 nM) from nSMase2 screening efforts, as a molecular template, a series of 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol derivatives were designed, synthesized, and evaluated. Systematic examination of various regions of DPTIP identified the key pharmacophore required for potent nSMase2 inhibition as well as a number of compounds with the 4-(1H-imidazol-2-yl)-2,6-dialkoxyphenol scaffold with similar or higher inhibitory potency against nSMase2 as compared to DPTIP. Among them, 4-(4,5-diisopropyl-1H-imidazol-2-yl)-2,6-dimethoxyphenol (25b) was found to be metabolically stable against P450 metabolism in liver microsomes and displayed higher plasma exposure following oral administration as compared to DPTIP. Analysis of plasma samples identified an O-glucuronide as the major metabolite. Blockade of the phase II metabolism should further facilitate our efforts to identify potent nSMase2 inhibitors with desirable ADME properties.
Collapse
Affiliation(s)
- Ondrej Stepanek
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Niyada Hin
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ranjeet Dash
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA,Address correspondence to: Tel: +1-410-614-0982;
| |
Collapse
|
35
|
Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-Glycosyltransferase (UGT) Superfamily: New Members, New Functions, and Novel Paradigms. Physiol Rev 2019; 99:1153-1222. [DOI: 10.1152/physrev.00058.2017] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.
Collapse
Affiliation(s)
- Robyn Meech
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Dong Gui Hu
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Ross A. McKinnon
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Siti Nurul Mubarokah
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Alex Z. Haines
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Pramod C. Nair
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Peter I. Mackenzie
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
36
|
Uno Y, Takahira R, Murayama N, Onozeki S, Kawamura S, Uehara S, Ikenaka Y, Ishizuka M, Ikushiro S, Yamazaki H. Functional and molecular characterization of UDP-glucuronosyltransferase 2 family in cynomolgus macaques. Biochem Pharmacol 2019; 163:335-344. [PMID: 30836059 DOI: 10.1016/j.bcp.2019.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/01/2019] [Indexed: 11/17/2022]
Abstract
UDP-glucuronosyltransferases (UGTs) are essential enzymes metabolizing endogenous and exogenous chemicals. However, characteristics of UGTs have not been fully investigated in molecular levels of cynomolgus macaques, one of non-human primates widely used in preclinical drug metabolism studies. In this study, three UGT2A cDNAs (UGT2A1, 2A2, and 2A3) were isolated and characterized along with seven UGT2Bs previously identified in cynomolgus macaques. Several transcript variants were found in cynomolgus UGT2A1 and UGT2A2, like human orthologs. Cynomolgus UGT2A and UGT2B amino acid sequences were highly identical (87-96%) to their human counterparts. By phylogenetic analysis, all these cynomolgus UGT2s were more closely clustered with their human homologs than with dog, rat, or mouse UGT2s. Especially, UGT2As showed orthologous relationships between humans and cynomolgus macaques. All the cynomolgus UGT2 mRNAs were expressed in livers, jejunum, and/or kidneys abundantly, except that UGT2A1 and UGT2A2 mRNAs were predominantly expressed in nasal mucosa, like human UGT2s. UGT2A and UGT2B genes together form a gene cluster in the cynomolgus and human genome. Among the seven cynomolgus UGT2Bs heterologously expressed in yeast, UGT2B9 and UGT2B30 showed activities in estradiol 17-O-glucuronidation and morphine 3-O-glucuronidation but did not show activities in estradiol 3-O-glucuronidation, similar to human UGT2Bs. In liver microsomes, cynomolgus macaques showed higher estradiol 17-O-glucuronidase and morphine 3-O-glucuronidase activities than humans, suggesting functional activities of the responsible UGT2B enzymes in cynomolgus macaques. Therefore, cynomolgus UGT2s had overall molecular similarities to human UGT2s, but also showed some differences in UGT2B enzyme properties.
Collapse
Affiliation(s)
- Yasuhiro Uno
- Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama 642-0017, Japan.
| | - Rika Takahira
- Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama 939-0398, Japan
| | - Norie Murayama
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Shunsuke Onozeki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Shu Kawamura
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Shotaro Uehara
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yoshinori Ikenaka
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Mayumi Ishizuka
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Shinichi Ikushiro
- Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama 939-0398, Japan.
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|
37
|
He K, Yonetani T, Asada Y, Echigo S, Itoh S. Simultaneous determination of carbamazepine-N-glucuronide and carbamazepine phase I metabolites in the wastewater by liquid chromatography-tandem mass spectrometry. Microchem J 2019. [DOI: 10.1016/j.microc.2018.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
38
|
Zhang T, Guo L, Yu F, Chen M, Wu B. The nuclear receptor Rev-erbα participates in circadian regulation of Ugt2b enzymes in mice. Biochem Pharmacol 2019; 161:89-97. [PMID: 30639455 DOI: 10.1016/j.bcp.2019.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 01/09/2019] [Indexed: 11/29/2022]
Abstract
Circadian clock is known to modulate phase I metabolism, however whether and how the phase II enzymes UDP-glucuronosyltransferases (UGTs) are regulated by circadian clock are largely unknown. In this study, we aimed to investigate a potential role of the clock gene Rev-erbα in regulation of Ugt2b enzymes. Ugt2b mRNA and protein expression in mouse livers were determined at a 4-h interval around the clock. Ugt2b activity was probed using morphine as a specific substrate. Regulation of Ugt2b by Rev-erbα was investigated using mouse hepatoma Hepa-1c1c7 cells and Rev-erbα knock-out (Rev-erbα-/-) mice. Luciferase reporter, mobility shift and chromatin immunoprecipitation (ChIP) assays were performed to identify the Rev-erbα binding site in Ugt2b36 promoter. Circadian variations in hepatic mRNA expression were observed for six Ugt2b genes (Ugt2b1, Ugt2b5, Ugt2b35, Ugt2b36, Ugt2b37, and Ugt2b38) in mice. Likewise, the total Ugt2b protein showed a circadian fluctuation. Glucuronidation of morphine (an Ugt2b substrate) both in vitro and in vivo was dosing-time dependent. Morphine glucuronidation was more extensive at the dosing time of ZT2 than at ZT14 consistent with the Ugt2b protein levels. Furthermore, Rev-erbα knockdown significantly increased Ugt2b mRNA and protein in Hepa-1c1c7 cells, whereas Rev-erbα overexpression or activation down-regulated Ugt2b expression. Moreover, Rev-erbα ablation in mice up-regulated the mRNA and protein expression of Ugt2b and blunted Ugt2b rhythmicity in the liver. In addition, Rev-erbα repressed the transcription of Ugt2b36 through specific binding to the -30 to -18 bp of promoter region based on a combination of luciferase reporter, mobility shift and ChIP assays. In summary, the clock gene Rev-erbα negatively regulates the expressions of Ugt2b genes, contributing to their circadian variations.
Collapse
Affiliation(s)
- Tianpeng Zhang
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| | - Lianxia Guo
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| | - Fangjun Yu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| | - Min Chen
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China.
| |
Collapse
|
39
|
In silico prediction of Heterocyclic Aromatic Amines metabolism susceptible to form DNA adducts in humans. Toxicol Lett 2019; 300:18-30. [DOI: 10.1016/j.toxlet.2018.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/02/2018] [Accepted: 10/08/2018] [Indexed: 11/19/2022]
|
40
|
Lapham K, Lin J, Novak J, Orozco C, Niosi M, Di L, Goosen TC, Ryu S, Riccardi K, Eng H, Cameron KO, Kalgutkar AS. 6-Chloro-5-[4-(1-Hydroxycyclobutyl)Phenyl]-1 H-Indole-3-Carboxylic Acid is a Highly Selective Substrate for Glucuronidation by UGT1A1, Relative to β-Estradiol. Drug Metab Dispos 2018; 46:1836-1846. [PMID: 30194276 DOI: 10.1124/dmd.118.083709] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/05/2018] [Indexed: 02/13/2025] Open
Abstract
6-Chloro-5-[4-(1-hydroxycyclobutyl)phenyl]-1H-indole-3-carboxylic acid (PF-06409577) is a direct activator of the human β1-containing adenosine monophosphate-activated protein kinase (ΑMPK) isoforms. The clearance mechanism of PF-06409577 in animals and humans involves uridine diphosphoglucuronosyl transferase (UGT)-mediated glucuronidation to an acyl glucuronide metabolite of PF-06409577 [(2S,3S,4S,5R,6S)-6-((6-chloro-5-(4-(1-hydroxycyclobutyl)phenyl)-1H-indole-3-carbonyl)oxy)-3,4,5-trihydroxytetrahydro-2H-pyran-2-carboxylic acid (M1)], which retains selective activation of human β1-containing AMPK isoforms. This paper describes a detailed characterization of the human UGT isoform(s) responsible for glucuronidation of PF-06409577 to M1. Studies using a panel of 13 human recombinant UGT (hrUGT) enzymes indicated that PF-06409577 was converted to M1 in a highly selective fashion by UGT1A1, which was further verified in human liver microsomes treated with specific chemical inhibitors, and in different UGT1A1 expressers. Conversion of PF-06409577 to M1 by UGT1A1 occurred in a relatively selective fashion, compared with β-estradiol (ES), a conventional probe substrate of UGT1A1. The Michaelis-Menten constant (K M) and V max values describing the formation of M1 from PF-06409577 in hrUGT1A1 and microsomal preparations from human intestine, liver, and kidney ranged from 131 to 212 μM (K M) and 107-3834 pmol/min per milligram (V max) in the presence of 2% bovine serum albumin. Relative activity factors (RAF) were determined for UGT1A1 using PF-06409577 and ES to enable estimation of intrinsic clearance from various tissues. RAF values from PF-06409577 and ES were generally comparable with the exception of intestinal microsomes, where ES overestimated the RAF of UGT1A1 due to glucuronidation by intestinal UGT1A8 and UGT1A10. Our results suggest the potential utility of PF-06409477 as a selective probe UGT1A1 substrate for UGT reaction phenotyping and inhibition studies in preclinical discovery/development.
Collapse
Affiliation(s)
- Kimberly Lapham
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Jian Lin
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Jonathan Novak
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Christine Orozco
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Mark Niosi
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Li Di
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Theunis C Goosen
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Sangwoo Ryu
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Keith Riccardi
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Heather Eng
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Kimberly O Cameron
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Inc., Groton, Connecticut (K.L., J.L., J.N., C.O., M.N., L.D., T.C.G., S.R., K.R., H.E.); and Medicine Design, Pfizer Inc., Cambridge, Massachusetts (K.O.C., A.S.K.)
| |
Collapse
|
41
|
Uno Y, Takahira R, Murayama N, Ishii Y, Ikenaka Y, Ishizuka M, Yamazaki H, Ikushiro S. Molecular and functional characterization of UDP-glucuronosyltransferase 1A in cynomolgus macaques. Biochem Pharmacol 2018; 155:172-181. [DOI: 10.1016/j.bcp.2018.06.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/22/2018] [Indexed: 12/19/2022]
|
42
|
Lu D, Dong D, Wu B. Highly selective N-glucuronidation of four piperazine-containing drugs by UDP-glucuronosyltransferase 2B10. Expert Opin Drug Metab Toxicol 2018; 14:989-998. [DOI: 10.1080/17425255.2018.1505862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Danyi Lu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| | - Dong Dong
- College of Medicine, Jinan University, Guangzhou, China
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, China
| |
Collapse
|
43
|
Lu D, Dong D, Xie Q, Li Z, Wu B. Disposition of Mianserin and Cyclizine in UGT2B10-Overexpressing Human Embryonic Kidney 293 Cells: Identification of UGT2B10 as a Novel N-Glucosidation Enzyme and Breast Cancer Resistance Protein as an N-Glucoside Transporter. Drug Metab Dispos 2018; 46:970-979. [PMID: 29691239 DOI: 10.1124/dmd.118.080804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/17/2018] [Indexed: 02/13/2025] Open
Abstract
UDP-glucuronosyltransferases (UGTs) play an important role in the metabolism and detoxification of amine-containing chemicals; however, the disposition mechanisms for amines via UGT metabolism are not fully clear. We aimed to investigate a potential role of UGT2B10 in N-glucosidation and to determine the transporters for the excretion of N-glucosides. We first established a human embryonic kidney cell line 293 (HEK293) that stably overexpressed UGT2B10. Incubation of mianserin or cyclizine with the cells generated one N-glucuronide and one N-glucoside. Chemical inhibition (using specific chemical inhibitor Ko143) and biologic inhibition [using specific short hairpin RNA of breast cancer resistance protein (BCRP)] resulted in a significant reduction in efflux of N-glucuronide. Similar results were observed when multidrug resistance-associated protein (MRP4) was inhibited. Furthermore, inhibition of BCRP led to increased intracellular N-glucoside, whereas inhibition of MRP4 caused no changes in disposition of N-glucoside. Overall, the data indicated that BCRP, not MRP4, was responsible for the excretion of N-glucosides, whereas both BCRP and MRP4 contributed to excretion of N-glucuronides. Interestingly, downregulation of N-glucuronidation led to increased N-glucosidation in the cells, supporting the glucosidation as a potential complementary pathway for conventional glucuronidation. In conclusion, UGT2B10 was for the first time identified as an N-glucosidation enzyme. Generated N-glucosides were excreted primarily by the BCRP transporter.
Collapse
Affiliation(s)
- Danyi Lu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy (D.L., Q.X., B.W.), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research (D.L., B.W.), and International Ocular Surface Research Centre and Institute of Ophthalmology, School of Medicine (D.D., Z.L.), Jinan University, Guangzhou, China; and Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (D.L.)
| | - Dong Dong
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy (D.L., Q.X., B.W.), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research (D.L., B.W.), and International Ocular Surface Research Centre and Institute of Ophthalmology, School of Medicine (D.D., Z.L.), Jinan University, Guangzhou, China; and Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (D.L.)
| | - Qian Xie
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy (D.L., Q.X., B.W.), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research (D.L., B.W.), and International Ocular Surface Research Centre and Institute of Ophthalmology, School of Medicine (D.D., Z.L.), Jinan University, Guangzhou, China; and Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (D.L.)
| | - Zhijie Li
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy (D.L., Q.X., B.W.), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research (D.L., B.W.), and International Ocular Surface Research Centre and Institute of Ophthalmology, School of Medicine (D.D., Z.L.), Jinan University, Guangzhou, China; and Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (D.L.)
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy (D.L., Q.X., B.W.), Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research (D.L., B.W.), and International Ocular Surface Research Centre and Institute of Ophthalmology, School of Medicine (D.D., Z.L.), Jinan University, Guangzhou, China; and Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (D.L.)
| |
Collapse
|
44
|
Sanchez-Dominguez CN, Gallardo-Blanco HL, Salinas-Santander MA, Ortiz-Lopez R. Uridine 5'-diphospho-glucronosyltrasferase: Its role in pharmacogenomics and human disease. Exp Ther Med 2018; 16:3-11. [PMID: 29896223 DOI: 10.3892/etm.2018.6184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022] Open
Abstract
Biotransformation is an enzyme-catalyzed process in which the body converts endogenous compounds, xenobiotics and toxic substances into harmless or easily excreted metabolites. The biotransformation reactions are classified as phase I and II reactions. Uridine 5'-diphospho (UDP)-glucuronosyltransferases (UGTs) are a superfamily of phase II enzymes which have roles in the conjugation of xenobiotics or endogenous compounds, including drugs and bilirubin, with glucuronic acid to make them easier to excrete. The method the human body uses to achieve glucuronidation may be affected by a large interindividual variation due to changes in the sequences of the genes encoding these enzymes. In the last five years, the study of the genetic variants of the UGTs at a molecular level has become important due to its association with several diseases and the ability to predict adverse events due to drug metabolism. In the present review, the structure and the prominent genetic variants of the UGT1A subfamily and their metabolic and clinical implications are described.
Collapse
Affiliation(s)
- Celia N Sanchez-Dominguez
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon 64460, Mexico
| | - Hugo L Gallardo-Blanco
- Department of Genetics, Faculty of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon 64460, Mexico
| | | | - Rocio Ortiz-Lopez
- Tecnologico de Monterrey, Medical School and Health Sciences, Monterrey, Nuevo Leon 64710, Mexico
| |
Collapse
|
45
|
Dwivedi P, Zhou X, Powell TG, Calafat AM, Ye X. Impact of enzymatic hydrolysis on the quantification of total urinary concentrations of chemical biomarkers. CHEMOSPHERE 2018; 199:256-262. [PMID: 29448192 PMCID: PMC5941949 DOI: 10.1016/j.chemosphere.2018.01.177] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 05/25/2023]
Abstract
Human exposure to consumer and personal care products chemicals such as phenols, including parabens and other antimicrobial agents, can be assessed through biomonitoring by quantifying urinary concentrations of the parent chemical or its metabolites, often after hydrolysis of phase II conjugates. Developing suitable analytical methods for the concurrent quantification of multiple exposure biomarkers is challenging because optimal conditions for the hydrolysis of such conjugates (e.g., O-glucuronides, N-glucuronides, sulfates) may differ depending on the biomarker. We evaluated the effectiveness of seven commercial hydrolytic enzymes to simultaneously hydrolyze N-glucuronides (using the antibacterial triclocarban as example compound) and other conjugates (using select phenols and parabens as examples) by using on-line solid phase extraction-high performance liquid chromatography-isotope dilution-tandem mass spectrometry. Incubation (30 min, 55 °C) with a genetically engineered β-glucuronidase (IMCS, ≥15 units/μL urine) hydrolyzed N-glucuronide triclocarban, but did not fully hydrolyze the conjugates of phenols and parabens. By contrast, incubation (4 h, 37 °C) with solid β-glucuronidase (Helix pomatia, Type H-1, ≥30 units/μL urine) or liquid β-glucuronidase/arylsulfatase (Helix pomatia, 30 units/μL urine [i.e., 30 μL/100 μL urine]) in the presence of 100 μL methanol for 100 μL urine completely hydrolyzed N-glucuronide triclocarban and the conjugates of several phenols and parabens, without cleaving the ester bond of the parabens to form p-hydroxybenzoic acid. These results highlight the relevance of method validation procedures that include optimizing the hydrolysis of phase II urinary conjugates (e.g., enzyme type and amount used, reaction time, temperature) to quantify accurately and concurrently multiple exposure biomarkers for biomonitoring purposes.
Collapse
Affiliation(s)
- Prabha Dwivedi
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Mailstop F-53, Atlanta, GA 30341, USA.
| | - Xiaoliu Zhou
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Mailstop F-53, Atlanta, GA 30341, USA
| | - Tolar G Powell
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Mailstop F-53, Atlanta, GA 30341, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Mailstop F-53, Atlanta, GA 30341, USA
| | - Xiaoyun Ye
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, 4770 Buford Hwy, Mailstop F-53, Atlanta, GA 30341, USA
| |
Collapse
|
46
|
Lee CA, Yang C, Shah V, Shen Z, Wilson DM, Ostertag TM, Girardet JL, Hall J, Gillen M. Metabolism and Disposition of Verinurad, a Uric Acid Reabsorption Inhibitor, in Humans. Drug Metab Dispos 2018; 46:532-541. [PMID: 29490903 DOI: 10.1124/dmd.117.078220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/23/2018] [Indexed: 12/18/2022] Open
Abstract
Verinurad (RDEA3170) is a second generation selective uric acid reabsorption inhibitor for the treatment of gout and asymptomatic hyperuricemia. Following a single oral solution of 10-mg dose of [14C]verinurad (500 μCi), verinurad was rapidly absorbed with a median time to occurrence of maximum observed concentration (Tmax) of 0.5 hours and terminal half-life of 15 hours. In plasma, verinurad constituted 21% of total radioactivity. Recovery of radioactivity in urine and feces was 97.1%. Unchanged verinurad was the predominant component in the feces (29.9%), whereas levels were low in the urine (1.2% excreted). Acylglucuronide metabolites M1 (direct glucuronidation) and M8 (glucuronidation of N-oxide) were formed rapidly after absorption of verinurad with terminal half-life values of approximately 13 and 18 hours, respectively. M1 and M8 constituted 32% and 31% of total radioactivity in plasma and were equimolar to verinurad on the basis of AUC ratios. M1 and M8 formed in the liver were biliary cleared with complete hydrolysis in the GI tract, as metabolites were not detected in the feces and/or efflux across the sinusoidal membrane; M1 and M8 accounted for 29.2% and 32.5% of the radioactive dose in urine, respectively. In vitro studies demonstrated that CYP3A4 mediated the formation of the N-oxide metabolite (M4), which was further metabolized by glucuronyl transferases (UGTs) to form M8, as M4 was absent in plasma and only trace levels were present in the urine. Several UGTs mediated the formation of M1, which could also be further metabolized by CYP2C8. Overall, the major clearance route of verinurad is metabolism via UGTs and CYP3A4 and CYP2C8.
Collapse
Affiliation(s)
- Caroline A Lee
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Chun Yang
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Vishal Shah
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Zancong Shen
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - David M Wilson
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Traci M Ostertag
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Jean-Luc Girardet
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Jesse Hall
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| | - Michael Gillen
- Preclinical and Clinical DMPK (C.A.L., C.Y.,V.S., Z.S.), Bioanalytical (D.M.W.), Biology (T.M.O.), Chemistry (J.-L.G.), and Clinical Development (J.H.) Ardea Biosciences, Inc., San Diego, California; and Early Clinical Development, IMED Biotech Unit, Quantitative Clinical Pharmacology (M.G.) AstraZeneca LP, Gaithersburg, Maryland
| |
Collapse
|
47
|
Labriet A, Allain EP, Rouleau M, Audet-Delage Y, Villeneuve L, Guillemette C. Post-transcriptional Regulation of UGT2B10 Hepatic Expression and Activity by Alternative Splicing. Drug Metab Dispos 2018; 46:514-524. [PMID: 29438977 PMCID: PMC5894810 DOI: 10.1124/dmd.117.079921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/31/2018] [Indexed: 12/17/2022] Open
Abstract
The detoxification enzyme UDP-glucuronosyltransferase UGT2B10 is specialized in the N-linked glucuronidation of many drugs and xenobiotics. Preferred substrates possess tertiary aliphatic amines and heterocyclic amines, such as tobacco carcinogens and several antidepressants and antipsychotics. We hypothesized that alternative splicing (AS) constitutes a means to regulate steady-state levels of UGT2B10 and enzyme activity. We established the transcriptome of UGT2B10 in normal and tumoral tissues of multiple individuals. The highest expression was in the liver, where 10 AS transcripts represented 50% of the UGT2B10 transcriptome in 50 normal livers and 44 hepatocellular carcinomas. One abundant class of transcripts involves a novel exonic sequence and leads to two alternative (alt.) variants with novel in-frame C termini of 10 or 65 amino acids. Their hepatic expression was highly variable among individuals, correlated with canonical transcript levels, and was 3.5-fold higher in tumors. Evidence for their translation in liver tissues was acquired by mass spectrometry. In cell models, they colocalized with the enzyme and influenced the conjugation of amitriptyline and levomedetomidine by repressing or activating the enzyme (40%-70%; P < 0.01) in a cell context-specific manner. A high turnover rate for the alt. proteins, regulated by the proteasome, was observed in contrast to the more stable UGT2B10 enzyme. Moreover, a drug-induced remodeling of UGT2B10 splicing was demonstrated in the HepaRG hepatic cell model, which favored alt. variants expression over the canonical transcript. Our findings support a significant contribution of AS in the regulation of UGT2B10 expression in the liver with an impact on enzyme activity.
Collapse
Affiliation(s)
- Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Eric P Allain
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Yannick Audet-Delage
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Québec, Canada Research Chair in Pharmacogenomics, Université Laval, Québec, Canada
| |
Collapse
|
48
|
N-3 Polyunsaturated Fatty Acids Stimulate Bile Acid Detoxification in Human Cell Models. Can J Gastroenterol Hepatol 2018; 2018:6031074. [PMID: 29850457 PMCID: PMC5907406 DOI: 10.1155/2018/6031074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/07/2018] [Indexed: 01/16/2023] Open
Abstract
Cholestasis is characterized by the accumulation of toxic bile acids (BAs) in liver cells. The present study aimed to evaluate the effects of n-3 polyunsaturated fatty acids (n-3 PUFAs), such as docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids, on BA homeostasis and toxicity in human cell models. The effects of EPA and/or DHA on the expression of genes involved in the maintenance of BA homeostasis were analyzed in human hepatoma (HepG2) and colon carcinoma (Caco-2) cells, as well as in primary culture of human intestinal (InEpC) and renal (RPTEC) cells. Extracellular BA species were quantified in culture media using LC-MS/MS. BA-induced toxicity was evaluated using caspase-3 and flow cytometry assays. Gene expression analyses of HepG2 cells reveal that n-3 PUFAs reduce the expression of genes involved in BA synthesis (CYP7A1, CYP27A1) and uptake (NTCP), while activating genes encoding metabolic enzymes (SULT2A1) and excretion transporters (MRP2, MRP3). N-3 PUFAs also generate a less toxic BA pool and prevent the BA-dependent activation of apoptosis in HepG2 cells. Conclusion. The present study reveals that n-3 PUFAs stimulate BA detoxification.
Collapse
|
49
|
Sandbaumhüter FA, Thormann W. Enantioselective capillary electrophoresis provides insight into the phase II metabolism of ketamine and its metabolites in vivo and in vitro. Electrophoresis 2018; 39:1478-1481. [PMID: 29572863 DOI: 10.1002/elps.201800012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/23/2018] [Accepted: 03/08/2018] [Indexed: 01/31/2023]
Abstract
Glucuronidation catalyzed by uridine-5'-diphospho-glucuronosyl-transferases (UGTs) is the most important reaction in phase II metabolism of drugs and other compounds. O-glucuronidation is more common than N-glucuronidation. The anesthetic, analgesic and antidepressive drug ketamine is metabolized in phase I by cytochrome P450 enzymes to norketamine, hydroxynorketamine (HNK) diastereomers and dehydronorketamine (DHNK). Equine urine samples collected two hours after ketamine injection were treated with β-glucuronidase and analyzed with three enantioselective capillary electrophoresis assays. Concentrations of HNK diastereomers and norketamine were significantly higher in comparison to untreated urine and an increase of ketamine and DHNK levels was found in selected but not all samples. This suggests that O-glucuronides of HNK and N-glucuronides of the other compounds are formed in equines. N-glucuronidation of norketamine was studied in vitro with liver microsomes of different species and the single human enzyme UGT1A4. With equine liver microsomes (ELM) a stereoselective N-glucuronidation of norketamine was found that compares well to the results obtained with urines collected after ketamine administration. No reaction was observed with canine liver microsomes, human liver microsomes and UGT1A4. Incubation of ketamine and DHNK with ELM did not reveal any glucuronidation. Enantioselective CE is suitable to provide insight into the phase II metabolism of ketamine and its metabolites.
Collapse
Affiliation(s)
- Friederike A Sandbaumhüter
- Clinical Pharmacology Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Wolfgang Thormann
- Clinical Pharmacology Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| |
Collapse
|
50
|
Exploring the Metabolism of (+)-[ 18F]Flubatine in Vitro and in Vivo: LC-MS/MS Aided Identification of Radiometabolites in a Clinical PET Study. Molecules 2018; 23:molecules23020464. [PMID: 29461507 PMCID: PMC6017759 DOI: 10.3390/molecules23020464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 01/25/2023] Open
Abstract
Both (+)-[18F]flubatine and its enantiomer (−)-[18F]flubatine are radioligands for the neuroimaging of α4β2 nicotinic acetylcholine receptors (nAChRs) by positron emission tomography (PET). In a clinical study in patients with early Alzheimer’s disease, (+)-[18F]flubatine ((+)-[18F]1) was examined regarding its metabolic fate, in particular by identification of degradation products detected in plasma and urine. The investigations included an in vivo study of (+)-flubatine ((+)-1) in pigs and structural elucidation of formed metabolites by LC-MS/MS. Incubations of (+)-1 and (+)-[18F]1 with human liver microsomes were performed to generate in vitro metabolites, as well as radiometabolites, which enabled an assignment of their structures by comparison of LC-MS/MS and radio-HPLC data. Plasma and urine samples taken after administration of (+)-[18F]1 in humans were examined by radio-HPLC and, on the basis of results obtained in vitro and in vivo, formed radiometabolites were identified. In pigs, (+)-1 was monohydroxylated at different sites of the azabicyclic ring system of the molecule. Additionally, one intermediate metabolite underwent glucuronidation, as also demonstrated in vitro. In humans, a fraction of 95.9 ± 1.9% (n = 10) of unchanged tracer remained in plasma, 30 min after injection. However, despite the low metabolic degradation, both radiometabolites formed in humans could be characterized as (i) a product of C-hydroxylation at the azabicyclic ring system, and (ii) a glucuronide conjugate of the precedingly-formed N8-hydroxylated (+)-[18F]1.
Collapse
|