1
|
Ueda H, Narumi K, Asano S, Saito Y, Furugen A, Kobayashi M. Comparative study on the occurrence of adverse effects in the concomitant use of azathioprine and aldehyde oxidase inhibitors. Expert Opin Drug Saf 2024; 23:89-97. [PMID: 38097359 DOI: 10.1080/14740338.2023.2295976] [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: 06/15/2023] [Accepted: 11/28/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES Aldehyde oxidase (AO) is a molybdenum-containing redox enzyme similar to xanthine oxidase that is involved in the thiopurine metabolism. This study investigated the effects of drug-drug interactions (DDIs) between azathioprine (AZA) and AO inhibitors on hematologic and hepatic disorders using the U.S. Food and Drug Administration Adverse Event Reporting System and the Japanese Adverse Drug Event Report database. METHODS The presence of DDI was assessed using the interaction signal scores (ISSs) calculated via the reporting odds ratios and 95% confidence intervals. The study used reports of 'azathioprine' as a suspect drug for adverse effects. AO inhibitors were selected based on previous in vitro reports. RESULTS Some drugs tested positive for ISSs in each database and type of adverse effect (hematologic or hepatic disorder) analysis. Among these drugs, chlorpromazine, clozapine, hydralazine, and quetiapine could inhibit AZA metabolism via AO, given the previously reported clinical blood concentration and inhibitory effects of each drug. CONCLUSION Concomitant use of AO inhibitors increased the signals for AZA-induced adverse effects. To date, no studies have evaluated the clinical importance of AO as a drug-metabolizing enzyme, and further in vitro and clinical research is needed to clarify the contribution of AO to the pharmacokinetics of thiopurines.
Collapse
Affiliation(s)
- Hinata Ueda
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Education Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shuho Asano
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshitaka Saito
- Department of Clinical Pharmaceutics & Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan
| | - Ayako Furugen
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Education Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Izat N, Bolleddula J, Abbasi A, Cheruzel L, Jones RS, Moss D, Ortega-Muro F, Parmentier Y, Peterkin VC, Tian DD, Venkatakrishnan K, Zientek MA, Barber J, Houston JB, Galetin A, Scotcher D. Challenges and Opportunities for In Vitro-In Vivo Extrapolation of Aldehyde Oxidase-Mediated Clearance: Toward a Roadmap for Quantitative Translation. Drug Metab Dispos 2023; 51:1591-1606. [PMID: 37751998 DOI: 10.1124/dmd.123.001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.
Collapse
Affiliation(s)
- Nihan Izat
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jayaprakasam Bolleddula
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Armina Abbasi
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Lionel Cheruzel
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Robert S Jones
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Darren Moss
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Fatima Ortega-Muro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Yannick Parmentier
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Vincent C Peterkin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Dan-Dan Tian
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Karthik Venkatakrishnan
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Michael A Zientek
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| |
Collapse
|
3
|
Gajula SNR, Nathani TN, Patil RM, Talari S, Sonti R. Aldehyde oxidase mediated drug metabolism: an underpredicted obstacle in drug discovery and development. Drug Metab Rev 2022; 54:427-448. [PMID: 36369949 DOI: 10.1080/03602532.2022.2144879] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aldehyde oxidase (AO) has garnered curiosity as a non-CYP metabolizing enzyme in drug development due to unexpected consequences such as toxic metabolite generation and high metabolic clearance resulting in the clinical failure of new drugs. Therefore, poor AO mediated clearance prediction in preclinical nonhuman species remains a significant obstacle in developing novel drugs. Various isoforms of AO, such as AOX1, AOX3, AOX3L1, and AOX4 exist across species, and different AO activity among humans influences the AO mediated drug metabolism. Therefore, carefully considering the unique challenges is essential in developing successful AO substrate drugs. The in vitro to in vivo extrapolation underpredicts AO mediated drug clearance due to the lack of reliable representative animal models, substrate-specific activity, and the discrepancy between absolute concentration and activity. An in vitro tool to extrapolate in vivo clearance using a yard-stick approach is provided to address the underprediction of AO mediated drug clearance. This approach uses a range of well-known AO drug substrates as calibrators for qualitative scaling new drugs into low, medium, or high clearance category drugs. So far, in vivo investigations on chimeric mice with humanized livers (humanized mice) have predicted AO mediated metabolism to the best extent. This review addresses the critical aspects of the drug discovery stage for AO metabolism studies, challenges faced in drug development, approaches to tackle AO mediated drug clearance's underprediction, and strategies to decrease the AO metabolism of drugs.
Collapse
Affiliation(s)
- Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Telangana, India
| | - Tanaaz Navin Nathani
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Telangana, India
| | - Rashmi Madhukar Patil
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Telangana, India
| | - Sasikala Talari
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Telangana, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Balanagar, Telangana, India
| |
Collapse
|
4
|
Rendić SP, Crouch RD, Guengerich FP. Roles of selected non-P450 human oxidoreductase enzymes in protective and toxic effects of chemicals: review and compilation of reactions. Arch Toxicol 2022; 96:2145-2246. [PMID: 35648190 PMCID: PMC9159052 DOI: 10.1007/s00204-022-03304-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022]
Abstract
This is an overview of the metabolic reactions of drugs, natural products, physiological compounds, and other (general) chemicals catalyzed by flavin monooxygenase (FMO), monoamine oxidase (MAO), NAD(P)H quinone oxidoreductase (NQO), and molybdenum hydroxylase enzymes (aldehyde oxidase (AOX) and xanthine oxidoreductase (XOR)), including roles as substrates, inducers, and inhibitors of the enzymes. The metabolism and bioactivation of selected examples of each group (i.e., drugs, "general chemicals," natural products, and physiological compounds) are discussed. We identified a higher fraction of bioactivation reactions for FMO enzymes compared to other enzymes, predominately involving drugs and general chemicals. With MAO enzymes, physiological compounds predominate as substrates, and some products lead to unwanted side effects or illness. AOX and XOR enzymes are molybdenum hydroxylases that catalyze the oxidation of various heteroaromatic rings and aldehydes and the reduction of a number of different functional groups. While neither of these two enzymes contributes substantially to the metabolism of currently marketed drugs, AOX has become a frequently encountered route of metabolism among drug discovery programs in the past 10-15 years. XOR has even less of a role in the metabolism of clinical drugs and preclinical drug candidates than AOX, likely due to narrower substrate specificity.
Collapse
Affiliation(s)
| | - Rachel D Crouch
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, 37204, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| |
Collapse
|
5
|
Xiao G, Chen YL, Dedic N, Xie L, Koblan KS, Galluppi GR. In Vitro ADME and Preclinical Pharmacokinetics of Ulotaront, a TAAR1/5-HT 1A Receptor Agonist for the Treatment of Schizophrenia. Pharm Res 2022; 39:837-850. [PMID: 35484370 PMCID: PMC9160101 DOI: 10.1007/s11095-022-03267-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/20/2022] [Indexed: 11/26/2022]
Abstract
Purpose Ulotaront (SEP-363856) is a TAAR1 agonist with 5-HT1A agonist activity currently in clinical development for the treatment of schizophrenia. The objectives of the current study were to characterize the in vitro ADME properties, preclinical PK, and to evaluate the DDI potential of ulotaront and its major metabolite SEP-383103. Methods Solubility, permeability, plasma protein binding, CYP inhibition and induction, transporter inhibition and uptake studies were conducted in vitro. Phenotyping studies were conducted using recombinant human CYPs and FMOs, human liver microsomes and human liver homogenates. Preclinical plasma and brain pharmacokinetics were determined after a single intraperitoneal, intravenous, and oral administration of ulotaront. Results Ulotaront is a compound of high solubility, high permeability, and low binding to plasma proteins. Ulotaront metabolism is mediated via both NADPH-dependent and NADPH-independent pathways, with CYP2D6 as the major metabolizing enzyme. Ulotaront is an inducer of CYP2B6, and an inhibitor of CYP2D6, OCT1 and OCT2, while SEP-383103 is neither a CYP inducer nor a potent inhibitor of CYPs and human transporters. Ulotaront exhibits rapid absorption, greater than 70% bioavailability, approximately 3.5 L/kg volume of distribution, 1.5-4 h half-life, 12-43 ml/min/kg clearance, and good penetration across the blood–brain barrier in preclinical species. Conclusions Ulotaront has been designated as a BCS1 compound by US FDA. The ability of ulotaront to penetrate the blood–brain barrier for CNS targeting has been demonstrated in mice and rats. The potential for ulotaront and SEP-383103 to act as perpetrators of CYP and transporter-mediated DDIs is predicted to be remote. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03267-1.
Collapse
Affiliation(s)
- Guangqing Xiao
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA.
| | - Yu-Luan Chen
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Nina Dedic
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Linghong Xie
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Kenneth S Koblan
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Gerald R Galluppi
- DMPK and Clinical Pharmacology, Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| |
Collapse
|
6
|
Mota C, Diniz A, Coelho C, Santos-Silva T, Esmaeeli M, Leimkühler S, Cabrita EJ, Marcelo F, Romão MJ. Interrogating the Inhibition Mechanisms of Human Aldehyde Oxidase by X-ray Crystallography and NMR Spectroscopy: The Raloxifene Case. J Med Chem 2021; 64:13025-13037. [PMID: 34415167 DOI: 10.1021/acs.jmedchem.1c01125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human aldehyde oxidase (hAOX1) is mainly present in the liver and has an emerging role in drug metabolism, since it accepts a wide range of molecules as substrates and inhibitors. Herein, we employed an integrative approach by combining NMR, X-ray crystallography, and enzyme inhibition kinetics to understand the inhibition modes of three hAOX1 inhibitors-thioridazine, benzamidine, and raloxifene. These integrative data indicate that thioridazine is a noncompetitive inhibitor, while benzamidine presents a mixed type of inhibition. Additionally, we describe the first crystal structure of hAOX1 in complex with raloxifene. Raloxifene binds tightly at the entrance of the substrate tunnel, stabilizing the flexible entrance gates and elucidating an unusual substrate-dependent mechanism of inhibition with potential impact on drug-drug interactions. This study can be considered as a proof-of-concept for an efficient experimental screening of prospective substrates and inhibitors of hAOX1 relevant in drug discovery.
Collapse
Affiliation(s)
- Cristiano Mota
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Ana Diniz
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Catarina Coelho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Teresa Santos-Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Mariam Esmaeeli
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Silke Leimkühler
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Eurico J Cabrita
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Filipa Marcelo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Maria João Romão
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| |
Collapse
|
7
|
Case Study 11: Considerations for Enzyme Mapping Experiments-Interaction Between the Aldehyde Oxidase Inhibitor Hydralazine and Glutathione. Methods Mol Biol 2021. [PMID: 34272718 DOI: 10.1007/978-1-0716-1554-6_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Often it may be convenient and efficient to address multiple research questions with a single experiment. In many instances, however, the best approach is to design the experiment to address one question at a time. The design of enzyme mapping experiments is discussed in this chapter, focusing on considerations pertinent to the study of aldehyde oxidase (AO) vs. cytochrome P450 metabolism. Specifically, a case is presented in which reduced glutathione (GSH) was included in an experiment with human liver S9 fraction to trap reactive metabolites generated from cytochrome P450-mediated metabolism of lapatinib and its O-dealkylated metabolite, M1 (question 1). The AO inhibitor hydralazine was included in this experiment to investigate the involvement of AO-mediated metabolism of M1 (question 2). The presence of GSH was found to interfere with the inhibitory activity of hydralazine. Consideration of the time-dependent nature of hydralazine inhibitory activity toward AO when designing this experiment could have predicted the potential for GSH to interfere with hydralazine. This case underscores the importance of clearly identifying the research question, tailoring the experimental protocol to answer that question, and then meticulously considering how the experimental conditions could influence the results, particularly if attempting to address multiple questions with a single experiment.
Collapse
|
8
|
Soltani S, Hallaj-Nezhadi S, Rashidi MR. A comprehensive review of in silico approaches for the prediction and modulation of aldehyde oxidase-mediated drug metabolism: The current features, challenges and future perspectives. Eur J Med Chem 2021; 222:113559. [PMID: 34119831 DOI: 10.1016/j.ejmech.2021.113559] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 01/09/2023]
Abstract
The importance of aldehyde oxidase (AOX) in drug metabolism necessitates the development and application of the in silico rational drug design methods as an integral part of drug discovery projects for the early prediction and modulation of AOX-mediated metabolism. The current study represents an up-to-date and thorough review of in silico studies of AOX-mediated metabolism and modulation methods. In addition, the challenges and the knowledge gap that should be covered have been discussed. The importance of aldehyde oxidase (AOX) in drug metabolism is a hot topic in drug discovery. Different strategies are available for the modulation of the AOX-mediated metabolism of drugs. Application of the rational drug design methods as an integral part of drug discovery projects is necessary for the early prediction of AOX-mediated metabolism. The current study represents a comprehensive review of AOX molecular structure, AOX-mediated reactions, AOX substrates, AOX inhibition, approaches to modify AOX-mediated metabolism, prediction of AOX metabolism/substrates/inhibitors, and the AOX related structure-activity relationship (SAR) studies. Furthermore, an up-to-date and thorough review of in silico studies of AOX metabolism has been carried out. In addition, the challenges and the knowledge gap that should be covered in the scientific literature have been discussed in the current review.
Collapse
Affiliation(s)
- Somaieh Soltani
- Pharmaceutical Analysis Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Somayeh Hallaj-Nezhadi
- Drug Applied Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Stem Cell and Regenerative Medicine Institute and Pharmacy faculty, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
9
|
Paragas EM, Choughule K, Jones JP, Barr JT. Enzyme Kinetics, Pharmacokinetics, and Inhibition of Aldehyde Oxidase. Methods Mol Biol 2021; 2342:257-284. [PMID: 34272698 DOI: 10.1007/978-1-0716-1554-6_10] [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] [Indexed: 06/13/2023]
Abstract
Aldehyde oxidase (AO) has emerged as an important drug metabolizing enzyme over the last decade. Several compounds have failed in the clinic because the clearance or toxicity was underestimated by preclinical species. Human AO is much more active than rodent AO, and dogs do not have functional AO. Metabolic products from AO-catalyzed oxidation are generally nonreactive and often they have much lower solubility. AO metabolism is not limited to oxidation as AO can also catalyze reduction of oxygen and nitrite. Reduction of oxygen leads to the reactive oxygen species (ROS) superoxide radical anion and hydrogen peroxide. Reduction of nitrite leads to the formation of nitric oxide with potential pharmacological implications. AO is also reported to catalyze the reductive metabolism of nitro-compounds, N-oxides, sulfoxides, isoxazoles, isothiazoles, nitrite, and hydroxamic acids. These reductive transformations may cause toxicity due to the formation of reactive metabolites. Moreover, the inhibition kinetics are complex, and multiple probe substrates should be used when assessing the potential for DDIs. Finally, AO appears to be amenable to computational predictions of both regioselectivity and rates of reaction, which holds promise for virtual screening.
Collapse
Affiliation(s)
- Erickson M Paragas
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Kanika Choughule
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, Boston, MA, USA
| | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - John T Barr
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck, South San Francisco, CA, USA.
| |
Collapse
|
10
|
Abbasi A, Joswig-Jones CA, Jones JP. Site-Directed Mutagenesis at the Molybdenum Pterin Cofactor Site of the Human Aldehyde Oxidase: Interrogating the Kinetic Differences Between Human and Cynomolgus Monkey. Drug Metab Dispos 2020; 48:1364-1371. [PMID: 33020066 DOI: 10.1124/dmd.120.000187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022] Open
Abstract
The estimation of the drug clearance by aldehyde oxidase (AO) has been complicated because of this enzyme's atypical kinetics and species and substrate specificity. Since human AO (hAO) and cynomolgus monkey AO (mAO) have a 95.1% sequence identity, cynomolgus monkeys may be the best species for estimating AO clearance in humans. Here, O6-benzylguanine (O6BG) and dantrolene were used under anaerobic conditions, as oxidative and reductive substrates of AO, respectively, to compare and contrast the kinetics of these two species through numerical modeling. Whereas dantrolene reduction followed the same linear kinetics in both species, the oxidation rate of O6BG was also linear in mAO and did not follow the already established biphasic kinetics of hAO. In an attempt to determine why hAO and mAO are kinetically distinct, we have altered the hAO V811 and F885 amino acids at the oxidation site adjacent to the molybdenum pterin cofactor to the corresponding alanine and leucine in mAO, respectively. Although some shift to a more monkey-like kinetics was observed for the V811A mutant, five more mutations around the AO cofactors still need to be investigated for this purpose. In comparing the oxidative and reductive rates of metabolism under anaerobic conditions, we have come to the conclusion that despite having similar rates of reduction (4-fold difference), the oxidation rate in mAO is more than 50-fold slower than hAO. This finding implies that the presence of nonlinearity in AO kinetics is dependent upon the degree of imbalance between the rates of oxidation and reduction in this enzyme. SIGNIFICANCE STATEMENT: Although they have as much as 95.1% sequence identity, human and cynomolgus monkey aldehyde oxidase are kinetically distinct. Therefore, monkeys may not be good estimators of drug clearance in humans.
Collapse
Affiliation(s)
- Armina Abbasi
- Department of Chemistry, Washington State University, Pullman, Washington
| | | | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, Washington
| |
Collapse
|
11
|
Dang NL, Matlock MK, Hughes TB, Swamidass SJ. The Metabolic Rainbow: Deep Learning Phase I Metabolism in Five Colors. J Chem Inf Model 2020; 60:1146-1164. [DOI: 10.1021/acs.jcim.9b00836] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Na Le Dang
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Matthew K. Matlock
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Tyler B. Hughes
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - S. Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| |
Collapse
|
12
|
Manevski N, King L, Pitt WR, Lecomte F, Toselli F. Metabolism by Aldehyde Oxidase: Drug Design and Complementary Approaches to Challenges in Drug Discovery. J Med Chem 2019; 62:10955-10994. [PMID: 31385704 DOI: 10.1021/acs.jmedchem.9b00875] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aldehyde oxidase (AO) catalyzes oxidations of azaheterocycles and aldehydes, amide hydrolysis, and diverse reductions. AO substrates are rare among marketed drugs, and many candidates failed due to poor pharmacokinetics, interspecies differences, and adverse effects. As most issues arise from complex and poorly understood AO biology, an effective solution is to stop or decrease AO metabolism. This perspective focuses on rational drug design approaches to modulate AO-mediated metabolism in drug discovery. AO biological aspects are also covered, as they are complementary to chemical design and important when selecting the experimental system for risk assessment. The authors' recommendation is an early consideration of AO-mediated metabolism supported by computational and in vitro experimental methods but not an automatic avoidance of AO structural flags, many of which are versatile and valuable building blocks. Preferably, consideration of AO-mediated metabolism should be part of the multiparametric drug optimization process, with the goal to improve overall drug-like properties.
Collapse
Affiliation(s)
- Nenad Manevski
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Lloyd King
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - William R Pitt
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Fabien Lecomte
- UCB Celltech , 208 Bath Road , Slough SL13WE , United Kingdom
| | - Francesca Toselli
- UCB BioPharma , Chemin du Foriest 1 , 1420 Braine-l'Alleud , Belgium
| |
Collapse
|
13
|
Cheshmazar N, Dastmalchi S, Terao M, Garattini E, Hamzeh-Mivehroud M. Aldehyde oxidase at the crossroad of metabolism and preclinical screening. Drug Metab Rev 2019; 51:428-452. [DOI: 10.1080/03602532.2019.1667379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Narges Cheshmazar
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mineko Terao
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Enrico Garattini
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
15
|
Chen S, Austin-Muttitt K, Zhang LH, Mullins JGL, Lau AJ. In Vitro and In Silico Analyses of the Inhibition of Human Aldehyde Oxidase by Bazedoxifene, Lasofoxifene, and Structural Analogues. J Pharmacol Exp Ther 2019; 371:75-86. [DOI: 10.1124/jpet.119.259267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/05/2019] [Indexed: 11/22/2022] Open
|
16
|
Deris-Abdolahpour F, Abdolalipouran-Sadegh L, Dastmalchi S, Hamzeh-Mivehroud M, Zarei O, Dehgan G, Rashidi MR. Effects of Phenothiazines on Aldehyde Oxidase Activity Towards Aldehydes and N-Heterocycles: an In Vitro and In Silico Study. Eur J Drug Metab Pharmacokinet 2019; 44:275-286. [PMID: 30382490 DOI: 10.1007/s13318-018-0514-6] [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] [Indexed: 11/27/2022]
Abstract
BACKGROUND Aldehyde oxidase (AOX) is an important molybdenum-containing enzyme with high similarity with xanthine oxidase (XO). AOX involved in the metabolism of a large array of aldehydes and N-heterocyclic compounds and its activity is highly substrate-dependent. OBJECTIVES The aim of this work was to study the effect of five important phenothiazine drugs on AOX activity using benzaldehyde and phenanthridine as aldehyde and N-heterocyclic substrates, respectively. METHODS The effect of trifluperazine, chlorpromazine, perphenazine, thioridazine and promethazine on rat liver AOX was measured spectrophotometrically. To predict the mode of interactions between the studied compounds and AOX, a combination of homology modeling and a molecular docking study was performed. RESULTS All phenothiazines could inhibit AOX activity measured either by phenanthridine or benzaldehyde with almost no effect on XO activity. In the case of benzaldehyde oxidation, the lowest and highest half-maximal inhibitory concentration (IC50) values were obtained for promethazine (IC50 = 0.9 µM), and trifluoperazine (IC50 = 3.9 µM), respectively; whereas perphenazine (IC50 = 4.3 µM), and trifluoperazine (IC50 = 49.6 µM) showed the strongest and weakest inhibitory activity against AOX-catalyzed phenanthridine oxidation, respectively. The in silico findings revealed that the binding site of thioridazine is near the dimer interference, and that hydrophobic interactions are of great importance in all the tested phenothiazines. CONCLUSION The five studied phenothiazine drugs showed dual inhibitory effects on AOX activity towards aldehydes and N-heterocycles as two major classes of enzyme substrates. Most of the interactions between the phenothiazine-related drugs and AOX in the binding pocket showed a hydrophobic nature.
Collapse
Affiliation(s)
| | | | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Zarei
- Neurosciences Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Gholamreza Dehgan
- Department of Zoology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, 51664-14766, Iran.
| |
Collapse
|
17
|
Fast Methods for Prediction of Aldehyde Oxidase-Mediated Site-of-Metabolism. Comput Struct Biotechnol J 2019; 17:345-351. [PMID: 30949305 PMCID: PMC6429535 DOI: 10.1016/j.csbj.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022] Open
Abstract
Aldehyde Oxidase (AO) is an enzyme involved in the metabolism of aldehydes and N-containing heterocyclic compounds. Many drug compounds contain heterocyclic moieties, and AO metabolism has lead to failure of several late-stage drug candidates. Therefore, it is important to take AO-mediated metabolism into account early in the drug discovery process, and thus, to have fast and reliable models to predict the site of metabolism (SOM). We have collected a dataset of 78 substrates of human AO with a total of 89 SOMs and 347 non-SOMs and determined atomic descriptors for each compound. The descriptors comprise NMR shielding and ESP charges from density functional theory (DFT), NMR chemical shift from ChemBioDraw, and Gasteiger charges from RDKit. Additionally, atomic accessibility was considered using 2D-SASA and relative span descriptors from SMARTCyp. Finally, stability of the product, the metabolite, was determined with DFT and also used as a descriptor. All descriptors have AUC larger than 0.75. In particular, descriptors related to the chemical shielding and chemical shift (AUC = 0.96) and ESP charges (AUC = 0.96) proved to be good descriptors. We recommend two simple methods to identify the SOM for a given molecule: 1) use ChemBioDraw to calculate the chemical shift or 2) calculate ESP charges or chemical shift using DFT. The first approach is fast but somewhat difficult to automate, while the second is more time-consuming, but can easily be automated. The two methods predict correctly 93% and 91%, respectively, of the 89 experimentally observed SOMs.
Collapse
|
18
|
Abbasi A, Paragas EM, Joswig-Jones CA, Rodgers JT, Jones JP. Time Course of Aldehyde Oxidase and Why It Is Nonlinear. Drug Metab Dispos 2019; 47:473-483. [PMID: 30787100 DOI: 10.1124/dmd.118.085787] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
Abstract
Many promising drug candidates metabolized by aldehyde oxidase (AOX) fail during clinical trial owing to underestimation of their clearance. AOX is species-specific, which makes traditional allometric studies a poor choice for estimating human clearance. Other studies have suggested using half-life calculated by measuring substrate depletion to measure clearance. In this study, we proposed using numerical fitting to enzymatic pathways other than Michaelis-Menten (MM) to avoid missing the initial high turnover rate of product formation. Here, product formation over a 240-minute time course of six AOX substrates-O6-benzylguanine, N-(2-dimethylamino)ethyl)acridine-4-carboxamide, zaleplon, phthalazine, BIBX1382 [N8-(3-Chloro-4-fluorophenyl)-N2-(1-methyl-4-piperidinyl)-pyrimido[5,4-d]pyrimidine-2,8-diamine dihydrochloride], and zoniporide-have been provided to illustrate enzyme deactivation over time to help better understand why MM kinetics sometimes leads to underestimation of rate constants. Based on the data provided in this article, the total velocity for substrates becomes slower than the initial velocity by 3.1-, 6.5-, 2.9-, 32.2-, 2.7-, and 0.2-fold, respectively, in human expressed purified enzyme, whereas the K m remains constant. Also, our studies on the role of reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, show that ROS did not significantly alter the change in enzyme activity over time. Providing a new electron acceptor, 5-nitroquinoline, did, however, alter the change in rate over time for mumerous compounds. The data also illustrate the difficulties in using substrate disappearance to estimate intrinsic clearance.
Collapse
Affiliation(s)
- Armina Abbasi
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Erickson M Paragas
- Department of Chemistry, Washington State University, Pullman, Washington
| | | | - John T Rodgers
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, Washington
| |
Collapse
|
19
|
Takaoka N, Sanoh S, Okuda K, Kotake Y, Sugahara G, Yanagi A, Ishida Y, Tateno C, Tayama Y, Sugihara K, Kitamura S, Kurosaki M, Terao M, Garattini E, Ohta S. Inhibitory effects of drugs on the metabolic activity of mouse and human aldehyde oxidases and influence on drug–drug interactions. Biochem Pharmacol 2018; 154:28-38. [DOI: 10.1016/j.bcp.2018.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
|
20
|
Le Dang N, Hughes TB, Miller GP, Swamidass SJ. Computationally Assessing the Bioactivation of Drugs by N-Dealkylation. Chem Res Toxicol 2018; 31:68-80. [PMID: 29355304 PMCID: PMC5871345 DOI: 10.1021/acs.chemrestox.7b00191] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cytochromes P450 (CYPs) oxidize alkylated amines commonly found in drugs and other biologically active molecules, cleaving them into an amine and an aldehyde. Metabolic studies usually neglect to report or investigate aldehydes, even though they can be toxic. It is assumed that they are efficiently detoxified into carboxylic acids and alcohols. Nevertheless, some aldehydes are reactive and escape detoxification pathways to cause adverse events by forming DNA and protein adducts. Herein, we modeled N-dealkylations that produce both amine and aldehyde metabolites and then predicted the reactivity of the aldehyde. This model used a deep learning approach previously developed by our group to predict other types of drug metabolism. In this study, we trained the model to predict N-dealkylation by human liver microsomes (HLM), finding that including isozyme-specific metabolism data alongside HLM data significantly improved results. The final HLM model accurately predicted the site of N-dealkylation within metabolized substrates (97% top-two and 94% area under the ROC curve). Next, we combined the metabolism, metabolite structure prediction, and previously published reactivity models into a bioactivation model. This combined model predicted the structure of the most likely reactive metabolite of a small validation set of drug-like molecules known to be bioactivated by N-dealkylation. Applying this model to approved and withdrawn medicines, we found that aldehyde metabolites produced from N-dealkylation may explain the hepatotoxicity of several drugs: indinavir, piperacillin, verapamil, and ziprasidone. Our results suggest that N-dealkylation may be an under-appreciated bioactivation pathway, especially in clinical contexts where aldehyde detoxification pathways are inhibited. Moreover, this is the first report of a bioactivation model constructed by combining a metabolism and reactivity model. These results raise hope that more comprehensive models of bioactivation are possible. The model developed in this study is available at http://swami.wustl.edu/xenosite/ .
Collapse
Affiliation(s)
- Na Le Dang
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Tyler B. Hughes
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| | - Grover P. Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - S. Joshua Swamidass
- Department of Pathology and Immunology, Washington University School of Medicine, Campus Box 8118, 660 S. Euclid Ave., St. Louis, Missouri 63110, United States
| |
Collapse
|
21
|
Paragas E, Humphreys SC, Min J, Joswig-Jones CA, Leimkühler S, Jones JP. ecoAO: A Simple System for the Study of Human Aldehyde Oxidases Role in Drug Metabolism. ACS OMEGA 2017; 2:4820-4827. [PMID: 28884164 PMCID: PMC5579547 DOI: 10.1021/acsomega.7b01054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/09/2017] [Indexed: 06/07/2023]
Abstract
Although aldehyde oxidase (AO) is an important hepatic drug-metabolizing enzyme, it remains understudied and is consequently often overlooked in preclinical studies, an oversight that has resulted in the failure of multiple clinical trials. AO's preclusion to investigation stems from the following: (1) difficulties synthesizing metabolic standards due to the chemospecificity and regiospecificity of the enzyme and (2) significant inherent variability across existing in vitro systems including liver cytosol, S9 fractions, and primary hepatocytes, which lack specificity and generate discordant expression and activity profiles. Here, we describe a practical bacterial biotransformation system, ecoAO, addressing both issues simultaneously. ecoAO is a cell paste of MoCo-producing Escherichia coli strain TP1017 expressing human AO. It exhibits specific activity toward known substrates, zoniporide, 4-trans-(N,N-dimethylamino)cinnamaldehyde, O6-benzylguanine, and zaleplon; it also has utility as a biocatalyst, yielding milligram quantities of synthetically challenging metabolite standards such as 2-oxo-zoniporide. Moreover, ecoAO enables routine determination of kcat and V/K, which are essential parameters for accurate in vivo clearance predictions. Furthermore, ecoAO has potential as a preclinical in vitro screening tool for AO activity, as demonstrated by its metabolism of 3-aminoquinoline, a previously uncharacterized substrate. ecoAO promises to provide easy access to metabolites with the potential to improve pharmacokinetic clearance predictions and guide drug development.
Collapse
Affiliation(s)
- Erickson
M. Paragas
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Sara C. Humphreys
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Joshua Min
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Carolyn A. Joswig-Jones
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| | - Silke Leimkühler
- Department
of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476 Potsdam, Germany
| | - Jeffrey P. Jones
- Department
of Chemistry, Washington State University, 99164-4630 Pullman, Washington, United States
| |
Collapse
|
22
|
Montefiori M, Jørgensen FS, Olsen L. Aldehyde Oxidase: Reaction Mechanism and Prediction of Site of Metabolism. ACS OMEGA 2017; 2:4237-4244. [PMID: 30023718 PMCID: PMC6044498 DOI: 10.1021/acsomega.7b00658] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/11/2017] [Indexed: 05/16/2023]
Abstract
Aldehyde oxidase (AO) is a molybdenum-containing enzyme involved in the clearance of drug compounds containing aldehydes and N-containing heterocyclic fragments. AO has gained considerable interest in recent years because of examples of too fast clearance of drug compounds in development. Thus, it is important to be able to predict AO-mediated drug metabolism. Therefore, we have characterized the structural and energetic aspects of different mechanisms with density functional theory using the molybdenum cofactor as a model for the reactive part of the enzyme. For a series of 6-substituted 4-quinazolinones, the trend in activation energies is the same for three tested reaction mechanisms. Using the concerted mechanism as a model for the enzymatic reaction, the transition states (TSs) for the formation of all possible metabolites for a series of known AO substrates were determined. The lowest activation energies correspond in all cases to the experimentally observed sites of metabolism (SOMs). Various molecular properties were calculated and investigated as more easily determinable markers for reactivity. The stabilities of both intermediates and products correlate to some extent with the TS energies and may be used to predict the SOM. The electrostatic-potential-derived charges are also good markers for the prediction of the experimental SOM for this set of compounds and may pave the way for the development of fast methods for the prediction of SOM for AO substrates.
Collapse
|
23
|
Bohnert T, Patel A, Templeton I, Chen Y, Lu C, Lai G, Leung L, Tse S, Einolf HJ, Wang YH, Sinz M, Stearns R, Walsky R, Geng W, Sudsakorn S, Moore D, He L, Wahlstrom J, Keirns J, Narayanan R, Lang D, Yang X. Evaluation of a New Molecular Entity as a Victim of Metabolic Drug-Drug Interactions-an Industry Perspective. ACTA ACUST UNITED AC 2016; 44:1399-423. [PMID: 27052879 DOI: 10.1124/dmd.115.069096] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/31/2016] [Indexed: 12/15/2022]
Abstract
Under the guidance of the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), scientists from 20 pharmaceutical companies formed a Victim Drug-Drug Interactions Working Group. This working group has conducted a review of the literature and the practices of each company on the approaches to clearance pathway identification (fCL), estimation of fractional contribution of metabolizing enzyme toward metabolism (fm), along with modeling and simulation-aided strategy in predicting the victim drug-drug interaction (DDI) liability due to modulation of drug metabolizing enzymes. Presented in this perspective are the recommendations from this working group on: 1) strategic and experimental approaches to identify fCL and fm, 2) whether those assessments may be quantitative for certain enzymes (e.g., cytochrome P450, P450, and limited uridine diphosphoglucuronosyltransferase, UGT enzymes) or qualitative (for most of other drug metabolism enzymes), and the impact due to the lack of quantitative information on the latter. Multiple decision trees are presented with stepwise approaches to identify specific enzymes that are involved in the metabolism of a given drug and to aid the prediction and risk assessment of drug as a victim in DDI. Modeling and simulation approaches are also discussed to better predict DDI risk in humans. Variability and parameter sensitivity analysis were emphasized when applying modeling and simulation to capture the differences within the population used and to characterize the parameters that have the most influence on the prediction outcome.
Collapse
Affiliation(s)
- Tonika Bohnert
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Aarti Patel
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ian Templeton
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Yuan Chen
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Chuang Lu
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - George Lai
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Louis Leung
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Susanna Tse
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Heidi J Einolf
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ying-Hong Wang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Michael Sinz
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ralph Stearns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Robert Walsky
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Wanping Geng
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Sirimas Sudsakorn
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - David Moore
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Ling He
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jan Wahlstrom
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Jim Keirns
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Rangaraj Narayanan
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Dieter Lang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | - Xiaoqing Yang
- Biogen, Cambridge, Massachusetts (T.B.); GlaxoSmithKline R&D, Hertfordshire, United Kingdom (A.P.); Janssen R&D, Spring House, Pennsylvania (I.T.); Genentech, South San Francisco, California (Y.C.); Takeda, Cambridge, Massachusetts (C.L.); Eisai Inc., Andover, Massachusetts (G.L.); Pfizer Inc., Groton, Connecticut (L.L., S.T.); Novartis, East Hanover, New Jersey (H.J.E.); Merck & Co., Inc., Kenilworth, New Jersey (Y.-H.W.); Bristol Myers Squibb, Wallingford, Connecticut (M.S.); Vertex Pharmaceuticals Inc., Boston, Massachusetts (R.S.); EMD Serono R&D Institute, Inc., Billerica, Massachusetts (R.W., W.G.); Sanofi, Waltham, Massachusetts (S.S.); Roche Innovation Center, New York, New York (D.M.); Daiichi Sankyo, Edison, New Jersey (L.H.); Amgen Inc., Thousand Oaks, California (J.W.); Astellas, Northbrook, Illinois (J.K.); Celgene Corporation, Summit, New Jersey (R.N.); Bayer Pharma AG, Wuppertal, Germany (D.L.); and Incyte Corporation, Wilmington, Delaware (X.Y.)
| | | |
Collapse
|
24
|
Rosa M, Bonnaillie P, Chanteux H. Prediction of drug–drug interactions with carbamazepine-10,11-epoxide using a new in vitro assay for epoxide hydrolase inhibition. Xenobiotica 2016; 46:1076-1084. [DOI: 10.3109/00498254.2016.1151088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Maria Rosa
- UCB Biopharma SPRL, Non-Clinical Development, Braine-L’alleud, Belgium
| | - Pierre Bonnaillie
- UCB Biopharma SPRL, Non-Clinical Development, Braine-L’alleud, Belgium
| | - Hugues Chanteux
- UCB Biopharma SPRL, Non-Clinical Development, Braine-L’alleud, Belgium
| |
Collapse
|
25
|
Interspecies differences in the metabolism of methotrexate: An insight into the active site differences between human and rabbit aldehyde oxidase. Biochem Pharmacol 2015; 96:288-95. [PMID: 26032640 DOI: 10.1016/j.bcp.2015.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/21/2015] [Indexed: 12/27/2022]
Abstract
Several drug compounds have failed in clinical trials due to extensive biotransformation by aldehyde oxidase (AOX) (EC 1.2.3.1). One of the main reasons is the difficulty in scaling clearance for drugs metabolised by AOX, from preclinical species to human. Using methotrexate as a probe substrate, we evaluated AOX metabolism in liver cytosol from human and commonly used laboratory species namely guinea pig, monkey, rat and rabbit. We found that the metabolism of methotrexate in rabbit liver cytosol was several orders of magnitude higher than any of the other species tested. The results of protein quantitation revealed that the amount of AOX1 in human liver was similar to rabbit liver. To understand if the observed differences in activity were due to structural differences, we modelled rabbit AOX1 using the previously generated human AOX1 homology model. Molecular docking of methotrexate into the active site of the enzyme led to the identification of important residues that could potentially be involved in substrate binding and account for the observed differences. In order to study the impact of these residue changes on enzyme activity, we used site directed mutagenesis to construct mutant AOX1 cDNAs by substituting nucleotides of human AOX1 with relevant ones of rabbit AOX1. AOX1 mutant proteins were expressed in Escherichia coli. Differences in the kinetic properties of these mutants have been presented in this study.
Collapse
|
26
|
Sodhi JK, Wong S, Kirkpatrick DS, Liu L, Khojasteh SC, Hop CECA, Barr JT, Jones JP, Halladay JS. A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos 2015; 43:908-15. [PMID: 25845827 DOI: 10.1124/dmd.114.061804] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/06/2015] [Indexed: 12/31/2022] Open
Abstract
GDC-0834, a Bruton's tyrosine kinase inhibitor investigated as a potential treatment of rheumatoid arthritis, was previously reported to be extensively metabolized by amide hydrolysis such that no measurable levels of this compound were detected in human circulation after oral administration. In vitro studies in human liver cytosol determined that GDC-0834 (R)-N-(3-(6-(4-(1,4-dimethyl-3-oxopiperazin-2-yl)phenylamino)-4-methyl-5-oxo- 4,5-dihydropyrazin-2-yl)-2-methylphenyl)-4,5,6,7-tetrahydrobenzo[b] thiophene-2-carboxamide) was rapidly hydrolyzed with a CLint of 0.511 ml/min per milligram of protein. Aldehyde oxidase (AO) and carboxylesterase (CES) were putatively identified as the enzymes responsible after cytosolic fractionation and mass spectrometry-proteomics analysis of the enzymatically active fractions. Results were confirmed by a series of kinetic experiments with inhibitors of AO, CES, and xanthine oxidase (XO), which implicated AO and CES, but not XO, as mediating GDC-0834 amide hydrolysis. Further supporting the interaction between GDC-0834 and AO, GDC-0834 was shown to be a potent reversible inhibitor of six known AO substrates with IC50 values ranging from 0.86 to 1.87 μM. Additionally, in silico modeling studies suggest that GDC-0834 is capable of binding in the active site of AO with the amide bond of GDC-0834 near the molybdenum cofactor (MoCo), orientated in such a way to enable potential nucleophilic attack on the carbonyl of the amide bond by the hydroxyl of MoCo. Together, the in vitro and in silico results suggest the involvement of AO in the amide hydrolysis of GDC-0834.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Susan Wong
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Donald S Kirkpatrick
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Lichuan Liu
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - S Cyrus Khojasteh
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Cornelis E C A Hop
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - John T Barr
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Jeffrey P Jones
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| | - Jason S Halladay
- Departments of Drug Metabolism and Pharmacokinetics (J.K.S., S.W., S.C.K., C.E.C.A.H., J.S.H.), Clinical Pharmacology (L.L.), and Protein Chemistry (D.S.K.), Genentech, Inc., South San Francisco, California; and Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., J.P.J.)
| |
Collapse
|
27
|
Sanoh S, Tayama Y, Sugihara K, Kitamura S, Ohta S. Significance of aldehyde oxidase during drug development: Effects on drug metabolism, pharmacokinetics, toxicity, and efficacy. Drug Metab Pharmacokinet 2015; 30:52-63. [DOI: 10.1016/j.dmpk.2014.10.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 12/28/2022]
|
28
|
Barr JT, Jones JP, Oberlies NH, Paine MF. Inhibition of human aldehyde oxidase activity by diet-derived constituents: structural influence, enzyme-ligand interactions, and clinical relevance. Drug Metab Dispos 2014; 43:34-41. [PMID: 25326286 DOI: 10.1124/dmd.114.061192] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The mechanistic understanding of interactions between diet-derived substances and conventional medications in humans is nascent. Most investigations have examined cytochrome P450-mediated interactions. Interactions mediated by other phase I enzymes are understudied. Aldehyde oxidase (AO) is a phase I hydroxylase that is gaining recognition in drug design and development programs. Taken together, a panel of structurally diverse phytoconstituents (n = 24) was screened for inhibitors of the AO-mediated oxidation of the probe substrate O(6)-benzylguanine. Based on the estimated IC50 (<100 μM), 17 constituents were advanced for Ki determination. Three constituents were described best by a competitive inhibition model, whereas 14 constituents were described best by a mixed-mode model. The latter model consists of two Ki terms, Kis and Kii, which ranged from 0.26-73 and 0.80-120 μM, respectively. Molecular modeling was used to glean mechanistic insight into AO inhibition. Docking studies indicated that the tested constituents bound within the AO active site and elucidated key enzyme-inhibitor interactions. Quantitative structure-activity relationship modeling identified three structural descriptors that correlated with inhibition potency (r(2) = 0.85), providing a framework for developing in silico models to predict the AO inhibitory activity of a xenobiotic based solely on chemical structure. Finally, a simple static model was used to assess potential clinically relevant AO-mediated dietary substance-drug interactions. Epicatechin gallate and epigallocatechin gallate, prominent constituents in green tea, were predicted to have moderate to high risk. Further characterization of this uncharted type of interaction is warranted, including dynamic modeling and, potentially, clinical evaluation.
Collapse
Affiliation(s)
- John T Barr
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (J.T.B., M.F.P.); Department of Chemistry, Washington State University, Pullman, Washington (J.P.J.); and Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.H.O.)
| | - Jeffrey P Jones
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (J.T.B., M.F.P.); Department of Chemistry, Washington State University, Pullman, Washington (J.P.J.); and Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.H.O.)
| | - Nicholas H Oberlies
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (J.T.B., M.F.P.); Department of Chemistry, Washington State University, Pullman, Washington (J.P.J.); and Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.H.O.)
| | - Mary F Paine
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (J.T.B., M.F.P.); Department of Chemistry, Washington State University, Pullman, Washington (J.P.J.); and Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.H.O.)
| |
Collapse
|
29
|
Choughule KV, Barnaba C, Joswig-Jones CA, Jones JP. In vitro oxidative metabolism of 6-mercaptopurine in human liver: insights into the role of the molybdoflavoenzymes aldehyde oxidase, xanthine oxidase, and xanthine dehydrogenase. Drug Metab Dispos 2014; 42:1334-40. [PMID: 24824603 DOI: 10.1124/dmd.114.058107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Anticancer agent 6-mercaptopurine (6MP) has been in use since 1953 for the treatment of childhood acute lymphoblastic leukemia (ALL) and inflammatory bowel disease. Despite being available for 60 years, several aspects of 6MP drug metabolism and pharmacokinetics in humans are unknown. Molybdoflavoenzymes such as aldehyde oxidase (AO) and xanthine oxidase (XO) have previously been implicated in the metabolism of this drug. In this study, we investigated the in vitro metabolism of 6MP to 6-thiouric acid (6TUA) in pooled human liver cytosol. We discovered that 6MP is metabolized to 6TUA through sequential metabolism via the 6-thioxanthine (6TX) intermediate. The role of human AO and XO in the metabolism of 6MP was established using the specific inhibitors raloxifene and febuxostat. Both AO and XO were involved in the metabolism of the 6TX intermediate, whereas only XO was responsible for the conversion of 6TX to 6TUA. These findings were further confirmed using purified human AO and Escherichia coli lysate containing expressed recombinant human XO. Xanthine dehydrogenase (XDH), which belongs to the family of xanthine oxidoreductases and preferentially reduces nicotinamide adenine dinucleotide (NAD(+)), was shown to contribute to the overall production of the 6TX intermediate as well as the final product 6TUA in the presence of NAD(+) in human liver cytosol. In conclusion, we present evidence that three enzymes, AO, XO, and XDH, contribute to the production of 6TX intermediate, whereas only XO and XDH are involved in the conversion of 6TX to 6TUA in pooled HLC.
Collapse
Affiliation(s)
- Kanika V Choughule
- Department of Chemistry, Washington State University, Pullman, Washington
| | - Carlo Barnaba
- Department of Chemistry, Washington State University, Pullman, Washington
| | | | - Jeffrey P Jones
- Department of Chemistry, Washington State University, Pullman, Washington
| |
Collapse
|
30
|
Barr JT, Choughule KV, Nepal S, Wong T, Chaudhry AS, Joswig-Jones CA, Zientek M, Strom SC, Schuetz EG, Thummel KE, Jones JP. Why do most human liver cytosol preparations lack xanthine oxidase activity? Drug Metab Dispos 2014; 42:695-9. [PMID: 24430612 DOI: 10.1124/dmd.113.056374] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
When investigating the potential for xanthine oxidase (XO)-mediated metabolism of a new chemical entity in vitro, selective chemical inhibition experiments are typically used. Most commonly, these inhibition experiments are performed using the inhibitor allopurinol (AP) and commercially prepared human liver cytosol (HLC) as the enzyme source. For reasons detailed herein, it is also a common practice to perfuse livers with solutions containing AP prior to liver harvest. The exposure to AP in HLC preparations could obviously pose a problem for measuring in vitro XO activity. To investigate this potential problem, an HPLC-MS/MS assay was developed to determine whether AP and its primary metabolite, oxypurinol, are retained within the cytosol for livers that were treated with AP during liver harvest. Differences in enzymatic activity for XO and aldehyde oxidase (AO) in human cytosol that can be ascribed to AP exposure were also evaluated. The results confirmed the presence of residual AP (some) and oxypurinol (all) human liver cytosol preparations that had been perfused with an AP-containing solution. In every case where oxypurinol was detected, XO activity was not observed. In contrast, the presence of AP and oxypurinol did not appear to have an impact on AO activity. Pooled HLC that was purchased from a commercial source also contained residual oxypurinol and did not show any XO activity. In the future, it is recommended that each HLC batch is screened for oxypurinol and/or XO activity prior to testing for XO-mediated metabolism of a new chemical entity.
Collapse
Affiliation(s)
- John T Barr
- Department of Chemistry, Washington State University, Pullman, Washington (J.T.B., K.V.C., S.N., C.A.J.-J., J.P.J.); Department of Pharmaceutics, University of Washington, Seattle, Washington (K.E.T., T.W.); Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (A.S.C., E.G.S.); Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden (S.C.S.); and Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Pfizer Inc., La Jolla, California (M.Z.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Weidert ER, Schoenborn SO, Cantu-Medellin N, Choughule KV, Jones JP, Kelley EE. Inhibition of xanthine oxidase by the aldehyde oxidase inhibitor raloxifene: implications for identifying molybdopterin nitrite reductases. Nitric Oxide 2014; 37:41-5. [PMID: 24406683 DOI: 10.1016/j.niox.2013.12.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/18/2013] [Accepted: 12/27/2013] [Indexed: 01/10/2023]
Abstract
Sources of nitric oxide alternative to nitric oxide synthases are gaining significant traction as crucial mediators of vessel function under hypoxic inflammatory conditions. For example, capacity to catalyze the one electron reduction of nitrite (NO2-) to ·NO has been reported for hemoglobin, myoglobin and molybdopterin-containing enzymes including xanthine oxidoreductase (XOR) and aldehyde oxidase (AO). For XOR and AO, use of selective inhibition strategies is therefore crucial when attempting to assign relative contributions to nitrite-mediated ·NO formation in cells and tissue. To this end, XOR inhibition has been accomplished with application of classic pyrazolopyrimidine-based inhibitors allo/oxypurinol or the newly FDA-approved XOR-specific inhibitor, Uloric® (febuxostat). Likewise, raloxifene, an estrogen receptor antagonist, has been identified as a potent (Ki=1.0 nM) inhibitor of AO. Herein, we characterize the inhibition kinetics of raloxifene for XOR and describe the resultant effects on inhibiting XO-catalyzed ·NO formation. Exposure of purified XO to raloxifene (PBS, pH 7.4) resulted in a dose-dependent (12.5-100 μM) inhibition of xanthine oxidation to uric acid. Dixon plot analysis revealed a competitive inhibition process with a Ki=13 μM. This inhibitory process was more effective under acidic pH; similar to values encountered under hypoxic/inflammatory conditions. In addition, raloxifene also inhibited anoxic XO-catalyzed reduction of NO2- to NO (EC50=64 μM). In contrast to having no effect on XO-catalyzed uric acid production, the AO inhibitor menadione demonstrated potent inhibition of XO-catalyzed NO2- reduction (EC50=60 nM); somewhat similar to the XO-specific inhibitor, febuxostat (EC50=4 nM). Importantly, febuxostat was found to be a very poor inhibitor of human AO (EC50=613 μM) suggesting its usefulness for validating XO-dependent contributions to NO2- reduction in biological systems. Combined, these data indicate care should be taken when choosing inhibition strategies as well as inhibitor concentrations when assigning relative NO2- reductase activity of AO and XOR.
Collapse
Affiliation(s)
- E R Weidert
- University of Pittsburgh School of Medicine, Department of Anesthesiology, United States
| | - S O Schoenborn
- University of Pittsburgh School of Medicine, Department of Anesthesiology, United States
| | - N Cantu-Medellin
- University of Pittsburgh School of Medicine, Department of Anesthesiology, United States; University of Pittsburgh School of Medicine, Vascular Medicine Institute, United States
| | - K V Choughule
- Washington State University, Department of Chemistry, United States
| | - J P Jones
- Washington State University, Department of Chemistry, United States
| | - E E Kelley
- University of Pittsburgh School of Medicine, Department of Anesthesiology, United States; University of Pittsburgh School of Medicine, Vascular Medicine Institute, United States.
| |
Collapse
|
32
|
Barr JT, Choughule K, Jones JP. Enzyme kinetics, inhibition, and regioselectivity of aldehyde oxidase. Methods Mol Biol 2014; 1113:167-186. [PMID: 24523113 DOI: 10.1007/978-1-62703-758-7_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The aldehyde oxidase (AO) enzyme family plays an increasing role in drug development. However, a number of compounds that are AO substrates have failed in the clinic because the clearance or toxicity is underestimated by preclinical species. Human AO is much more active than rodent AO, and dogs do not have functional AO. While AOs normally make non-reactive metabolites such as lactams, the metabolic products often have much lower solubility that can lead to renal failure. While an endogenous substrate for the oxidation reaction is not known, electron acceptors for the reductive part of the reaction include oxygen and nitrites. Reduction of oxygen leads to the reactive oxygen species (ROS) superoxide radical anion, and hydrogen peroxide. Reduction of nitrite leads to the formation of nitric oxide with potential pharmacological implications. To date, no clinically important drug-drug interactions (DDIs) have been observed for AOs. However, the inhibition kinetics are complex, and multiple probe substrates should be used when assessing the potential for DDIs. Finally, AO appears to be amenable to computational predictions of both regioselectivity and rates of reaction, which holds promise for virtual screening.
Collapse
Affiliation(s)
- John T Barr
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | | | | |
Collapse
|
33
|
Barr JT, Jones JP, Joswig-Jones CA, Rock DA. Absolute quantification of aldehyde oxidase protein in human liver using liquid chromatography-tandem mass spectrometry. Mol Pharm 2013; 10:3842-9. [PMID: 24006961 DOI: 10.1021/mp4003046] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The function of the enzyme human aldehyde oxidase (AOX1) is uncertain; however, recent studies have implicated significant biochemical involvement in humans. AOX1 has also rapidly become an important drug-metabolizing enzyme. Until now, quantitation of AOX1 in complex matrices such as tissue has not been achieved. Herein, we developed and employed a trypsin digest and subsequent liquid chromatography-tandem mass spectrometry analysis to determine absolute amounts of AOX1 in human liver. E. coli expressed human purified AOX1 was used to validate the linearity, sensitivity, and selectivity of the method. Overall, the method is highly efficient and sensitive for determination of AOX1 in cytosolic liver fractions. Using this method, we observed substantial batch-to-batch variation in AOX1 content (21-40 pmol AOX1/mg total protein) between various pooled human liver cytosol preparations. We also observed interbatch variation in Vmax (3.3-4.9 nmol min(-1) mg(-1)) and a modest correlation between enzyme concentration and activity. In addition, we measured a large difference in kcat/Km, between purified (kcat/Km of 1.4) and human liver cytosol (kcat/Km of 15-20) indicating cytosol to be 11-14 times more efficient in the turnover of DACA than the E. coli expressed purified enzyme. Finally, we discussed the future impact of this method for the development of drug metabolism models and understanding the biochemical role of this enzyme.
Collapse
Affiliation(s)
- John T Barr
- Department of Chemistry, Washington State University , P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | | | | | | |
Collapse
|
34
|
Choughule KV, Barr JT, Jones JP. Evaluation of rhesus monkey and guinea pig hepatic cytosol fractions as models for human aldehyde oxidase. Drug Metab Dispos 2013; 41:1852-8. [PMID: 23918666 DOI: 10.1124/dmd.113.052985] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Aldehyde oxidase (AOX) is a cytosolic enzyme expressed across a wide range of species, including guinea pig and rhesus monkey. These species are believed to be the best preclinical models for studying human AOX-mediated metabolism. We compared AOX activity in rhesus monkeys, guinea pigs, and humans using phthalazine and N-[2-(dimethylamino)ethyl]acridone-4-carboxamide (DACA) as substrates and raloxifene as an inhibitor. Michaelis-Menten kinetics was observed for phthalazine oxidation in rhesus monkey, guinea pig, and human liver cytosol, whereas substrate inhibition was seen with DACA oxidase activity in all three livers. Raloxifene inhibited phthalazine and DACA oxidase activity uncompetitively in guinea pig, whereas mixed-mode inhibition was seen in rhesus monkey. Our analysis of the primary sequence alignment of rhesus monkey, guinea pig, and human aldehyde oxidase isoform 1 (AOX1) along with homology modeling has led to the identification of several amino acid residue differences within the active site and substrate entrance channel of AOX1. We speculate that some of these residues might be responsible for the differences observed in activity. Overall, our data indicate that rhesus monkeys and guinea pigs would overestimate intrinsic clearance in humans and would be unsuitable to use as animal models. Our study also showed that AOX metabolism in species is substrate-dependent and no single animal model can be reliably used to predict every drug response in humans.
Collapse
Affiliation(s)
- Kanika V Choughule
- Department of Chemistry, Washington State University, Pullman, Washington
| | | | | |
Collapse
|