1
|
Zhu SX. Human absorption, distribution, metabolism, and excretion studies: Conventional or microtracer? Drug Metab Dispos 2025; 53:100067. [PMID: 40198958 DOI: 10.1016/j.dmd.2025.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
A human absorption, distribution, metabolism, and excretion (hADME) study is an essential clinical pharmacology study for small-molecule drugs. The study provides insights into circulating drug-related materials and the drug's elimination pathways in humans, which can guide future studies on safety and drug-drug interaction of metabolites as well as organ impairment and drug-drug interaction of the parent drug. The 2 hADME study types, namely conventional and microtracer, are comprehensively compared in this manuscript. A review of literature found that conventional hADME studies were approximately 7 times that of microtracer hADME studies for small molecule and peptide drugs based on publications in 3 peer-reviewed journals from 2010 to 2024. Each study type has advantages and disadvantages. The advantages of conventional hADME studies primarily include the ease, low cost, and flexibility of radiometric sample analysis. In contrast, the advantages of microtracer hADME studies primarily include exemption from prerequisite studies and use of non-good manufacturing practice 14C-labeled materials. The disadvantages of each study type are essentially the advantages of the other. The manuscript also discusses scenarios where a microtracer hADME study may be preferable. Finally, recommendations are provided on selecting the appropriate hADME study type for an investigational drug. SIGNIFICANCE STATEMENT: The manuscript discusses 2 primary human absorption, distribution, metabolism, and excretion study types: conventional and microtracer. It covers published literature studies, the pros and cons of each type, scenarios for conducting microtracer studies, and a recommended decision tree for selecting the appropriate human absorption, distribution, metabolism, and excretion study type.
Collapse
Affiliation(s)
- Sean Xiaochun Zhu
- Drug Metabolism and Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, Massachusetts.
| |
Collapse
|
2
|
Tang LWT, Shi Y, Sharma R, Obach RS. The Drug-Drug Interaction between Erlotinib and OSI-930 Is Mediated through Aldehyde Oxidase Inhibition. Drug Metab Dispos 2024; 52:1020-1028. [PMID: 38889967 DOI: 10.1124/dmd.124.001802] [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: 05/15/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
The propensity for aldehyde oxidase (AO) substrates to be implicated in drug-drug interactions (DDIs) is not well understood due to the dearth of potent inhibitors that elicit in vivo inhibition of AO. Although there is only one reported instance of DDI that has been ascribed to the inhibition of AO to date, the supporting evidence for this clinical interaction is rather tenuous, and its veracity has been called into question. Our group recently reported that the epidermal growth factor receptor inhibitor erlotinib engendered potent time-dependent inhibition of AO with inactivation kinetic constants in the same order of magnitude as its free circulating plasma concentrations. At the same time, it was previously reported that the concomitant administration of erlotinib with the investigational drug OSI-930 culminated in a an approximately twofold increase in its systemic exposure. Although the basis underpinning this interaction remains unclear, the structure of OSI-930 contains a quinoline motif that is amenable to oxidation at the electrophilic carbon adjacent to the nitrogen atom by molybdenum-containing hydroxylases like AO. In this study, we conducted metabolite identification that revealed that OSI-930 undergoes AO metabolism to a mono-oxygenated 2-oxo metabolite and assessed its formation kinetics in human liver cytosol. Additionally, reaction phenotyping in human hepatocytes revealed that AO contributes nearly 50% to the overall metabolism of OSI-930. Finally, modeling the interaction between erlotinib and OSI-930 using a mechanistic static model projected an ∼1.85-fold increase in the systemic exposure of OSI-930, which accurately recapitulated clinical observations. SIGNIFICANCE STATEMENT: This study delineates an aldehyde oxidase (AO) metabolic pathway in the investigational drug OSI-930 for the first time and confirmed that it represented a major route of metabolism through reaction phenotyping in human hepatocytes. Our study provided compelling mechanistic and modeling evidence for the first instance of an AO-mediated clinical drug-drug interaction stemming from the in vivo inhibition of the AO-mediated quinoline 2-oxidation pathway in OSI-930 by erlotinib.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Yuanyuan Shi
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Raman Sharma
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
3
|
Subash S, Singh DK, Ahire D, Khojasteh SC, Murray BP, Zientek MA, Jones RS, Kulkarni P, Zubair F, Smith BJ, Heyward S, Leeder JS, Prasad B. Ontogeny of Human Liver Aldehyde Oxidase: Developmental Changes and Implications for Drug Metabolism. Mol Pharm 2024; 21:2740-2750. [PMID: 38717252 DOI: 10.1021/acs.molpharmaceut.3c01147] [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: 06/04/2024]
Abstract
Despite the increasing importance of aldehyde oxidase (AO) in the drug metabolism of clinical candidates, ontogeny data for AO are limited. The objective of our study was to characterize the age-dependent AO content and activity in the human liver cytosolic fraction (HLC) and human hepatocytes (HH). HLC (n = 121 donors) and HH (n = 50 donors) were analyzed for (1) AO protein content by quantitative proteomics and (2) enzyme activity using carbazeran as a probe substrate. AO activity showed high technical variability and poor correlation with the content in HLC samples, whereas hepatocyte samples showed a strong correlation between the content and activity. Similarly, AO content and activity showed no significant age-dependent differences in HLC samples, whereas the average AO content and activity in hepatocytes increased significantly (∼20-40-fold) from the neonatal levels (0-28 days). Based on the hepatocyte data, the age at which 50% of the adult AO content is reached (age50) was 3.15 years (0.32-13.97 years, 95% CI). Metabolite profiling of carbazeran revealed age-dependent metabolic switching and the role of non-AO mechanisms (glucuronidation and desmethylation) in carbazeran elimination. The content-activity correlation in hepatocytes improved significantly (R2 = 0.95; p < 0.0001) in samples showing <10% contribution of glucuronidation toward the overall metabolism, confirming that AO-mediated oxidation and glucuronidation are the key routes of carbazeran metabolism. Considering the confounding effect of glucuronidation on AO activity, AO content-based ontogeny data are a more direct reflection of developmental changes in protein expression. The comprehensive ontogeny data of AO in HH samples are more reliable than HLC data, which are important for developing robust physiologically based pharmacokinetic models for predicting AO-mediated metabolism in children.
Collapse
Affiliation(s)
- Sandhya Subash
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - Dilip K Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - Deepak Ahire
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - S Cyrus Khojasteh
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Bernard P Murray
- Drug Metabolism, Gilead Sciences, Inc., Foster City, California 94404, United States
| | - Michael A Zientek
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Robert S Jones
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Priyanka Kulkarni
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Faizan Zubair
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Bill J Smith
- Terminal Phase Consulting LLC, Colorado Springs, Colorado 94404, United States
| | - Scott Heyward
- BioIVT, Inc., Baltimore, Maryland 21227, United States
| | - J Steven Leeder
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri 64108, United States
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| |
Collapse
|
4
|
Tang LWT, DaSilva E, Lapham K, Obach RS. Evaluation of Icotinib as a Potent and Selective Inhibitor of Aldehyde Oxidase for Reaction Phenotyping in Human Hepatocytes. Drug Metab Dispos 2024; 52:565-573. [PMID: 38565303 DOI: 10.1124/dmd.124.001693] [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: 02/12/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Aldehyde oxidase (AO) is a molybdenum cofactor-containing cytosolic enzyme that has gained prominence due to its involvement in the developmental failure of several drug candidates in first-in-human trials. Unlike cytochrome P450s (P450) and glucuronosyltransferase, AO substrates have been plagued by poor in vitro to in vivo extrapolation, leading to low systemic exposures and underprediction of human dose. However, apart from measuring a drug's AO clearance rates, it is also important to determine the relative contribution to metabolism by this enzyme (fm,AO). Although hydralazine is the most well-studied time-dependent inhibitor (TDI) of AO and is frequently employed for AO reaction phenotyping in human hepatocytes to derive fm,AO, multiple studies have expressed concerns pertaining to its utility in providing accurate estimates of fm,AO values due to its propensity to significantly inhibit P450s at the concentrations typically used for reaction phenotyping. In this study, we characterized icotinib, a cyclized analog of erlotinib, as a potent TDI of AO-inactivating human liver cytosolic zoniporide 2-oxidation equipotently with erlotinib with a maximal inactivate rate/inactivator concentration at half maximal inactivation rate (K I) ratio of 463 and 501 minute-1mM-1 , respectively. Moreover, icotinib also exhibits selectivity against P450 and elicits significantly weaker inhibition against human liver microsomal UGT1A1/3 as compared with erlotinib. Finally, we evaluated icotinib as an inhibitor of AO for reaction phenotyping in cryopreserved human hepatocytes and demonstrated that it can yield more accurate prediction of fm,AO compared with hydralazine and induce sustained suppression of AO activity at higher cell densities, which will be important for reaction phenotyping endeavors of low clearance drugs SIGNIFICANCE STATEMENT: In this study, we characterized icotinib as a potent time-dependent inhibitor of AO with ample selectivity margins against the P450s and UGT1A1/3 and demonstrated its utility for reaction phenotyping in human hepatocytes to obtain accurate estimates of fm,AO for victim DDI risk predictions. We envisage the adoption of icotinib in place of hydralazine in AO reaction phenotyping.
Collapse
Affiliation(s)
- Lloyd Wei Tat Tang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Ethan DaSilva
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Kimberly Lapham
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
5
|
Shanu-Wilson J, Coe S, Evans L, Steele J, Wrigley S. Small molecule drug metabolite synthesis and identification: why, when and how? Drug Discov Today 2024; 29:103943. [PMID: 38452922 DOI: 10.1016/j.drudis.2024.103943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/19/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
The drug discovery and development process encompasses the interrogation of metabolites arising from the biotransformation of drugs. Here we look at why, when and how metabolites of small-molecule drugs are synthesised from the perspective of a specialist contract research organisation, with particular attention paid to projects for which regulatory oversight is relevant during this journey. To illustrate important aspects, we look at recent case studies, trends and learnings from our experience of making and identifying metabolites over the past ten years, along with with selected examples from the literature.
Collapse
Affiliation(s)
- Julia Shanu-Wilson
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK.
| | - Samuel Coe
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Liam Evans
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Jonathan Steele
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| | - Stephen Wrigley
- Hypha Discovery Ltd., 154B Brook Drive, Milton Park, Oxfordshire OX14 4SD, UK
| |
Collapse
|
6
|
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
|
7
|
Uehara S, Yasuda M, Higuchi Y, Yoneda N, Kawai K, Suzuki M, Yamazaki H, Suemizu H. SGX523 causes renal toxicity through aldehyde oxidase-mediated less-soluble metabolite formation in chimeric mice with humanized livers. Toxicol Lett 2023; 388:48-55. [PMID: 37806366 DOI: 10.1016/j.toxlet.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
SGX523 is a c-Met tyrosine kinase inhibitor that failed in clinical trials because of renal toxicity caused by crystal deposits in renal tubules. SGX523 is metabolized by aldehyde oxidase (AOX) in a species-dependent manner to the considerably less soluble 2-quinolinone-SGX523, which is likely involved in the clinically observed obstructive nephropathy. This study investigated the metabolism and renal toxicity of SGX523 in chimeric mice with humanized livers (humanized-liver mice). The 2-quinolinone-SGX523 formation activity was higher in humanized-liver mouse and human hepatocytes than in mouse hepatocytes. Additionally, this activity in the liver cytosolic fraction from humanized-liver mice was inhibited by the AOX inhibitors raloxifene and hydralazine. After oral SGX523 administration, higher maximum concentrations, larger areas under the plasma concentration versus time curves, and higher urinary concentrations of 2-quinolinone-SGX523 were observed in humanized-liver mice than in non-humanized mice. Serum creatinine and blood urea nitrogen levels were elevated in humanized-liver mice following repeated oral SGX523 administration. The accumulation of amorphous material in the tubules and infiltration of inflammatory cells around tubules were observed in the kidneys of humanized-liver mice after repeated oral SGX523 administration. These findings demonstrate that humanized-liver mice are useful for understanding the metabolism and toxicity of SGX523.
Collapse
Affiliation(s)
- Shotaro Uehara
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan.
| | - Masahiko Yasuda
- Pathology Center, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Yuichiro Higuchi
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Nao Yoneda
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Kenji Kawai
- Pathology Center, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Masami Suzuki
- Translational Research Division, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Hiroshi Yamazaki
- Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Hiroshi Suemizu
- Liver Engineering Laboratory, Department of Applied Research for Laboratory Animals, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| |
Collapse
|
8
|
Subash S, Singh DK, Ahire DS, Khojasteh SC, Murray BP, Zientek MA, Jones RS, Kulkarni P, Smith BJ, Heyward S, Cronin CN, Prasad B. Dissecting Parameters Contributing to the Underprediction of Aldehyde Oxidase-Mediated Metabolic Clearance of Drugs. Drug Metab Dispos 2023; 51:1362-1371. [PMID: 37429730 DOI: 10.1124/dmd.123.001379] [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: 04/27/2023] [Revised: 06/01/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023] Open
Abstract
We investigated the effect of variability and instability in aldehyde oxidase (AO) content and activity on the scaling of in vitro metabolism data. AO content and activity in human liver cytosol (HLC) and five recombinant human AO preparations (rAO) were determined using targeted proteomics and carbazeran oxidation assay, respectively. AO content was highly variable as indicated by the relative expression factor (REF; i.e., HLC to rAO content) ranging from 0.001 to 1.7 across different in vitro systems. The activity of AO in HLC degrades at a 10-fold higher rate in the presence of the substrate as compared with the activity performed after preincubation without substrate. To scale the metabolic activity from rAO to HLC, a protein-normalized activity factor (pnAF) was proposed wherein the activity was corrected by AO content, which revealed up to sixfold higher AO activity in HLC versus rAO systems. A similar value of pnAF was observed for another substrate, ripasudil. Physiologically based pharmacokinetic (PBPK) modeling revealed a significant additional clearance (CL; 66%), which allowed for the successful prediction of in vivo CL of four other substrates, i.e., O-benzyl guanine, BIBX1382, zaleplon, and zoniporide. For carbazeran, the metabolite identification study showed that the direct glucuronidation may be contributing to around 12% elimination. Taken together, this study identified differential protein content, instability of in vitro activity, role of additional AO clearance, and unaccounted metabolic pathways as plausible reasons for the underprediction of AO-mediated drug metabolism. Consideration of these factors and integration of REF and pnAF in PBPK models will allow better prediction of AO metabolism. SIGNIFICANCE STATEMENT: This study elucidated the plausible reasons for the underprediction of aldehyde oxidase (AO)-mediated drug metabolism and provided recommendations to address them. It demonstrated that integrating protein content and activity differences and accounting for the loss of AO activity, as well as consideration of extrahepatic clearance and additional pathways, would improve the in vitro to in vivo extrapolation of AO-mediated drug metabolism using physiologically based pharmacokinetic modeling.
Collapse
Affiliation(s)
- Sandhya Subash
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Dilip K Singh
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Deepak S Ahire
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - S Cyrus Khojasteh
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bernard P Murray
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Michael A Zientek
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Robert S Jones
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Priyanka Kulkarni
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bill J Smith
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Ciarán N Cronin
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| |
Collapse
|
9
|
Inoue K, Kikuchi K, Takahashi K, Hitaoka S, Kusano K, Komori T. In Vitro Metabolism of 2-Methoxy-N-[3-[4-[3-methyl-4-[(6-methyl- 3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) by Aldehyde Oxidase and Predicting Its Percent Contribution Relative to CYP-Mediated Metabolism. Drug Metab Dispos 2023; 51:962-969. [PMID: 37188528 DOI: 10.1124/dmd.122.000995] [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: 06/15/2022] [Revised: 02/19/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) is an anticancer drug that was discontinued due to hepatotoxicity found in clinical studies. Metabolite analysis of CP-724,714 was conducted using human hepatocytes, in which twelve oxidative metabolites and one hydrolyzed metabolite were formed. Among the three mono-oxidative metabolites, the formation of two was inhibited by adding 1-aminobenzotriazole, a pan-CYP inhibitor. In contrast, the remaining one was not affected by this inhibitor but partially inhibited by hydralazine, indicating that aldehyde oxidase (AO) was involved in metabolizing CP-724,714, which contains a quinazoline substructure, a heterocyclic aromatic quinazoline ring, known to be preferably metabolized by AO. One of the oxidative metabolites of CP-724,714 observed in human hepatocytes was also generated in recombinant human AO. Although CP-724,714 is metabolized by both CYPs and AO in human hepatocytes, the contribution level of AO could not be evaluated using its specific inhibitors because of low AO activity in in vitro human materials. Here, we present a metabolic pathway for CP-724,714 in human hepatocytes and the involvement of AO in CP-724,714 metabolism. We showed here a plausible workflow for predicting AO contribution to the metabolism of CP-724,714 based on DMPK screening data. SIGNIFICANCE STATEMENT: 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) was identified as a substrate of aldehyde oxidase (AO) rather than xanthine oxidase. Since CP-724,714 is also metabolized by cytochrome P450s (CYPs), the contribution levels of AO and CYPs in the metabolism of CP-724,714 were estimated simultaneously based on in vitro drug metabolism screening data.
Collapse
Affiliation(s)
- Kazuko Inoue
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kiyomi Kikuchi
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kazumi Takahashi
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Seiji Hitaoka
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kazutomi Kusano
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Takafumi Komori
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| |
Collapse
|
10
|
Cerny MA, Spracklin DK, Obach RS. Human Absorption, Distribution, Metabolism, and Excretion Studies: Origins, Innovations, and Importance. Drug Metab Dispos 2023; 51:647-656. [PMID: 36973000 DOI: 10.1124/dmd.122.001006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 03/13/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Human absorption, distribution, metabolism, and excretion (hADME) studies represent one of the most important clinical studies in terms of obtaining a comprehensive and quantitative overview of the total disposition of a drug. This article will provide background on the origins of hADME studies as well as provide an overview of technological innovations that have impacted how hADME studies are carried out and analyzed. An overview of the current state of the art for hADME studies will be provided, the impacts of advances in technology and instrumentation on the timing of and approaches to hADME studies will be discussed, and a summary of the parameters and information obtained from these studies will be offered. Additionally, aspects of the ongoing debate over the importance of animal absorption, distribution, metabolism, and excretion studies versus a "human-first, human-only strategy" will be presented. Along with the information above, this manuscript will highlight how, for over 50 years, Drug Metabolism and Disposition has served as an important outlet for the reporting of hADME studies. SIGNIFICANCE STATEMENT: Human absorption, distribution, metabolism, and excretion (hADME) studies have and will continue to be important to the understanding and development of drugs. This manuscript provides a historical perspective on the origins of hADME studies as well as advancements resulting in the current-state-of the art practice for these studies.
Collapse
Affiliation(s)
- Matthew A Cerny
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut
| | - Douglas K Spracklin
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Groton, Connecticut
| |
Collapse
|
11
|
Young GC, Spracklin DK, James AD, Hvenegaard MG, Scarfe G, Wagner DS, Georgi K, Schieferstein H, Bjornsdottir I, van Groen B, Romeo AA, Cassidy KC, Da-Violante G, Bister B, Blech S, Lyer R, Schulz SI, Cuyckens F, Moliner P. Considerations for Human ADME Strategy and Design Paradigm Shift(s) - An Industry White Paper. Clin Pharmacol Ther 2023; 113:775-781. [PMID: 35733280 DOI: 10.1002/cpt.2691] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022]
Abstract
The human absorption, distribution, metabolism, and excretion (hADME) study is the cornerstone of the clinical pharmacology package for small molecule drugs, providing comprehensive information on the rates and routes of disposition and elimination of drug-related material in humans through the use of 14 C-labeled drug. Significant changes have already been made in the design of the hADME study for many companies, but opportunity exists to continue to re-think both the design and timing of the hADME study in light of the potential offered by newer technologies, that enable flexibility in particular to reducing the magnitude of the radioactive dose used. This paper provides considerations on the variety of current strategies that exist across a number of pharmaceutical companies and on some of the ongoing debates around a potential move to the so called "human first/human only" approach, already adopted by at least one company. The paper also provides a framework for continuing the discussion in the application of further shifts in the paradigm.
Collapse
Affiliation(s)
- Graeme C Young
- GlaxoSmithKline Research & Development Ltd., David Jack Centre, Ware, UK
| | | | | | | | - Graeme Scarfe
- AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Katrin Georgi
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | | | - Andrea A Romeo
- Roche Pharma Research and Early Development, Basel, Switzerland
| | | | | | - Bojan Bister
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Stefan Blech
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | | | | |
Collapse
|
12
|
Huang M, Zhu K, Wang Y, Lou C, Sun H, Li W, Tang Y, Liu G. In Silico Prediction of Metabolic Reaction Catalyzed by Human Aldehyde Oxidase. Metabolites 2023; 13:metabo13030449. [PMID: 36984889 PMCID: PMC10059660 DOI: 10.3390/metabo13030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Aldehyde oxidase (AOX) plays an important role in drug metabolism. Human AOX (hAOX) is widely distributed in the body, and there are some differences between species. Currently, animal models cannot accurately predict the metabolism of hAOX. Therefore, more and more in silico models have been constructed for the prediction of the hAOX metabolism. These models are based on molecular docking and quantum chemistry theory, which are time-consuming and difficult to automate. Therefore, in this study, we compared traditional machine learning methods, graph convolutional neural network methods, and sequence-based methods with limited data, and proposed a ligand-based model for the metabolism prediction catalyzed by hAOX. Compared with the published models, our model achieved better performance (ACC = 0.91, F1 = 0.77). What's more, we built a web server to predict the sites of metabolism (SOMs) for hAOX. In summary, this study provides a convenient and automatable model and builds a web server named Meta-hAOX for accelerating the drug design and optimization stage.
Collapse
Affiliation(s)
- Mengting Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Keyun Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yimeng Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chaofeng Lou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Huimin Sun
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
13
|
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: 0.7] [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
|
14
|
Toselli F, Golding M, Nicolaï J, Gillent E, Chanteux H. Drug clearance by aldehyde oxidase: can we avoid clinical failure? Xenobiotica 2022; 52:890-903. [PMID: 36170034 DOI: 10.1080/00498254.2022.2129519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite increased awareness of aldehyde oxidase (AO) as a major drug-metabolising enzyme, predicting the pharmacokinetics of its substrates remains challenging. Several drug candidates have been terminated due to high clearance, which were subsequently discovered to be AO substrates. Even retrospective extrapolation of human clearance, from models more sensitive to AO activity, often resulted in underprediction.The questions of the current work thus were: Is there an acceptable degree of in vitro AO metabolism that does not result in high in vivo human clearance? And, if so, how can this be predicted?We built an in vitro/in vivo correlation using known AO substrates, combining multiple in vitro parameters to calculate the blood metabolic clearance mediated by AO (CLbAO). This value was compared with observed blood clearance (CLb-obs), establishing cut-off CLbAO values, to discriminate between low and high CLb-obs. The model was validated using additional literature compounds, and CLb-obs was predicted in the correct category.This simple, categorical, semi-quantitative yet multi-factorial model is readily applicable in drug discovery. Further, it is valuable for high-clearance compounds, as it predicts the CLb group, rather than an exact CLb value, for the substrates of this poorly-characterised enzyme.
Collapse
Affiliation(s)
| | | | - Johan Nicolaï
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| | - Eric Gillent
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| | - Hugues Chanteux
- Development Science, UCB Biopharma, Braine-l'Alleud, Belgium
| |
Collapse
|
15
|
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: 4.0] [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
|
16
|
Kozminski KD, Selimkhanov J, Heyward S, Zientek MA. Contribution of Extrahepatic Aldehyde Oxidase Activity to Human Clearance. Drug Metab Dispos 2021; 49:743-749. [PMID: 34162687 DOI: 10.1124/dmd.120.000313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/10/2021] [Indexed: 11/22/2022] Open
Abstract
Aldehyde oxidase (AOX) is a soluble, cytosolic enzyme that metabolizes various N-heterocyclic compounds and organic aldehydes. It has wide tissue distribution with highest levels found in liver, kidney, and lung. Human clearance projections of AOX substrates by in vitro assessments in isolated liver fractions (cytosol, S9) and even hepatocytes have been largely underpredictive of clinical outcomes. Various hypotheses have been suggested as to why this is the case. One explanation is that extrahepatic AOX expression contributes measurably to AOX clearance and is at least partially responsible for the often observed underpredictions. Although AOX expression has been confirmed in several extrahepatic tissues, activities therein and potential contribution to overall human clearance have not been thoroughly studied. In this work, the AOX enzyme activity using the S9 fractions of select extrahepatic human tissues (kidney, lung, vasculature, and intestine) were measured using carbazeran as a probe substrate. Measured activities were scaled to a whole-body clearance using best-available parameters and compared with liver S9 fractions. Here, the combined scaled AOX clearance obtained from the kidney, lung, vasculature, and intestine is very low and amounted to <1% of liver. This work suggests that AOX metabolism from extrahepatic sources plays little role in the underprediction of activity in human. One of the notable outcomes of this work has been the first direct demonstration of AOX activity in human vasculature. SIGNIFICANCE STATEMENT: This work demonstrates aldehyde oxidase (AOX) activity is measurable in a variety of extrahepatic human tissues, including vasculature, yet activities and potential contributions to human clearance are relatively low and insignificant when compared with the liver. Additionally, the modeling of the tissue-specific in vitro kinetic data suggests that AOX may be influenced by the tissue it resides in and thus show different affinity, activity, and modified activity over time.
Collapse
Affiliation(s)
- Kirk D Kozminski
- Takeda Pharmaceuticals Limited, San Diego, California (K.D.K., J.S., M.A.Z.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Jangir Selimkhanov
- Takeda Pharmaceuticals Limited, San Diego, California (K.D.K., J.S., M.A.Z.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Scott Heyward
- Takeda Pharmaceuticals Limited, San Diego, California (K.D.K., J.S., M.A.Z.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Michael A Zientek
- Takeda Pharmaceuticals Limited, San Diego, California (K.D.K., J.S., M.A.Z.); and BioIVT, Baltimore, Maryland (S.H.)
| |
Collapse
|
17
|
Dhuria NV, Haro B, Kapadia A, Lobo KA, Matusow B, Schleiff MA, Tantoy C, Sodhi JK. Recent developments in predicting CYP-independent metabolism. Drug Metab Rev 2021; 53:188-206. [PMID: 33941024 DOI: 10.1080/03602532.2021.1923728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
As lead optimization efforts have successfully reduced metabolic liabilities due to cytochrome P450 (CYP)-mediated metabolism, there has been an increase in the frequency of involvement of non-CYP enzymes in the metabolism of investigational compounds. Although there have been numerous notable advancements in the characterization of non-CYP enzymes with respect to their localization, reaction mechanisms, species differences and identification of typical substrates, accurate prediction of non-CYP-mediated clearance, with a particular emphasis with the difficulties in accounting for any extrahepatic contributions, remains a challenge. The current manuscript comprehensively summarizes the recent advancements in the prediction of drug metabolism and the in vitro to in vitro extrapolation of clearance for substrates of non-CYP drug metabolizing enzymes.
Collapse
Affiliation(s)
- Nikhilesh V Dhuria
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bianka Haro
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Amit Kapadia
- California Poison Control Center, University of California San Francisco, San Diego, CA, USA
| | | | - Bernice Matusow
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA
| | - Mary A Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Christina Tantoy
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA
| | - Jasleen K Sodhi
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA.,Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
18
|
De Sousa Mendes M, L Orton A, Humphries HE, Jones B, Gardner I, Neuhoff S, Pilla Reddy V. A Laboratory-Specific Scaling Factor to Predict the In Vivo Human Clearance of Aldehyde Oxidase Substrates. Drug Metab Dispos 2020; 48:1231-1238. [PMID: 32893186 DOI: 10.1124/dmd.120.000082] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/22/2020] [Indexed: 02/13/2025] Open
Abstract
Aldehyde oxidase (AO) efficiently metabolizes a range of compounds with N-containing heterocyclic aromatic rings and/or aldehydes. The limited knowledge of AO activity and abundance (in vitro and in vivo) has led to poor prediction of in vivo systemic clearance (CL) using in vitro-to-in vivo extrapolation approaches, which for drugs in development can lead to their discontinuation. We aimed to identify appropriate scaling factors to predict AO CL of future new chemical entities (NCEs). The metabolism of six AO substrates was measured in human liver cytosol (HLC) and S9 fractions. Measured blood-to-plasma ratios and free fractions (in the in vitro system and in plasma) were used to develop physiologically based pharmacokinetic models for each compound. The impact of extrahepatic metabolism was explored, and the intrinsic clearance required to recover in vivo profiles was estimated and compared with in vitro measurements. Using HLC data and assuming only hepatic metabolism, a systematic underprediction of clearance was observed (average fold underprediction was 3.8). Adding extrahepatic metabolism improved the accuracy of the results (average fold error of 1.9). A workflow for predicting metabolism of an NCE by AO is proposed, and an empirical (laboratory-specific) scaling factor of three on the predicted intravenous CL allows a reasonable prediction of the available clinical data. Alternatively, considering also extrahepatic metabolism, an scaling factor of 6.5 applied on the intrinsic clearance could be used. Future research should focus on the impact of the in vitro study designs and the contribution of extrahepatic metabolism to AO-mediated clearance to understand the mechanisms behind the systematic underprediction. SIGNIFICANCE STATEMENT: This works describes the development of scaling factors to allow in vitro-in vivo extrapolation of the clearance of compounds by aldehyde oxidase metabolism in humans. In addition, physiologically based pharmacokinetic models were developed for each of the aldehyde oxidase substrate compounds investigated.
Collapse
Affiliation(s)
- Mailys De Sousa Mendes
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alexandra L Orton
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Helen E Humphries
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Barry Jones
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Iain Gardner
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sibylle Neuhoff
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Venkatesh Pilla Reddy
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom (M.D.S.M., H.E.H., I.G., S.N.) and Oncology DMPK Research & Early Development (A.O., B.J.) and Modelling and Simulation, Research & Early Development (V.P.R.), Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
19
|
Irie K, Nakagawa A, Fujita H, Tamura R, Eto M, Ikesue H, Muroi N, Tomii K, Hashida T. Pharmacokinetics of Favipiravir in Critically Ill Patients With COVID-19. Clin Transl Sci 2020; 13:880-885. [PMID: 32475019 PMCID: PMC7300626 DOI: 10.1111/cts.12827] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022] Open
Abstract
Since December 2019, a novel coronavirus (severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2)) infection has been rapidly spreading worldwide and causing the respiratory illness, coronavirus disease 2019 (COVID‐19). The antiretroviral drug favipiravir (FPV) has been experimentally used for COVID‐19 treatment since March 2020 in Japan. However, the pharmacokinetics of FPV in critically ill patients is unknown. We measured the serum concentration of FPV using high‐performance liquid chromatography in patients with severe COVID‐19 who were admitted to the intensive care unit and placed on mechanical ventilation. The patients were administered 1,600 mg of FPV twice daily on day 1, followed by 600 mg twice daily from day 2 to day 5 (or more if needed). Suspensions of FPV tablets were administered through a nasogastric tube. Seven patients were enrolled in this study. Forty‐nine blood samples were obtained from the eligible patients to evaluate FPV concentration. The FPV trough (after 8–12 hours) concentrations of most samples were lower than the lower limit of quantification (1 µg/mL) and half‐maximal effective concentration (9.7 µg/mL) against SARS‐CoV‐2 previously tested in vitro. FPV trough concentration in critically ill patients was much lower than that of healthy subjects in a previous clinical trial, which is a cause for great concern. Further study is required to determine the optimal strategy for treatment of patients with severe COVID‐19.
Collapse
Affiliation(s)
- Kei Irie
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan.,Department of Pharmaceutics, Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| | - Atsushi Nakagawa
- Department of Respiratory Medicine, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hirotoshi Fujita
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Ryo Tamura
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Masaaki Eto
- Department of Clinical Laboratory, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hiroaki Ikesue
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Nobuyuki Muroi
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Tohru Hashida
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| |
Collapse
|
20
|
Williamson B, Colclough N, Fretland AJ, Jones BC, Jones RDO, McGinnity DF. Further Considerations Towards an Effective and Efficient Oncology Drug Discovery DMPK Strategy. Curr Drug Metab 2020; 21:145-162. [PMID: 32164508 DOI: 10.2174/1389200221666200312104837] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/06/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND DMPK data and knowledge are critical in maximising the probability of developing successful drugs via the application of in silico, in vitro and in vivo approaches in drug discovery. METHODS The evaluation, optimisation and prediction of human pharmacokinetics is now a mainstay within drug discovery. These elements are at the heart of the 'right tissue' component of AstraZeneca's '5Rs framework' which, since its adoption, has resulted in increased success of Phase III clinical trials. With the plethora of DMPK related assays and models available, there is a need to continually refine and improve the effectiveness and efficiency of approaches best to facilitate the progression of quality compounds for human clinical testing. RESULTS This article builds on previously published strategies from our laboratories, highlighting recent discoveries and successes, that brings our AstraZeneca Oncology DMPK strategy up to date. We review the core aspects of DMPK in Oncology drug discovery and highlight data recently generated in our laboratories that have influenced our screening cascade and experimental design. We present data and our experiences of employing cassette animal PK, as well as re-evaluating in vitro assay design for metabolic stability assessments and expanding our use of freshly excised animal and human tissue to best inform first time in human dosing and dose escalation studies. CONCLUSION Application of our updated drug-drug interaction and central nervous system drug exposure strategies are exemplified, as is the impact of physiologically based pharmacokinetic and pharmacokinetic-pharmacodynamic modelling for human predictions.
Collapse
Affiliation(s)
- Beth Williamson
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Nicola Colclough
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Adrian John Fretland
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Boston MA, United States
| | - Barry Christopher Jones
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Rhys Dafydd Owen Jones
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dermot Francis McGinnity
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
21
|
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: 67] [Impact Index Per Article: 11.2] [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
|
22
|
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: 50] [Impact Index Per Article: 8.3] [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
|
23
|
Toselli F, Fredenwall M, Svensson P, Li XQ, Johansson A, Weidolf L, Hayes MA. Hip To Be Square: Oxetanes as Design Elements To Alter Metabolic Pathways. J Med Chem 2019; 62:7383-7399. [DOI: 10.1021/acs.jmedchem.9b00030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Francesca Toselli
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Marlene Fredenwall
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Peder Svensson
- Integrative Research Laboratories, Arvid Wallgrens Backe 20, Gothenburg 413 46, Sweden
| | - Xue-Qing Li
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Johansson
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Weidolf
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Martin A. Hayes
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
24
|
Affiliation(s)
- Christine Beedham
- Honorary Senior Lecturer, Faculty of Life Sciences, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
25
|
Hausner EA, Elmore SA, Yang X. Overview of the Components of Cardiac Metabolism. Drug Metab Dispos 2019; 47:673-688. [PMID: 30967471 PMCID: PMC7333657 DOI: 10.1124/dmd.119.086611] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Metabolism in organs other than the liver and kidneys may play a significant role in how a specific organ responds to chemicals. The heart has metabolic capability for energy production and homeostasis. This homeostatic machinery can also process xenobiotics. Cardiac metabolism includes the expression of numerous organic anion transporters, organic cation transporters, organic carnitine (zwitterion) transporters, and ATP-binding cassette transporters. Expression and distribution of the transporters within the heart may vary, depending on the patient's age, disease, endocrine status, and various other factors. Several cytochrome P450 (P450) enzyme classes have been identified within the heart. The P450 hydroxylases and epoxygenases within the heart produce hydroxyeicosatetraneoic acids and epoxyeicosatrienoic acids, metabolites of arachidonic acid, which are critical in regulating homeostatic processes of the heart. The susceptibility of the cardiac P450 system to induction and inhibition from exogenous materials is an area of expanding knowledge, as are the metabolic processes of glucuronidation and sulfation in the heart. The susceptibility of various transcription factors and signaling pathways of the heart to disruption by xenobiotics is not fully characterized but is an area with implications for disruption of normal postnatal development, as well as modulation of adult cardiac health. There are knowledge gaps in the timelines of physiologic maturation and deterioration of cardiac metabolism. Cross-species characterization of cardiac-specific metabolism is needed for nonclinical work of optimum translational value to predict possible adverse effects, identify sensitive developmental windows for the design and conduct of informative nonclinical and clinical studies, and explore the possibilities of organ-specific therapeutics.
Collapse
Affiliation(s)
- Elizabeth A Hausner
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Susan A Elmore
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| | - Xi Yang
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland (E.A.H., X.Y.); and National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina (S.A.E.)
| |
Collapse
|
26
|
Padilha EC, Wang J, Kerns E, Lee A, Huang W, Jiang JK, McKew J, Mutlib A, Peccinini RG, Yu PB, Sanderson P, Xu X. Application of in vitro Drug Metabolism Studies in Chemical Structure Optimization for the Treatment of Fibrodysplasia Ossificans Progressiva (FOP). Front Pharmacol 2019; 10:234. [PMID: 31068801 PMCID: PMC6491728 DOI: 10.3389/fphar.2019.00234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/22/2019] [Indexed: 12/14/2022] Open
Abstract
Currently no approved treatment exists for fibrodysplasia ossificans progressiva (FOP) patients, and disease progression results in severe restriction of joint function and premature mortality. LDN-193189 has been demonstrated to be efficacious in a mouse FOP disease model after oral administration. To support species selection for drug safety evaluation and to guide structure optimization for back-up compounds, in vitro metabolism of LDN-193189 was investigated in liver microsome and cytosol fractions of mouse, rat, dog, rabbit, monkey and human. Metabolism studies included analysis of reactive intermediate formation using glutathione and potassium cyanide (KCN) and analysis of non-P450 mediated metabolites in cytosol fractions of various species. Metabolite profiles and metabolic soft spots of LDN-193189 were elucidated using LC/UV and mass spectral techniques. The in vitro metabolism of LDN-193189 was significantly dependent on aldehyde oxidase, with formation of the major NIH-Q55 metabolite. The piperazinyl moiety of LDN-193189 was liable to NADPH-dependent metabolism which generated reactive iminium intermediates, as confirmed through KCN trapping experiments, and aniline metabolites (M337 and M380), which brought up potential drug safety concerns. Subsequently, strategies were employed to avoid metabolic liabilities leading to the synthesis of Compounds 1, 2, and 3. This study demonstrated the importance of metabolite identification for the discovery of novel and safe drug candidates for the treatment of FOP and helped medicinal chemists steer away from potential metabolic liabilities.
Collapse
Affiliation(s)
- Elias C Padilha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States.,Department of Natural Active Principles and Toxicology, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Jianyao Wang
- Department of Pharmacokinetics, Dynamics and Metabolism, Discovery Sciences, Janssen Research and Development, Spring House, PA, United States.,Frontage Laboratories, Inc., Department of Drug Metabolism, Exton, PA, United States
| | - Ed Kerns
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Arthur Lee
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Wenwei Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Jian-Kang Jiang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - John McKew
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Abdul Mutlib
- Frontage Laboratories, Inc., Department of Drug Metabolism, Exton, PA, United States
| | - Rosangela G Peccinini
- Department of Natural Active Principles and Toxicology, School of Pharmaceutical Sciences, Universidade Estadual Paulista (UNESP), Araraquara, Brazil
| | - Paul B Yu
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Philip Sanderson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
27
|
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: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
|
28
|
Li AC, Cui D, Yu E, Dobson K, Hellriegel ET, Robertson Jr P. Identification and human exposure prediction of two aldehyde oxidase-mediated metabolites of a methylquinoline-containing drug candidate. Xenobiotica 2018; 49:302-312. [PMID: 29473769 DOI: 10.1080/00498254.2018.1444815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Austin C. Li
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| | - Donghui Cui
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| | - Erya Yu
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| | - Kyle Dobson
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| | - Edward T. Hellriegel
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| | - Philmore Robertson Jr
- Department of Drug Metabolism and Pharmacokinetics, Teva Pharmaceuticals, West Chester, PA, USA
| |
Collapse
|
29
|
Crouch RD, Blobaum AL, Felts AS, Conn PJ, Lindsley CW. Species-Specific Involvement of Aldehyde Oxidase and Xanthine Oxidase in the Metabolism of the Pyrimidine-Containing mGlu 5-Negative Allosteric Modulator VU0424238 (Auglurant). Drug Metab Dispos 2017; 45:1245-1259. [PMID: 28939686 DOI: 10.1124/dmd.117.077552] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/20/2017] [Indexed: 02/13/2025] Open
Abstract
Aldehyde oxidase (AO) and xanthine oxidase (XO) are molybdo-flavoenzymes that catalyze oxidation of aromatic azaheterocycles. Differences in AO activity have been reported among various species, including rats, humans, and monkeys. Herein we report a species difference in the enzymes responsible for the metabolism of the negative allosteric modulator of metabotropic glutamate receptor subtype 5 (mGlu5 NAM) VU0424238 (VU238, auglurant). Hepatic S9 incubations with AO and XO specific inhibitors hydralazine and allopurinol indicated that rats and cynomolgus monkeys both oxidized VU238 to the 6-oxopyrimidine metabolite M1 via an AO-mediated pathway, whereas secondary oxidation to the 2,6-dioxopyrimidine metabolite M2 was mediated predominantly by AO in monkeys and XO in rats. Despite differences in enzymatic pathways, intrinsic clearance (CLint) of M1 was similar between species (cynomolgus and rat CLint = 2.00 ± 0.040 and 2.19 ± 0.201 μl/min per milligram of protein, respectively). Inhibitor studies in the S9 of multiple species indicated that oxidation of VU238 to M1 was mediated predominantly by AO in humans, cynomolgus and rhesus monkeys, rats, mice, guinea pigs, and minipigs. Oxidation of M1 to M2 was mediated predominantly by XO in rats and mice and by AO in monkeys and guinea pigs, whereas low turnover prevented enzyme phenotyping in humans and minipigs. Additionally, inhibitor experiments indicated that oxidation at the 2-position of the pyrimidine ring of the known AO substrate, BIBX1382, was mediated by AO in all species, although production of this metabolite was comparatively low in rats and mice. These data may suggest low reactivity of rat AO toward 2-oxidation of pyrimidine-containing compounds and highlight the importance of thoroughly characterizing AO-metabolized drug candidates in multiple preclinical species.
Collapse
Affiliation(s)
- Rachel D Crouch
- Vanderbilt Center for Neuroscience Drug Discovery (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), Departments of Pharmacology (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), and Chemistry (C.W.L.), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Anna L Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), Departments of Pharmacology (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), and Chemistry (C.W.L.), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Andrew S Felts
- Vanderbilt Center for Neuroscience Drug Discovery (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), Departments of Pharmacology (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), and Chemistry (C.W.L.), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), Departments of Pharmacology (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), and Chemistry (C.W.L.), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), Departments of Pharmacology (R.D.C., A.L.B., A.S.F., P.J.C., C.W.L.), and Chemistry (C.W.L.), Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|